The American Psychiatric Publishing Textbook of Geriatric Psychiatry, 5th Edition
The American Psychiatric Publishing Textbook of Geriatric Psychiatry, 5th Edition
TEXTBOOK OF
GERIATRIC PSYCHIATRY
FIFTH EDITION
The American Psychiatric Publishing
TEXTBOOK OF
GERIATRIC PSYCHIATRY
FIFTH EDITION
EDITED BY
David C. Steffens, M.D., M.H.S.
Dan G. Blazer, M.D., Ph.D.
Mugdha E. Thakur, M.D.
Note: The authors have worked to ensure that all information in this book is
accurate at the time of publication and consistent with general psychiatric
and medical standards, and that information concerning drug dosages,
schedules, and routes of administration is accurate at the time of publication
and consistent with standards set by the U.S. Food and Drug Administration
and the general medical community. As medical research and practice
continue to advance, however, therapeutic standards may change.
Moreover, specific situations may require a specific therapeutic response
not included in this book. For these reasons and because human and
mechanical errors sometimes occur, we recommend that readers follow the
advice of physicians directly involved in their care or the care of a member
of their family.
If you would like to buy between 25 and 99 copies of this or any other
American Psychiatric Publishing title, you are eligible for a 20% discount;
please contact Customer Service at [email protected] or 800-368-5777. If
you wish to buy 100 or more copies of the same title, please e-mail us at
[email protected] for a price quote.
PART I
Introduction to Geriatric Psychiatry
PART II
Evaluation of Psychiatric Disorders in Late Life
PART III
Presentation of Psychiatric Disorders in Late Life
7 Delirium
Jane S. Saczynski, Ph.D.
Sharon K. Inouye, M.D., M.P.H.
9 Depressive Disorders
Dan G. Blazer, M.D., Ph.D.
David C. Steffens, M.D., M.H.S.
5 Bereavement
Moria J. Smoski, Ph.D.
Stephanie T. Jenal, Ph.D.
Larry W. Thompson, Ph.D.
8 Personality Disorders
Thomas E. Oxman, M.D.
0 Psychopharmacology
Benoit H. Mulsant, M.D.
Bruce G. Pollock, M.D., Ph.D.
Index
Contributors
Marc E. Agronin, M.D.
Medical Director for Mental Health and Clinical Research, Miami Jewish
Health Systems; Affiliate Associate Professor of Psychiatry and Neurology,
University of Miami Miller School of Medicine, Miami, Florida
Disclosure of Interests
The following contributors to this book have indicated a financial interest
in or other affiliation with a commercial supporter, a manufacturer of a
commercial product, a provider of a commercial service, a
nongovernmental organization, and/or a government agency, as listed
below:
Demography
In 2010, approximately 40 million persons ages 65 years and older lived in
the United States, accounting for 13% of the population. The number of
persons in this age group has steadily increased, from 3.1 million in 1900
and 12.3 million in 1950 to the current estimate. With the aging of the baby
boomer cohort (those born between 1946 and 1964), the size of the elderly
population is projected to continue to increase over the next several decades
and to reach 72.1 million by the year 2030 and 88.5 million by 2050,
accounting for an estimated 20.2% of the population. Even more
astounding, the number of oldest old, or persons ages 85 years and older,
was 5.5 million in 2010 and was projected to reach 19.0 million by 2050
(Federal Interagency Forum on Aging-Related Statistics 2012).
In 2009, about 3% of adults ages 65 years and older resided in
community housing with at least one service available, whereas 4% resided
in long-term-care facilities. Among individuals ages 85 years and older, 8%
resided in community housing with services and 14% resided in long-term
care facilities (Federal Interagency Forum on Aging-Related Statistics
2012). Many of these residents are placed in residential care because of
psychiatric disorders, especially the behavior problems that result from
Alzheimer’s disease.
The current older population of the United States is predominantly
female and white. In 2010, women accounted for 57% of the population
ages 65 years and older and 67% of those ages 85 years and older (Federal
Interagency Forum on Aging-Related Statistics 2012). The racial and ethnic
composition of the older population is projected to change over the next
several decades. In 2010, approximately 80% of individuals ages 65 years
and older were non-Hispanic white, 8% non-Hispanic black, 7% Hispanic,
3% Asian, and 2% other race/ethnic group. By 2050, the proportion of non-
Hispanic blacks is projected to be 12%, Asians 9%, and Hispanics of any
race 20%, whereas the proportion of non-Hispanic whites is expected to
decrease to 58% (Federal Interagency Forum on Aging-Related Statistics
2012).
Life expectancy in the United States in 1900 was 50.7 years for women
and 47.9 years for men, whereas the life expectancy at birth in 2009 was
80.9 years for women and 76.0 for men. In 2009, a person age 65 could
expect to live an average of 19.2 more years, and a person age 85 could
expect to live 6.7 additional years (Federal Interagency Forum on Aging-
Related Statistics 2012).
The percentage of older adults in the labor force—those who are either
working or looking for work—has changed over the last five decades. The
percentage of men ages 65–69 in the labor force declined from 40.9% in
1963 to 24.4% in 1985 and then increased to 37.4% in 2011. The
percentage of men ages 70 years and older in the labor force declined from
20.8% in 1963 to 10.5% in 1985 and then increased to 15.4% in 2011. The
percentage of women ages 65–69 years in the labor force increased from
16.5% in 1963 to 27.3% in 2011 (Federal Interagency Forum on Aging-
Related Statistics 2012). Whether or not older adults choose to remain in or
leave the workforce, the effect of an aging population on the economy of
the United States—not to mention the need for health care—will be
dramatic.
If an older person develops a psychiatric disorder, the disorder may
become chronic and the person may live many years with a decreased
quality of life because of psychiatric morbidity. In addition, the great
majority of older persons with psychiatric disorders experience a comorbid
physical illness. Currently, the proportion of younger and middle-aged
adults with a psychiatric disorder is higher than the proportion of older
adults with a psychiatric disorder, suggesting that as the younger adults age,
the proportion of older adults with a lifetime psychiatric disorder may also
increase and create a potential crisis in geriatric mental health (Jeste et al.
1999).
What can psychiatric epidemiological studies contribute to mental health
services for older adults? Morris (1975) suggested the following uses of
epidemiology:
Case Identification
Clinicians constantly face the task of distinguishing abnormality from
normality. Although most epidemiologists and clinicians agree on the core
symptoms of psychiatric disorders throughout the life cycle, the absolute
distinction between cases and noncases—that is, persons who require
psychiatric attention versus those who do not require such care—is not
easily established. Many of the symptoms and signs of a psychiatric
disorder in late life may be ubiquitous with the aging process, thus blurring
the distinction between cases and noncases. Epidemiologists can assist the
clinician in identifying meaningful clusters of symptoms and significant
degrees of symptom severity. Case identification is also the foundation of
descriptive epidemiology. “Cases” are the numerator of the equation from
which prevalence (the proportion of disease that is present in the population
during a specified time period) and incidence (the proportion of new cases
that develop in a population at risk over a specified time period such as 1
year) are derived from community and clinical samples (the denominator).
What is a case? Copeland (1981) suggested that this question be turned
by epidemiologists, with advantage, to “A case for what?” The choice of a
construct for a case depends on the particular scientific or clinical inquiry of
the investigator. If, to determine a value of a new short-acting sedative-
hypnotic agent, the clinician wishes to identify a group of older adults with
initial insomnia, the prevalence and severity of a target symptom (initial
insomnia) define the case. The sleep difficulty may result from several
different underlying disorders, but diagnosis would be irrelevant to the
purpose of the study. For most clinicians, however, the goal of case
identification is to identify subjects experiencing uniform underlying
psychopathology, as is implicit in DSM-5 (American Psychiatric
Association 2013).
Diagnostic categories that approximate true disease processes have
several characteristics, including the following (Weissman and Klerman
1978):
Historical Studies
Psychiatrists typically follow patients for relatively short periods during the
course of their illnesses. In addition, they usually interact with each patient
within a relatively brief window of historical time. Epidemiological studies
add a historical perspective to current cross-sectional findings in population
and clinical surveys. Historical studies in psychiatric epidemiology are rare.
Temporal changes that occur with most behaviors that are of psychiatric
interest must be determined over years rather than months. Longitudinal
studies introduce problems of loss to follow-up. With the caveat that case
identification methods change over time and place, rendering the detection
of historical trends subject to misclassification error, historical studies have
contributed importantly to estimating the separate effects of age, historical
events, and cohort behavior on the incidence of mental illness. For example,
the benefits of historical studies can be seen in trends of suicide mortality
among older adults.
Etiological Studies
One of the basic contributions of epidemiology is to identify determinants
of disease or to identify causal factors that can offer the possibility of
disease prevention. Within geriatric psychiatry, it is important to identify
factors that can either predispose individuals to developing psychiatric
disorders or precipitate the recurrence of such disorders. These disorders
may have their initial onset in late life, or the disorders may have an early
onset and recur later in life. Other factors can be identified that are
associated with the prevalence of a disorder, but the antecedent-consequent
relationship has not been established. For practical purposes in this
discussion, we identify all of these as risk factors. These factors generally
fall into several categories, including genetic or biological factors,
environmental or chemical factors, and social factors. Examples of each are
provided in the following subsections. In addition, the presence of a
comorbid physical or mental condition or disorder often leads to the
development of psychiatric symptoms or another disorder, and these are
described in chapters related to specific disorders throughout this textbook.
Genetic Factors
The genetic predisposition to psychiatric disorders is covered in other
chapters of this textbook (e.g., see Chapter 3, “Genomics in Geriatric
Psychiatry”) and is not reviewed here. Research that focuses on the
interaction between genetic and environmental factors and the occurrence
of disease (Hernandez and Blazer 2006), however, can potentially offer new
information on the etiology of psychiatric disorders. For example, Hendrie
et al. (2004) reported a significant association between the ε4 allele of the
apolipoprotein E gene (APOE*E4) and Alzheimer’s disease in African
Americans in Indianapolis, Indiana, but these alleles were not associated
with an increased risk for Alzheimer’s disease among Yoruba living in
Ibadan, Nigeria, suggesting that the interaction between gene and
environment may play a role in the etiology of the disease. Gatz et al.
(1992) studied genetic and environmental contributions to self-reported
depressive symptoms in older adults in a sample of twin pairs. Genetic
influence accounted for 16% of the variance in depression score and for
19% of the variance in psychomotor and somatic complaints, but
heritability was minimal for depressed mood and well-being. Although
shared experiences contributed to the variance, the most important correlate
of late-life depressive symptoms was nonshared experiences.
Social Factors
By far the most frequently investigated environmental factors associated
with psychiatric disorders are social factors. Many investigators believe that
the changing roles and circumstances of older adults can cause stress and
thereby contribute to the onset of psychiatric disorders and cognitive
difficulties. In a study of 986 community-dwelling older adults, Blazer
(1980b) found the crude estimate of relative risk for mental health
impairment to be 2.14, given a life event score of 150 or greater on the
Schedule of Recent Events (Holmes and Rahe 1967). A relative risk of 1.73
(P<0.01) was estimated when a binary regression procedure was used,
controlling for physical health, economic status, social support, and age. In
a study of individuals ages 55 years and older, Murrell et al. (1983) found
that social factors, including widowhood, divorce, separation, and
decreased income, were related to depressive symptomatology in the
community. In the Hispanic EPESE, economic stressors and conditions
such as chronic financial strain were associated with depressive symptoms
in Mexican American elders (Black et al. 1998).
In the Longitudinal Aging Study Amsterdam, major depression was
associated with unmarried status, functional limitation, perceived
loneliness, internal locus of control, poorer self-perceived health, and lack
of instrumental social support (Beekman et al. 1995). In the Duke ECA
study, the recent experience of negative life events and poor social support
were associated with major depression (Blazer et al. 1987b). Perceived
health and loneliness were some of the correlates of depressive symptoms
in the Leiden 85-plus study (Stek et al. 2004). Depression has also been
linked with variables suggesting increased dependency (Anstey et al. 2007).
Impairment or dissatisfaction with one’s social network has been reported
to be associated with anxiety symptoms in late life (Forsell and Winblad
1998). Nevertheless, the study of social factors in relation to psychiatric
disorders must not be viewed simplistically. The mitigating effect of social
support, the perception of a stressful life event (as well as the actual
occurrence of the event), the expectancy of an event, and the perceived
importance of an event all may contribute to the effect of environmental
stress on the older adult.
In the study of social factors associated with psychiatric disorders across
the life cycle, context is essential. One study of the social and
environmental characteristics of a healthy community involved an in-depth
evaluation of selected Chicago neighborhoods (not specifically older adults)
(Sampson 2003). The investigators found that physical and mental health
were facilitated by the capacity of residents to achieve social control over
their environment and to engage in collective action for the common good.
They found that the strong ties among neighbors necessary for such
proactive action are no longer the norm in many urban communities
because friends and social support networks are not organized locally.
Given this reality, the investigators proposed a “collective efficacy theory,”
in which they focused on the combination of working trust and shared
willingness of residents to intervene in social control, a move away from
reliance on personal ties. They placed the emphasis on shared beliefs in a
neighborhood’s conjoint capability for action to achieve an effect. This type
of approach may be especially relevant to communal housing arrangements
for older adults.
Summary
From these examples, it is clear that both psychiatric disorders and
symptoms in late life can have multiple causes and that these factors may
interact with one another to produce adverse outcomes. Skoog (2004)
suggested that the science of epidemiology has much to contribute to
increased knowledge of the etiology of mental disorders in older adults and
that to maximize that contribution, future population studies should be
longitudinal and should include assessments of psychosocial risk factors as
well as biological markers such as brain imaging, neurochemical analyses,
and genetic information.
Conclusion
The number of persons age 65 years or older is expected to increase over
the next several decades. Projections show that a higher proportion of these
older adults than observed in previous generations will have experienced a
DSM disorder in their lifetime, putting them at risk for chronic or recurrent
disorders. Epidemiological studies, particularly large-scale, community-
based surveys, can provide clues as to the etiology of these disorders by
observing risk factors as they occur in the general population. These
surveys can also provide important information about the use of health
services among older individuals and help quantify areas where there is
need.
Key Points
• The proportion of older adults in the United States is expected to
dramatically increase over the next 50 years and to be accompanied by
an increase in the number of older adults with psychiatric disorders.
• The prevalence of clinically significant psychiatric symptoms is
generally higher than the prevalence of psychiatric disorders, and the
prevalence of both symptoms and disorders is higher in clinical samples
than in community samples. Psychiatric syndromes, rather than
disorders, are the more realistic diagnostic entities in geriatric psychiatry.
• Alzheimer’s disease is the most prevalent form of dementia, and its
prevalence increases with age.
• Sleep problems, anxiety symptoms, and depressive symptoms are the
most prevalent psychiatric symptoms among older adults.
• The frequency of binge drinking is higher than expected among older
adults.
• Besides dementia, anxiety disorders (particularly phobic disorders) are
the most prevalent psychiatric disorders in older adults in community
samples.
• Genetic, environmental, and social factors, as well as their interaction,
can predispose individuals to psychiatric disorders in late life or be risk
factors for the recurrence of psychiatric symptoms.
• The high suicide rate among older adults is due primarily to the high rate
in older white males.
• Older adults are less likely than younger adults to seek treatment for
mental health problems, and if treatment is sought, it is likely to be
within the primary care setting.
• Even though a high proportion of older adults use psychotropic
medications, psychiatric disorders, particularly depression, are generally
untreated in older persons.
References
Alexopoulos GS, Meyers BS, Young RC, et al: “Vascular depression”
hypothesis. Arch Gen Psychiatry 54(10):915–922, 1997 9337771
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 3rd Edition. Washington, DC, American Psychiatric
Association, 1980
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Anstey KJ, von Sanden C, Sargent-Cox K, et al: Prevalence and risk factors
for depression in a longitudinal, population-based study including
individuals in the community and residential care. Am J Geriatr
Psychiatry 15(6):497–505, 2007 17545450
Bachman DL, Wolf PA, Linn RT, et al: Incidence of dementia and probable
Alzheimer’s disease in a general population: the Framingham Study.
Neurology 43(3 Pt 1):515–519, 1993 8450993
Barnes DE, Kaup A, Kirby KA, et al: Traumatic brain injury and risk of
dementia in older veterans. Neurology 83(4):312–319, 2014 24966406
Beekman ATF, Deeg DJH, van Tilburg T, et al: Major and minor depression
in later life: a study of prevalence and risk factors. J Affect Disord
36(1–2):65–75, 1995 8988267
Black SA, Markides KS, Miller TQ: Correlates of depressive
symptomatology among older community-dwelling Mexican
Americans: the Hispanic EPESE. J Gerontol B Psychol Sci Soc Sci
53(4):S198–S208, 1998 9679521
Bland RC, Newman SC, Orn H: Prevalence of psychiatric disorders in the
elderly in Edmonton. Acta Psychiatr Scand Suppl 338 (suppl 338):57–
63, 1988 3165596
Blazer D: The diagnosis of depression in the elderly. J Am Geriatr Soc
28(2):52–58, 1980a 7351450
Blazer D: Life events, mental health functioning and the use of health care
services by the elderly. Am J Public Health 70(11): 1174–1179, 1980b
7425190
Blazer DG 2nd: Controversies in community-based psychiatric
epidemiology: let the data speak for themselves. Arch Gen Psychiatry
57(3):227–228, 2000 10711908
Blazer DG: Psychiatry and the oldest old. Am J Psychiatry 157(12):1915–
1924, 2000 11097951
Blazer DG, Wu LT: The epidemiology of alcohol use disorders and
subthreshold dependence in a middle-aged and elderly community
sample. Am J Geriatr Psychiatry 19(8):685–694, 2011 21785289
Blazer D, Bachar JR, Hughes DC: Major depression with melancholia: a
comparison of middle-aged and elderly adults. J Am Geriatr Soc
35(10):927–932, 1987a 3655175
Blazer D, Hughes DC, George LK: The epidemiology of depression in an
elderly community population. Gerontologist 27(3):281–287, 1987b
3609795
Blazer D, Swartz M, Woodbury M, et al: Depressive symptoms and
depressive diagnoses in a community population. Use of a new
procedure for analysis of psychiatric classification. Arch Gen
Psychiatry 45(12):1078–1084, 1988 3058087
Blazer D, Woodbury M, Hughes DC, et al: A statistical analysis of the
classification of depression in a mixed community and clinical sample.
J Affect Disord 16(1):11–20, 1989 2521645
Blazer D, Burchett B, Service C, et al: The association of age and
depression among the elderly: an epidemiologic exploration. J Gerontol
46(6):M210–M215, 1991 1834726
Bruce ML, McAvay GJ, Raue PJ, et al: Major depression in elderly home
health care patients. Am J Psychiatry 159(8):1367–1374, 2002
12153830
Busse A, Bischkopf J, Riedel-Heller SG, et al: Mild cognitive impairment:
prevalence and incidence according to different diagnostic criteria.
Results of the Leipzig Longitudinal Study of the Aged (LEILA75+). Br
J Psychiatry 182:449–454, 2003 12724250
Callahan CM, Hendrie HC, Tierney WM: Documentation and evaluation of
cognitive impairment in elderly primary care patients. Ann Intern Med
122(6):422–429, 1995 7856990
Canadian Study of Health and Aging Working Group: Canadian study of
health and aging: study methods and prevalence of dementia. CMAJ
150(6):899–913, 1994 8131123
Centers for Disease Control and Prevention (CDC): Suicide among adults
aged 35–64 years—United States, 1999–2010. MMWR Morb Mortal
Wkly Rep 62(17):321–325, 2013 23636024
Chou KL, Mackenzie CS, Liang K, et al: Three-year incidence and
predictors of first-onset of DSM-IV mood, anxiety, and substance use
disorders in older adults: results from Wave 2 of the National
Epidemiologic Survey on Alcohol and Related Conditions. J Clin
Psychiatry 72(2):144–155, 2011 21382305
Christenson R, Blazer D: Epidemiology of persecutory ideation in an
elderly population in the community. Am J Psychiatry 141(9):1088–
1091, 1984 6235752
Copeland J: What is a “case”? A case for what? In What is a Case: The
Problem of Definition in Psychiatric Community Surveys. Edited by
Wing J, Bebbington P, Robins L. London, Grant McIntyre, 1981, pp 9–
11
Copeland JRM, Dewey ME, Scott A, et al: Schizophrenia and delusional
disorder in older age: community prevalence, incidence, comorbidity,
and outcome. Schizophr Bull 24(1):153–161, 1998 9502553
Cornoni-Huntley J, Brock D, Ostfeld A, et al: Established Populations for
Epidemiologic Studies of the Elderly: Resource Data Book (NIH Publ
No 86–2443). Bethesda, MD, National Institutes of Health, 1986
Demyttenaere K, Bruffaerts R, Posada-Villa J, et al: Prevalence, severity,
and unmet need for treatment of mental disorders in the World Health
Organization World Mental Health Surveys. JAMA 291(21):2581–
2590, 2004 15173149
Eaton WW, Kramer M, Anthony JC, et al: The incidence of specific
DIS/DSM-III mental disorders: data from the NIMH Epidemiologic
Catchment Area Program. Acta Psychiatr Scand 79(2):163–178, 1989
2784251
Eden J, Le M, Maslow K, et al: The Mental Health and Substance Use
Workforce for Older Adults: In Whose Hands? Washington, DC,
National Academies Press, 2012
Evans DA, Funkenstein HH, Albert MS, et al: Prevalence of Alzheimer’s
disease in a community population of older persons. Higher than
previously reported. JAMA 262(18):2551–2556, 1989 2810583
Federal Interagency Forum on Aging-Related Statistics: Older Americans
2012 Key Indicators of Well-Being. Washington, DC, US Government
Printing Office, 2012
Feldman HH, Jacova C: Mild cognitive impairment. Am J Geriatr
Psychiatry 13(8):645–655, 2005 16085780
First MB, Spitzer RL, Gibbon M, et al: Structured Clinical Interview for
DSM-IV Axis I Disorders, Research Version. Washington, DC,
American Psychiatric Association, 1997
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Forsell Y, Winblad B: Feelings of anxiety and associated variables in a very
elderly population. Int J Geriatr Psychiatry 13(7):454–458, 1998
9695033
Forsell Y, Winblad B: Incidence of major depression in a very elderly
population. Int J Geriatr Psychiatry 14(5):368–372, 1999 10389040
Ganguli M, Dodge HH, Shen C, et al: Mild cognitive impairment, amnestic
type: an epidemiologic study. Neurology 63(1):115–121, 2004
15249620
Gatz M, Pedersen NL, Plomin R, et al: Importance of shared genes and
shared environments for symptoms of depression in older adults. J
Abnorm Psychol 101(4): 701–708, 1992 1430610
German PS, Shapiro S, Skinner EA: Mental health of the elderly: use of
health and mental health services. J Am Geriatr Soc 33(4):246–252,
1985 3989185
González HM, Croghan T, West B, et al: Antidepressant use in black and
white populations in the United States. Psychiatr Serv 59(10):1131–
1138, 2008 18832498
González HM, Tarraf W, Whitfield KE, et al: The epidemiology of major
depression and ethnicity in the United States. J Psychiatr Res
44(15):1043–1051, 2010 20537350
Graham JE, Rockwood K, Beattie BL, et al: Prevalence and severity of
cognitive impairment with and without dementia in an elderly
population. Lancet 349(9068): 1793–1796, 1997 9269213
Grant BF, Dawson DA, Hasin DS: The Alcohol Use Disorder and
Associated Disabilities Interview Schedule DSM-IV. Bethesda, MD,
National Institute on Alcohol Abuse and Alcoholism, 2001
Grant BF, Hasin DS, Blanco C, et al: The epidemiology of social anxiety
disorder in the United States: results from the National Epidemiologic
Survey on Alcohol and Related Conditions. J Clin Psychiatry
66(11):1351–1361, 2005a 16420070
Grant BF, Hasin DS, Stinson FS, et al: Prevalence, correlates, co-morbidity,
and comparative disability of DSM-IV generalized anxiety disorder in
the USA: results from the National Epidemiologic Survey on Alcohol
and Related Conditions. Psychol Med 35(12):1747–1759, 2005b
16202187
Grant BF, Hasin DS, Stinson FS, et al: The epidemiology of DSM-IV panic
disorder and agoraphobia in the United States: results from the
National Epidemiologic Survey on Alcohol and Related Conditions. J
Clin Psychiatry 67(3):363–374, 2006 16649821
Gum AM, King-Kallimanis B, Kohn R: Prevalence of mood, anxiety, and
substance-abuse disorders for older Americans in the National
Comorbidity Survey-Replication. Am J Geriatr Psychiatry 17(9):769–
781, 2009 19700949
Hänninen T, Hallikainen M, Tuomainen S, et al: Prevalence of mild
cognitive impairment: a population-based study in elderly subjects.
Acta Neurol Scand 106(3):148–154, 2002 12174174
Harris T, Cook DG, Victor C, et al: Onset and persistence of depression in
older people—results from a 2-year community follow-up study. Age
Ageing 35(1):25–32, 2006 16303774
Hasin DS, Goodwin RD, Stinson FS, et al: Epidemiology of major
depressive disorder: results from the National Epidemiologic Survey on
Alcoholism and Related Conditions. Arch Gen Psychiatry
62(10):1097–1106, 2005 16203955
Hasin DS, Stinson FS, Ogburn E, et al: Prevalence, correlates, disability,
and comorbidity of DSM-IV alcohol abuse and dependence in the
United States: results from the National Epidemiologic Survey on
Alcohol and Related Conditions. Arch Gen Psychiatry 64(7):830–842,
2007 17606817
Hazzard W: Introduction: the practice of geriatric medicine, in Principles of
Geriatric Medicine and Gerontology. Edited by Hazzard W, Bierman E,
Blass J, et al. New York, McGraw-Hill, 1994, pp xxiii–xxiv
Hebert LE, Scherr PA, Beckett LA, et al: Age-specific incidence of
Alzheimer’s disease in a community population. JAMA 273(17):1354–
1359, 1995 7715060
Heeren TJ, Lagaay AM, Hijmans W, et al: Prevalence of dementia in the
“oldest old” of a Dutch community. J Am Geriatr Soc 39(8):755–759,
1991 2071805
Heeringa SG, Wagner J, Torres M, et al: Sample designs and sampling
methods for the Collaborative Psychiatric Epidemiology Studies
(CPES). Int J Methods Psychiatr Res 13(4):221–240, 2004 15719530
Henderson AS, Korten AE, Jacomb PA, et al: The course of depression in
the elderly: a longitudinal community-based study in Australia.
Psychol Med 27(1):119–129, 1997 9122292
Hendrie HC, Hall KS, Ogunniyi A, et al: Alzheimer’s disease, genes, and
environment: the value of international studies. Can J Psychiatry
49(2):92–99, 2004 15065742
Hernandez LM, Blazer DG: Beyond the Nature/Nurture Debate:
Connecting Genes, Behavior, and the Social Environment. Washington,
DC, National Academies Press, 2006
Holmes TH, Rahe RH: The social readjustment rating scale. J Psychosom
Res 11(2):213–218, 1967 6059863
Hulshoff Pol HE, Hoek HW, Susser E, et al: Prenatal exposure to famine
and brain morphology in schizophrenia. Am J Psychiatry 157(7):1170–
1172, 2000 10873931
Jeste DV, Alexopoulos GS, Bartels SJ, et al: Consensus statement on the
upcoming crisis in geriatric mental health: research agenda for the next
2 decades. Arch Gen Psychiatry 56(9):848–853, 1999 12884891
Jimenez DE, Bartels SJ, Cardenas V, et al: Cultural beliefs and mental
health treatment preferences of ethnically diverse older adult
consumers in primary care. Am J Geriatr Psychiatry 20(6):533–542,
2012 21992942
Jongenelis K, Pot AM, Eisses AMH, et al: Prevalence and risk indicators of
depression in elderly nursing home patients: the AGED study. J Affect
Disord 83(2–3):135–142, 2004 15555706
Kessler RC, Ustün TB: The World Mental Health (WMH) Survey Initiative
Version of the World Health Organization (WHO) Composite
International Diagnostic Interview (CIDI). Int J Methods Psychiatr Res
13(2):93–121, 2004 15297906
Kessler RC, Berglund P, Demler O, et al: Life-time prevalence and age-of-
onset distributions of DSM-IV disorders in the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):593–602, 2005
15939837
Kim G, Parton JM, DeCoster J, et al: Regional variation of racial disparities
in mental health service use among older adults. Gerontologist
53(4):618–626, 2013 22859437
Klap R, Unroe KT, Unützer J: Caring for mental illness in the United
States: a focus on older adults. Am J Geriatr Psychiatry 11(5):517–524,
2003 14506085
Kleinbaum DG, Kupper LL, Morgenstern H: Stratified analysis, in
Epidemiologic Research: Principles and Quantitative Methods. New
York, Van Nostrand Reinhold Company, 1982, pp 320–376
Krishnan KRR: Concept of disease in geriatric psychiatry. Am J Geriatr
Psychiatry 15(1):1–11, 2007 17095750
Kroenke K, Spitzer RL, Williams JB: The PHQ-9: validity of a brief
depression severity measure. J Gen Intern Med 16(9):606–613, 2001
11556941
Li S, Yan F, Li G, et al: Is the dementia rate increasing in Beijing?
Prevalence and incidence of dementia 10 years later in an urban elderly
population. Acta Psychiatr Scand 115(1):73–79, 2007 17201869
Lopez OL, Jagust WJ, DeKosky ST, et al: Prevalence and classification of
mild cognitive impairment in the Cardiovascular Health Study
Cognition Study: part 1. Arch Neurol 60(10):1385–1389, 2003
14568808
Luber MP, Meyers BS, Williams-Russo PG, et al: Depression and service
utilization in elderly primary care patients. Am J Geriatr Psychiatry
9(2):169–176, 2001 11316621
Lyketsos CG, Sheppard JM, Steinberg M, et al: Neuropsychiatric
disturbance in Alzheimer’s disease clusters into three groups: the
Cache County study. Int J Geriatr Psychiatry 16(11):1043–1053, 2001
11746650
Lyness JM, King DA, Cox C, et al: The importance of subsyndromal
depression in older primary care patients: prevalence and associated
functional disability. J Am Geriatr Soc 47(6):647–652, 1999 10366161
Manly JJ, Bell-McGinty S, Tang M-X, et al: Implementing diagnostic
criteria and estimating frequency of mild cognitive impairment in an
urban community. Arch Neurol 62(11):1739–1746, 2005 16286549
McCusker J, Cole M, Dufouil C, et al: The prevalence and correlates of
major and minor depression in older medical inpatients. J Am Geriatr
Soc 53(8):1344–1353, 2005 16078960
Meesters PD, de Haan L, Comijs HC, et al: Schizophrenia spectrum
disorders in later life: prevalence and distribution of age at onset and
sex in a Dutch catchment area. Am J Geriatr Psychiatry 20(1):18–28,
2012 22183010
Morris JN: Uses of Epidemiology, 3rd Edition. London, England, Churchill
Livingstone, 1975
Murrell SA, Himmelfarb S, Wright K: Prevalence of depression and its
correlates in older adults. Am J Epidemiol 117(2):173–185, 1983
6829547
National Center for Health Statistics: Health, United States, 2012: With
Special Feature on Emergency Care. Hyattsville, MD, U.S.
Government Printing Office, 2012
Olafsdóttir M, Marcusson J, Skoog I: Mental disorders among elderly
people in primary care: the Linköping study. Acta Psychiatr Scand
104(1):12–18, 2001 11437744
Ostling S, Skoog I: Psychotic symptoms and paranoid ideation in a
nondemented population-based sample of the very old. Arch Gen
Psychiatry 59(1):53–59, 2002 11779282
Palsson SP, Ostling S, Skoog I: The incidence of first-onset depression in a
population followed from the age of 70 to 85. Psychol Med
31(7):1159–1168, 2001 11681542
Panza F, D’Introno A, Colacicco AM, et al: Current epidemiology of mild
cognitive impairment and other predementia syndromes. Am J Geriatr
Psychiatry 13(8): 633–644, 2005 16085779
Parmelee PA, Katz IR, Lawton MP: Depression among institutionalized
aged: assessment and prevalence estimation. J Gerontol 44(1):M22–
M29, 1989 2783434
Paulose-Ram R, Safran MA, Jonas BS, et al: Trends in psychotropic
medication use among U.S. adults. Pharmacoepidemiol Drug Saf
16(5):560–570, 2007 17286304
Pietrzak RH, Kinley J, Afifi TO, et al: Subsyndromal depression in the
United States: prevalence, course, and risk for incident psychiatric
outcomes. Psychol Med 43(7): 1401–1414, 2013 23111093
Plassman BL, Havlik RJ, Steffens DC, et al: Documented head injury in
early adulthood and risk of Alzheimer’s disease and other dementias.
Neurology 55(8):1158–1166, 2000 11071494
Plassman BL, Langa KM, Fisher GG, et al: Prevalence of dementia in the
United States: the aging, demographics, and memory study.
Neuroepidemiology 29(1–2):125–132, 2007 17975326
Plassman BL, Langa KM, Fisher GG, et al: Prevalence of cognitive
impairment without dementia in the United States. Ann Intern Med
148(6):427–434, 2008 18347351
Radloff LS: The CES-D scale: A self-report depression scale for research in
the general population. Appl Psychol Meas 1:385–401, 1977
Regier DA, Myers JK, Kramer M, et al: The NIMH Epidemiologic
Catchment Area program. Historical context, major objectives, and
study population characteristics. Arch Gen Psychiatry 41(10):934–941,
1984 6089692
Regier DA, Boyd JH, Burke JD Jr, et al: One-month prevalence of mental
disorders in the United States. Based on five Epidemiologic Catchment
Area sites. Arch Gen Psychiatry 45(11):977–986, 1988 3263101
Reynolds CF 3rd, Frank E, Perel JM, et al: Nortriptyline and interpersonal
psychotherapy as maintenance therapies for recurrent major
depression: a randomized controlled trial in patients older than 59
years. JAMA 281(1):39–45, 1999 9892449
Riedel-Heller SG, Busse A, Aurich C, et al: Prevalence of dementia
according to DSM-III-R and ICD-10: results of the Leipzig
Longitudinal Study of the Aged (LEILA75+), part 1. Br J Psychiatry
179: 250–254, 2001 11532803
Robins LN, Helzer JE, Croughan J, et al: National Institute of Mental
Health Diagnostic Interview Schedule. Its history, characteristics, and
validity. Arch Gen Psychiatry 38(4):381–389, 1981 6260053
Rosch E: Principles of categorization, in Cognition and Categorization.
Edited by Rosch E, Lloyd B. Hillsdale, NJ, Lawrence Erlbaum, 1978,
pp 3–27
Roses AD: Apolipoprotein E affects the rate of Alzheimer disease
expression: beta-amyloid burden is a secondary consequence
dependent on APOE genotype and duration of disease. J Neuropathol
Exp Neurol 53:429–437, 1994 8083686
Roses AD: Genetic testing for Alzheimer disease. Practical and ethical
issues. Arch Neurol 54(10):1226–1229, 1997 9341568
Rovner BW, Kafonek S, Filipp L, et al: Prevalence of mental illness in a
community nursing home. Am J Psychiatry 143(11): 1446–1449, 1986
3777239
Sachdev PS, Lipnicki DM, Crawford J, et al: Risk profiles for mild
cognitive impairment vary by age and sex: the Sydney Memory and
Ageing study. Am J Geriatr Psychiatry 20(10):854–865, 2012
22673190
Sampson RJ: The neighborhood context of well-being. Perspect Biol Med
46 (3 suppl): S53–S64, 2003 14563074
Shapiro S, Skinner EA, Kessler LG, et al: Utilization of health and mental
health services. Three Epidemiologic Catchment Area sites. Arch Gen
Psychiatry 41(10): 971–978, 1984 6477055
Sheehan B, Bass C, Briggs R, et al: Somatization among older primary care
attenders. Psychol Med 33(5):867–877, 2003 12877401
Shoghi-Jadid K, Small GW, Agdeppa ED, et al: Localization of
neurofibrillary tangles and beta-amyloid plaques in the brains of living
patients with Alzheimer disease. Am J Geriatr Psychiatry 10(1):24–35,
2002 11790632
Skoog I: Psychiatric epidemiology of old age: the H70 study—the NAPE
lecture 2003. Acta Psychiatr Scand 109(1):4–18, 2004 14674953
Smalbrugge M, Pot AM, Jongenelis K, et al: Prevalence and correlates of
anxiety among nursing home patients. J Affect Disord 88(2):145–153,
2005 16122811
Stek ML, Gussekloo J, Beekman ATF, et al: Prevalence, correlates and
recognition of depression in the oldest old: the Leiden 85-plus study. J
Affect Disord 78(3):193–200, 2004 15013243
Stevens T, Livingston G, Kitchen G, et al: Islington study of dementia
subtypes in the community. Br J Psychiatry 180:270–276, 2002
11872521
Stinson FS, Dawson DA, Patricia Chou S, et al: The epidemiology of DSM-
IV specific phobia in the USA: results from the National
Epidemiologic Survey on Alcohol and Related Conditions. Psychol
Med 37(7):1047–1059, 2007 17335637
Strain J, Didehbani N, Cullum CM, et al: Depressive symptoms and white
matter dysfunction in retired NFL players with concussion history.
Neurology 81(1):25–32, 2013 23709590
Strauss JS, Gabriel KR, Kokes RF, et al: Do psychiatric patients fit their
diagnoses? Patterns of symptomatology as described with the biplot. J
Nerv Ment Dis 167(2): 105–113, 1979 368291
Susser ES, Lin SP: Schizophrenia after prenatal exposure to the Dutch
Hunger Winter of 1944–1945. Arch Gen Psychiatry 49(12):983–988,
1992 1449385
Teresi J, Abrams R, Holmes D, et al: Prevalence of depression and
depression recognition in nursing homes. Soc Psychiatry Psychiatr
Epidemiol 36(12):613–620, 2001 11838834
Trollor JN, Anderson TM, Sachdev PS, et al: Prevalence of mental
disorders in the elderly: the Australian National Mental Health and
Well-Being Survey. Am J Geriatr Psychiatry 15(6):455–466, 2007
17545446
Unützer J, Simon G, Belin TR, et al: Care for depression in HMO patients
aged 65 and older. J Am Geriatr Soc 48(8):871–878, 2000 10968289
Wang PS, Berglund P, Olfson M, et al: Failure and delay in initial treatment
contact after first onset of mental disorders in the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):603–613, 2005a
15939838
Wang PS, Lane M, Olfson M, et al: Twelve-month use of mental health
services in the United States: results from the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):629–640, 2005b
15939840
Wang PS, Demler O, Olfson M, et al: Changing profiles of service sectors
used for mental health care in the United States. Am J Psychiatry
163(7):1187–1198, 2006 16816223
Weissman MM, Klerman GL: Epidemiology of mental disorders: emerging
trends in the United States. Arch Gen Psychiatry 35(6):705–712, 1978
655769
World Health Organization: Composite International Diagnostic Interview
(CIDI, Version 1.0). Geneva, World Health Organization, 1990
Yesavage JA, Brink TL, Rose TL, et al: Development and validation of a
geriatric depression screening scale: a preliminary report. J Psychiatr
Res 17(1):37–49, 1982–1983 7183759
Suggested Readings
Eden J, Le M, Maslow K, Blazer D: The Mental Health and Substance Use
Workforce for Older Adults: In Whose Hands? Washington, DC,
National Academies Press, 2012
Gum AM, King-Kallimanis B, Kohn R: Prevalence of mood, anxiety, and
substance-abuse disorders for older Americans in the National
Comorbidity Survey-Replication. Am J Geriatr Psychiatry 17(9):769–
781, 2009
Jeste DV, Alexopoulos GS, Bartels SJ, et al: Consensus statement on the
upcoming crisis in geriatric mental health: research agenda for the next
2 decades. Arch Gen Psychiatry 56(9):848–853, 1999
Kessler RC, Berglund P, Demler O, et al: Lifetime prevalence and age-of-
onset distributions of DSM-IV disorders in the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):593–602, 2005
Wang PS, Lane M, Olfson M, et al: Twelve-month use of mental health
services in the United States: results from the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):629–640, 2005
CHAPTER 2
—Robert Browning
Will you still need me, Will you still feed me When I’m sixty-four?
Cardiovascular System
The heart and blood vessels of aging individuals undergo significant
anatomical alterations. These structural changes lead to changes in function.
In addition, age-associated changes occur in the autonomic nervous system
and in the response of the cardiovascular system to it, and these changes
have important physiological effects. The ability of the heart to beat faster
and pump efficiently and the ease with which blood vessels dilate or
constrict are markedly affected. Both cardiac output and cardiac reserve
decrease (O’Rourke and Hashimoto 2006; Seals and Esler 2000).
With age, human blood vessels stiffen. Anatomically, the intima and
media both thicken with age, but according to autopsy studies, this
thickening occurs disproportionately in the intima, with intimal hyperplasia
observed. Oxidative stress and vascular inflammation may both contribute
to endothelial changes and dysfunction. The vessels are thicker and less
distensible. The ability of arterial vessels to transmit blood is not
appreciably affected, but the cushioning effect of the arterial system is
adversely altered. The physiological results are a greater pulse wave
velocity, early reflected pulse waves, and higher systolic blood pressures
and aortic pulse pressures in older individuals. Higher pressures can
increase the load on the heart and lead to left ventricular enlargement as
well as increased left ventricular oxygen requirements, thereby increasing
the risk of congestive heart failure. There is an increased need for coronary
blood flow oxygen, but there is a decreased ability to provide such flow due
to decreased aortic pressure during diastole as well as a reduced period of
diastole. This situation may cause cardiac ischemia by worsening
ventricular function and cardiac perfusion during diastole. Increased
pressures can also result in damage to the endothelium and media in the
microcirculation of the brain and kidneys and may be connected with organ
dysfunction (O’Rourke and Hashimoto 2006; Ungvari et al. 2010).
The function of the heart during exercise, including the force and rate of
contraction, is mediated by the sympathetic nervous system. Age-related
changes in that system affect the adaptability of the heart and blood vessels
to stress. In general, sympathetic nervous system activity rises in elderly
patients, as evidenced by higher circulating levels of norepinephrine.
Norepinephrine fills cardiac and vascular surface cell receptors, making
them less sensitive. The β-adrenergic response of the heart during exercise
is attenuated; a lower maximum heart rate and decreased force of
contraction are the result. Similarly, large arteries do not respond as well to
β-adrenergic stimulation, and their ability to dilate is reduced (Lakatta
1999; Seals and Esler 2000).
The older heart dilates during exercise to increase end-diastolic volume
and maintain stroke volume, but cardiac output nonetheless declines with
age. Because the heart stiffens, it empties less completely. This is the result
of several factors, including increased afterload, decreased contractility of
the heart, and the reduced inotropic effect of β-adrenergic stimulation to the
heart. The decline in cardiac output also adversely affects oxygen use in
older adults. The decline in cardiac function with age may explain 50% of
the reduction in maximum oxygen consumption that occurs (Lakatta 1999).
Respiratory System
As individuals age, the chest wall becomes stiffer and less compliant. From
age 24 to age 75 years, compliance declines by 31%. A number of factors
contribute to stiffening of the chest wall, including degenerative joint
disease and the calcification of the costal cartilages and chondrosternal
junction. In older individuals, osteoporosis may cause partial or wedge
fractures of the vertebrae, with resulting kyphosis and an increase in the
anteroposterior diameter of the thorax; these physical alterations can in turn
adversely affect chest wall mechanics. When an older individual generates a
breath, the relative contribution of the diaphragm and abdominal muscles is
increased compared with the thoracic muscles, as a result of decreased chest
wall compliance. Respiratory muscle function also declines because of
changes in the rib cage, decreased chest wall compliance, and decreased
elastic recoil of the lung. Respiratory muscles weaken with age, as a result
of nutritional deficiencies, anatomical changes in skeletal muscle, and
physiological decline (Janssens 2005; Sharma and Goodwin 2006).
Changes in the lung itself and in the control of breathing negatively
affect the respiratory system. A loss of elastic tissue in the lung occurs, with
a loss of elastic recoil, and the alveolar ducts and respiratory bronchioles
enlarge. This enlargement leads to a loss of alveolar surface area; less tissue
is available for gas exchange, and partial pressure of oxygen (PO2)
decreases with age but the decline is not uniform. The diffusing capacity of
the lung also declines. Higher closing volumes make full expansion of the
airways more difficult, especially in the dependent areas of the lung. The
loss of surface area combined with decreased expansion results in
ventilation-perfusion mismatch and decreased oxygenation. The control of
breathing is also altered in elderly patients. Low oxygen tension and high
carbon dioxide levels fail to provide the same physiological stimulus to
breathe, but the decreased response to hypercapnia is not consistent in all
studies.
In older people, the lung is less able to guard itself against infection
(Taffet 1999). The mucociliary tree lining the respiratory tract is slower in
trapping and expelling invading particles and microorganisms (Ho et al.
2001). With age, the person’s ability to generate a sufficiently strong cough
declines. The development of higher closing volumes further complicates
defense against infection by making it harder to expel secretions from the
lower areas of the lungs.
Some studies suggest that exercise training can slow the respiratory
decline that occurs with aging. The age-related decrease in maximum
oxygen consumption, as well as the decreased responsiveness to low
oxygen tension or high carbon dioxide levels, can improve with exercise.
Although exercise is helpful, it cannot prevent the eventual decline in
pulmonary function (Schwartz and Kohrt 2003). The extent to which
exercise is helpful may not be consistent across all groups of older patients
(Sharma and Goodwin 2006).
Gastrointestinal System
As people age, numerous anatomical changes take place throughout the
gastrointestinal tract, some of which are functionally significant (Firth and
Prather 2002; Hall and Wiley 2003; Majumdar et al. 1997; Taffet 1999).
The production of saliva declines with age. A recent study demonstrated
that healthy adults ages 70 years and older have lower levels of stimulated
whole saliva than do adults ages 50 years and younger (Smith et al. 2013).
In older individuals, receding gums make the teeth more susceptible to
dental caries and subsequent tooth loss. The mastication of food may be
incomplete. There are fewer myenteric ganglion cells, which affects the
coordination of swallowing and may predispose some elderly patients to
aspiration. The strength of esophageal contractions is diminished, but food
nonetheless traverses the length of the esophagus uneventfully. The
production of acid and pepsin by the stomach is mostly preserved. Both the
stomach and the small intestine do not dilate as easily as a bolus of food
enters, and transit through the large bowel may be slower. The small bowel
is less effective at absorbing vitamins and minerals (e.g., vitamin D,
calcium, and iron) and sugars (e.g., xylose and lactose). The motor function
of the colon is not significantly affected by aging.
The liver, gallbladder, and pancreas continue to function well in elderly
patients (Hall and Wiley 2003; Majumdar et al. 1997; Oskvig 1999; Taffet
1999). The liver loses hepatocytes and becomes smaller; those cells
remaining are less able to regenerate. In general, the liver’s ability to
manufacture binding proteins and metabolize drugs is stable, although
considerable variability can be found across individuals. The liver makes
fewer vitamin K–dependent clotting factors. Liver transaminases and
alkaline phosphatase remain unchanged. Few significant anatomical
changes occur in the gallbladder, and its function remains intact. In the
aging pancreas, there is parenchymal fibrosis, acinar atrophy, and fatty
infiltration but no resulting impairment in the synthesis of pancreatic
enzymes and bicarbonate. The role of the pancreas in digestive function is
therefore unaffected.
Endocrine System
Prolactin
Levels of prolactin in aging women have been reported to increase,
decrease, or remain the same, whereas those in aging men are slightly
increased. None of these changes is believed to have an effect on normal
function (Gruenewald and Matsumoto 2003).
Antidiuretic Hormone
Aging causes significant changes in antidiuretic hormone (ADH) and the
body’s response to it, which alter the older patient’s ability to excrete free
water—resulting in hyponatremia—or to prevent volume losses—resulting
in dehydration. Basal ADH levels are normal to increased in older
individuals; because renal free water clearance decreases with age,
hyponatremia can more easily occur. However, when volume loss takes
place, with subsequent hypotension, less ADH is released in older
individuals. In this particular clinical situation, other age-related changes
are also at work to produce dehydration: 1) the kidney is less responsive to
ADH, which impairs its effort to make more concentrated urine, and 2)
aldosterone activity decreases and natriuretic hormone activity increases,
both of which inhibit renal conservation of sodium and restoration of
normal volume. The impaired thirst mechanism in elderly individuals
further exacerbates this scenario by preventing them from drinking
adequate amounts of fluid to correct free water losses, thereby contributing
further to dehydration (Gruenewald and Matsumoto 2003; Oskvig 1999;
Perry 1999).
Adrenal Androgens
Both dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate
(DHEA-S) decrease significantly in older individuals. DHEA production
peaks at age 20 years and then declines (Fried and Walston 2003;
Gruenewald and Matsumoto 2003).
Growth Hormone
Growth hormone levels peak at puberty and then decrease by 14% per
decade. Both a decrease in growth hormone–releasing hormone secretion
and an increase in somatostatin are responsible for the decline in growth
hormone. Insulin-like growth factor (IGF-1), which is produced by the liver
and mediates the actions of growth hormone in the body, also diminishes
gradually, at a rate of 7%–13% per decade. The decline in growth hormone
with age may result in a decrease in both lean body mass and bone mass
(Gruenewald and Matsumoto 2003; Perry 1999).
Estrogen
Estrogen declines precipitously with menopause. Both fibrosis and
involution of the ovary, as well as atrophy of the uterus and vagina, take
place. The number of ovarian follicles declines, and a corresponding
decrease in the secretion of both estrogen and androgens occurs; after
menopause, the ratio of estrogens to androgens decreases. Menopause is
also marked by an alteration in gonadotropin-releasing hormone secretion
and high follicle-stimulating hormone levels, although luteinizing hormone
levels remain the same. The loss of estrogen affects bone mass and places
women at risk for osteoporosis. Women also lose the beneficial effects of
estrogen on lipids, with rising low-density lipoprotein levels, and are at
higher risk for cardiovascular disease. The lack of estrogen causes atrophy
of the vaginal endothelium; the endothelium thins, less lubrication occurs
with intercourse, and dyspareunia can result (Gruenewald and Matsumoto
2003; Perry 1999; Taffet 1999). The results of a widely cited randomized
clinical trial, the Women’s Health Initiative, demonstrated that estrogen
replacement in postmenopausal women did not produce the expected
reductions in cardiovascular events, stroke, fractures, and dementia that had
been anticipated based on previous observational studies (Grimes and Lobo
2002; Nelson et al. 2002). Some obstetricians and gynecologists have
nonetheless suggested that estrogen can be safely prescribed, at lower
dosages than those given in the Women’s Health Initiative trial, for the
relief of common postmenopausal symptoms such as hot flashes (Grimes
and Lobo 2002).
Thyroid
Although there are age-related changes in the thyroid gland, these changes
have no corresponding effect on thyroid function. The aging thyroid is more
fibrotic and nodular in composition and decreases in size. Overall, thyroid-
stimulating hormone tends to rise with age. Thyroid autoantibodies tend to
be higher in women than in men. Although the thyroid continues to make
sufficient amounts of thyroxine (T4), it fails to metabolize T4 as well. The
synthesis of T4 actually declines, but its level is unchanged. Peripheral
deiodination of T4 to triiodothyronine (T3) also decreases, and the level of
T3 declines by 10%–20% in elderly people. Reverse T3 levels do not
change. Thyroxine-binding globulin levels remain normal with age
(McNabney and Fedarko 2013;Veldhuis 2013).
Insulin
Elderly patients have a tendency toward hyperglycemia. Circulating insulin
levels may rise but are less efficiently utilized. Although insulin secretion
by the pancreatic β cells is preserved with age, insulin clearance declines
and insulin levels increase. Peripheral uptake of insulin is affected by
insulin resistance in peripheral tissues; some of these tissues, particularly
adipocytes, have fewer receptors, thereby decreasing their sensitivity to
insulin.
The presence in the bloodstream of free fatty acids and inflammatory
adipocytokines may also contribute to pancreatic β cell dysfunction. Elderly
individuals have decreased muscle mass and a higher percentage of fat and
therefore an increased number of adipocytes. These notable changes in
insulin secretion and tissue sensitivity in the periphery may lead to observed
increases in fasting glucose in older individuals. Another factor that leads to
higher glucose levels is that IGF-1, which acts at insulin receptors to
promote glucose uptake, is less abundant in older adults (Halter 2003; Perry
1999; Rizvi 2007; Taffet 1999). A number of other factors contribute to the
increased prevalence of glucose intolerance and type II diabetes mellitus in
elderly people, including changes in body composition, a reduction in
physical activity, and increased comorbid illness and medication use.
According to the Centers for Disease Control and Prevention (2014a), the
incidence of diagnosed diabetes in the U.S. population ages 65–79 years
was 15.4% in 2011.
Musculoskeletal System
In general, elderly people are weaker and less muscular. Sarcopenia, a
decline in skeletal muscle mass, occurs. In the fourth decade, both muscle
mass and strength begin to decrease. There are smaller numbers of type II
fast-twitch fibers and fewer motor units and synapses; slow muscle fibers
predominate. The motor units may enlarge. There are fewer and smaller
ventral spinal motor neurons in the cervical and lumbar regions. Age,
however, does not affect conduction velocity. Biochemical changes also
occur, with decreased activity of glycolytic enzymes, including
triosephosphate dehydrogenase, lactate dehydrogenase, glycerophosphate
dehydrogenase, and citrate synthase. Exercise may modify age-associated
changes in muscle mass and strength. Sarcopenia places older people at risk
for significant physical disability and a decline in their ability to perform
activities of daily living (ADLs) and may ultimately undermine their ability
to live independently (Loeser and Delbono 2003; Taffet 1999).
Elderly people also develop demonstrable changes in cartilage, tendons,
and ligaments. Cartilage becomes less cellular with age. Alterations in the
structure of proteoglycans affect the ability of these molecules to bind water
and maintain the hydration of cartilage. Cartilage weakens as the number of
proteoglycan monomers decreases and the protein links between the
monomers are broken. An increase in collagen cross-linking and in the
diameter of collagen fibrils occurs, which makes collagen stiffer; this
stiffness results in the compression of proteoglycans, which further
interferes with water retention. The overall effect of age-related changes in
cartilage is to decrease both its tensile strength and its stiffness, adversely
affecting its response to mechanical stress (Loeser and Delbono 2003;
Taffet 1999).
Older tendons and ligaments may be stiffer because of an increase in
cross-linking of type I collagen, and their water content is also decreased.
These alterations may make them less able to withstand mechanical stress
and more susceptible to fatigue. Biomechanical studies confirm the
weakening of tendons and ligaments with age. These changes may decrease
the range of motion of joints in older adults and make them more prone to
tendonitis, ligament tears, and ligament ruptures (Loeser and Delbono 2003;
Taffet 1999).
Age-related changes in the structure of both cortical and trabecular bone
occur. Cortical bone becomes thinner and more porous; trabecular bone also
thins, and whole trabeculae are lost. Bones are therefore weaker. As an
individual ages, the number of osteoblasts and osteocytes may decrease.
Osteoclasts continue to function normally. Mechanical strain, an important
stimulus to bone formation, has less of an effect in older people, and less
bone is made. Older individuals are at increased risk for bone loss. Without
estrogen replacement, women can lose significant bone mass after
menopause. Elderly men with testosterone deficiency also may develop
osteoporosis. Other factors that contribute to bone loss in both men and
women include low peak bone density, poor calcium intake, secondary
hyperparathyroidism (as discussed earlier), and insufficient exercise
(Prestwood and Duque 2003). Bisphosphonates have been studied
extensively and widely prescribed to treat osteoporosis, although reports of
unusual fractures in patients on long-term therapy have led to
reconsideration of exactly how long these medications should be prescribed
(Whitaker et al. 2012). Concern about the routine use of calcium
supplementation and the resultant increased risk of coronary artery disease
has also been raised in a systematic review (Bolland et al. 2010).
Renal System
As a person ages, a progressive decrease occurs in the size of the kidney
due to fatty infiltration, fibrosis, and the dropout of cortical nephrons. The
rate of decline of nephrons is 0.5%–1.0% per year; by age 60, 30%–50% of
functioning glomeruli have been eliminated. Cortical nephrons become
diffusely sclerotic. Glomeruli outside the cortex have fewer capillary loops
and epithelial cells, but more mesangial cells. Other areas of the kidney
may also be affected; interstitial fibrosis may adversely affect the renal
pyramids. These losses do not automatically lead to a failure of the kidney
to keep fluids and electrolytes in balance, but decreased reserve capacity
does predispose the kidney to possible dysfunction or failure. Creatinine
clearance, a widely accepted measure of kidney function, declines 7.5%–
10.0% per decade (Oskvig 1999; Taffet 1999; Weinstein and Anderson
2010).
These anatomical changes have important physiological consequences,
including the decreased ability of the kidney to acidify urine or to excrete
an acid or a water load. The response of the renin-angiotensin-aldosterone
system is less supple, renin activity declines, and less renin is produced in
the face of decreased intravascular volume or a depletion of salt. The
kidney is able to maintain its output of erythropoietin, but the hydroxylation
of vitamin D declines. Levels of atrial natriuretic peptide rise. The kidney
less reliably metabolizes hormones such as glucagon, calcitonin, and
parathyroid hormone; drug metabolism is also significantly affected
(Oskvig 1999; Taffet 1999; Weinstein and Anderson 2010), as discussed in
the next subsection, “Effects of Aging on Pharmacokinetics and
Pharmacodynamics.”
Pharmacokinetics
Absorption
Age has no significant effect on drug absorption. Although acid secretion,
gastrointestinal perfusion, and membrane transport all may decrease and
thereby lower absorption, gastrointestinal transit time is prolonged and
increases absorption, and thus no net change occurs.
Volume of Distribution
The volume of distribution is significantly affected by the changes in body
mass and total body water that occur with aging. Older patients, with
decreased lean body mass and total body water, have a smaller volume of
distribution. This is particularly relevant when choosing proper dosages for
drugs, such as antibiotics or lithium, that are primarily distributed in water.
Protein binding also can affect the volume of distribution; it is generally
unaffected by age. In frail elderly patients, there may be significant
decreases in albumin levels, which affect the binding of potentially harmful
drugs such as warfarin, which must be vigilantly titrated.
Clearance Rate
With age, renal mass and renal blood flow are decreased, resulting in a
decline in glomerular filtration rate and creatinine clearance. This decrease
in clearance can alter the rate at which drugs are excreted, and dosages must
be appropriately adjusted. Certain drugs, such as nonsteroidal anti-
inflammatory drugs and angiotensin-converting enzyme inhibitors, also
may alter renal blood flow and thereby depress kidney function. Hepatic
drug clearance is decreased by an age-related decline in hepatic blood flow;
oxidative metabolism in the cytochrome P450 system is slower, thereby
affecting elimination, but conjugation is not slower. Underlying hepatic
disease and drug interactions also may significantly affect the metabolism
of drugs by the liver.
Elimination Half-Life
The elimination half-life—the time required for the drug concentration to
decrease by half—of certain drugs increases in older adults. This may
require adjustment of the drug dosing interval. For example, aspirin, certain
antibiotics (e.g., vancomycin), digoxin, and the calcium channel blockers
(diltiazem, felodipine, and nifedipine) all have higher elimination half-lives,
and the dosages must be adjusted downward.
Pharmacodynamics
The pharmacodynamic effects of drugs in elderly patients must also be
considered. Frequently, older adults are more sensitive to medications, and
drugs often must be given in lower doses. For example, because their
response to anticholinergic drugs is increased, elderly individuals develop
side effects, including constipation, urinary retention, and delirium, more
frequently than do younger individuals. Other notable examples of drugs
with enhanced pharmacodynamic effects in elderly people include
diazepam, morphine, and theophylline.
Geriatric Syndromes
Several common syndromes—known generally as geriatric syndromes—
are found more frequently in older patients. Elderly people can be
predisposed to delirium, a waxing and waning disorder of inattention, and
many elderly patients develop dementia, which doubles in the population
every 5 years after age 65. Falls are multifactorial phenomena that increase
with age. Both elderly men and elderly women often develop chronic
difficulty with control of urination, or urinary incontinence. The tendency
of elderly people to use multiple drugs—or polypharmacy—is also a well-
known geriatric syndrome. Frailty is a complex geriatric syndrome in which
the health of the elderly individual declines after cumulative loss of
physiological reserve, with sarcopenia, osteopenia, weight loss, and
progressive functional deterioration. Five of the most characteristic geriatric
syndromes—dementia, falls, urinary incontinence, polypharmacy, and
frailty—are discussed in the following subsections.
Falls
Falls are a common phenomenon in older patients; every year, one-half of
all nursing home residents and one-third of all community-dwelling elderly
have a fall. These falls produce notable morbidity: 20%–30% of falls result
in moderate to severe injuries such as lacerations, hip fractures, and head
trauma (Centers for Disease Control 2014b). Half of the persons who fall
experience minor injuries. In the aftermath of falls, disability may result.
People who fall frequently are at risk for a decline in their ADLs and
instrumental ADLs (IADLs) (assessment of such functions is discussed
later in this chapter in the subsection “Fundamentals of Geriatric
Assessment”). A decline in these functions can ultimately undermine
independence and also might result in hospitalization. The leading cause of
death from injury in adults over age 65 is complications resulting from falls
(Berry and Kiel 2013; Fried 2000; King and Tinetti 1995; Rubenstein and
Josephson 2006; Rubenstein et al. 1994).
Falls are generally multifactorial and are caused by 1) intrinsic factors,
2) situational factors, 3) extrinsic factors, and 4) medications (Alexander
1999; King and Tinetti 1995). Intrinsic factors are disease-specific deficits
in an individual patient that might contribute to falling; these factors include
neurological problems (central, neuromuscular, vestibular, visual, and
proprioceptive) as well as systemic illness. Situational factors relate to the
particular activity that is taking place. Extrinsic factors relate to the
demands and hazards of a particular environment. Medications may
adversely affect mental status, cognition, balance, circulation, and
neuromuscular function and therefore predispose patients to falls.
The proper evaluation of a fall requires 1) taking a detailed history and
review of systems and 2) performing a thorough physical examination and
neurological examination (Rubenstein and Josephson 2006). The fall may
indeed be a nonspecific presentation of a serious medical condition such as
cardiac ischemia, infection, intravascular volume depletion, or
hypothyroidism, and these should be initially considered. The clinician
should ask about any symptoms and situational or extrinsic factors that
might have led to the fall and determine exactly how the fall occurred. A
medication list should be compiled. The physical examination should rule
out any cardiac abnormalities; the neurological examination must carefully
assess the patient for any deficits in vision, strength, sensation, joint
mobility, balance, cerebellar function, gait, or proprioception.
The prevention of falls focuses on altering both intrinsic and extrinsic
factors (Gillespie et al. 2003; King and Tinetti 1995; Rubenstein and
Josephson 2006). With regard to intrinsic factors, one can 1) prescribe
medication appropriately, 2) optimally treat disease, 3) improve balance and
gait through physical therapy, and 4) improve conditioning and strength
through exercise. With regard to extrinsic factors, one can 1) improve the
environment by reducing or eliminating hazards, 2) monitor patients more
carefully by increasing staff supervision and using motion detection, 3)
eliminate restraints and the risk of injury they pose, 4) encourage patients to
wear hip protectors, and 5) install protective flooring. Preventing falls
ultimately requires multiple steps to produce successful results.
A meta-analysis of five randomized clinical trials suggested that vitamin
D supplementation may reduce the risk of falls in ambulatory or
institutionalized older individuals by more than 20% (Bischoff-Ferrari et al.
2004).
Urinary Incontinence
Urinary incontinence is a prevalent condition in older adults that causes
significant morbidity and affects quality of life (DuBeau 2006; Tannenbaum
et al. 2001). One-half of all nursing home residents and up to one-third of
persons older than age 65 years who reside in the community carry the
diagnosis. It is a condition with multiple causes, including age-related
changes, genitourinary tract abnormalities, and coexisting illnesses.
Urinary incontinence can be classified into two main categories: 1)
transient incontinence and 2) established incontinence. Transient
incontinence is reversible and can be easily treated. For example, transient
incontinence, which could be a consequence of an acute urinary tract
infection, inadequately controlled diabetes mellitus, or a recent prescription
of a diuretic, will resolve with the correction of those conditions.
Established incontinence is divided into the following three subcategories:
Polypharmacy
Polypharmacy, defined as the simultaneous use of multiple medications or
the prescribing of more medications than is clinically appropriate, is a
common problem in older adults (Hanlon et al. 2001; Stewart 2001). The
high use of drugs by elderly individuals is attributable to several factors.
Older patients more commonly have chronic illness and experience more
symptoms. The elderly are also more frequent consumers of medical care.
Drug use is also influenced by individual physician prescribing practices
and by drug advertising. According to studies conducted in outpatients,
elderly patients typically use 3.1–7.9 prescription and nonprescription drugs
simultaneously. Polypharmacy carries with it certain consequences,
including adverse drug reactions, drug interactions, and patient
noncompliance; polypharmacy also increases the incidence of geriatric
syndromes such as urinary incontinence, falls, cognitive impairment, and
delirium.
Clinicians should take several steps to ensure that medicines are
prescribed appropriately. They should take a careful, comprehensive
medication history, including allergies and adverse drug reactions. Current
use of alcohol, tobacco, and recreational drugs should be documented.
Medications should be prescribed only if they have a known benefit, and
they should be given at the lowest effective dose. Instructions about
medication use should be communicated clearly to patients. Patients who
are taking medication should be carefully monitored for therapeutic
effectiveness and for side effects (Semla and Rochon 2006). In a systematic
review of 14 clinical trials on optimizing prescribing in older individuals,
Spinewine et al. (2007) reported that certain clinical interventions,
including geriatric medicine services, the participation of a pharmacist in
clinical care, and computerized decision support, had beneficial effects on
prescribing. In 2012, an expert panel of the American Geriatrics Society
revised the Beers Criteria for Potentially Inappropriate Medication Use in
Older Adults and issued new evidence-based guidelines (American
Geriatrics Society 2012 Beers Criteria Update Expert Panel 2012).
Frailty
The sum effect of physiological decline in the older individual, combined
with the cumulative and simultaneous burden of chronic disease, may result
in the geriatric syndrome known as frailty (Cohen 2000; Hamerman 1999;
Morley et al. 2006). Frailty has been defined by Fried and Walston (2003)
as “a state of age-related physiologic vulnerability resulting from impaired
homeostatic mechanisms and a reduced capacity of the organism to
withstand stress” (p. 1489). Older patients have less pulmonary, cardiac,
and renal reserve. They are less able to mount an effective immune
response. They also have higher sympathetic nervous tone, which may
increase cortisol production and further impair the immune system. In older
patients, cortisol also may have catabolic effects on bone and muscle and
result in insulin resistance. Elderly patients also may have higher levels of
circulating cytokines—such as interleukin-6, interleukin-1B, and tumor
necrosis factor-α—which also may have deleterious catabolic effects on
muscle. Changes in neuroendocrine function—the decline in sex steroids,
growth hormone, and DHEA—can have corresponding negative effects on
the size and strength of muscle and, in the case of estrogen and testosterone,
on bone mass. The frail older individual is characteristically weak as a
result of declining muscle and bone mass; a tendency toward a sedentary
state may lead to deconditioning, further weakness, and fatigue. Poor oral
intake may lead to weight loss and nutritional compromise, adding even
more to the tendency to tire easily. Progressive weakness may adversely
affect the patient’s balance and ability to ambulate. Ultimately, the frail
older individual loses the capacity to function independently and may
require skilled assistance in a facility outside the home. Frailty also carries
with it a higher risk of medical illness and mortality (Fried and Walston
2003).
Geriatric Assessment
The presentation of acute illness in older adults may not be typical of that in
younger adults, and diagnosis in the geriatric patient therefore poses special
challenges. For example, the geriatric patient may experience a change in
mental status that suggests an acute neurological event but is instead due to
pneumonia, urinary tract infection, myocardial infarction, or an adverse
reaction to medication. The symptoms of the older patient are very often
nonspecific. The astute clinician must consider this carefully when
constructing a differential diagnosis. Something as straightforward as
functional decline might signal a more serious medical problem. The
physiological changes that occur in elderly people also may markedly alter
how patients present. Older persons may not always develop fever and
leukocytosis in response to infection, and the clinician must recognize other
clues. The decline in functional reserve in many organ systems may
predispose elderly people to harm even when a precipitating event seems
minor. An elderly person with an impaired thirst mechanism may not
replenish water losses quickly enough on a hot summer day, and
dehydration could then happen very suddenly. The challenges of
deciphering symptoms and preventing harm are heightened by the difficulty
of gathering information from elderly patients. Those who are capable of
giving a good history may be reluctant to report that anything is wrong;
they may be depressed, fearful of a new diagnosis, convinced that their
problem is nothing but “old age,” or skeptical that any doctor or medical
system can help them. In some cases, medical illness can exacerbate
psychiatric illness—making the depressed patient more disengaged or the
schizophrenic patient more agitated—and clinicians must be willing to
consider diagnoses beyond mere worsening of an ongoing psychiatric
condition and other possibilities.
The effective evaluation and treatment of the geriatric patient—from the
fully functioning community-dwelling older adult to the frail older adult in
decline—require a global approach, which includes, but reaches beyond, a
consideration of the patient’s medical problems. Reuben (2003) defined
geriatric assessment as a comprehensive patient evaluation, conducted by
an individual clinician or an interdisciplinary team, which considers the
effect of key medical, social, psychological, and environmental factors on
health and pays careful attention to function. During the medical
assessment, the clinician performs a complete history and physical
examination. He or she reviews the medication list for appropriateness and
evidence of poly-pharmacy; checks for deficits in vision, hearing,
ambulation, and balance; and screens for common geriatric problems such
as falling, incontinence, and malnutrition. Vision is tested with Snellen’s
eye chart. Hearing is screened with the whispering voice test or a handheld
audiometer. The patient’s weight and height are measured, and the body
mass index is calculated. In addition to the standard neurological
examination, the patient’s mobility and balance can be determined by a “get
up and go” test; the patient is asked to stand, walk 10 feet, turn around,
return, and be seated. The task is timed; a time greater than 20 seconds
suggests that more extensive evaluation is needed.
Cognitive assessment is performed with the Mini-Mental State
Examination (Folstein et al. 1975). The Geriatric Depression Scale
(Yesavage et al. 1983) is used to screen for depression. Fundamental day-to-
day functioning is determined by documenting ADLs—bathing, dressing,
toileting, feeding, and transferring—and IADLs—driving, shopping,
cooking, housekeeping, using the telephone, and managing finances. The
clinician also must gather other important information about function: 1)
the extent, strength, and reliability of the patient’s social support system
(most often the patient’s family); 2) the patient’s economic resources; and
3) the safety of the patient’s home and its proximity to medical care and
other essential services. The patient’s spiritual preferences and needs are
also assessed. After the assessment is completed, recommendations are
developed and a care plan is implemented.
Although the results across clinical trials have not been consistent, the
effectiveness of comprehensive geriatric assessment and management has
been validated in several studies. Increased diagnostic accuracy has been
noted. Patients have shown significant improvements in functional status.
Affect and cognition have improved. The use of health care services, as
measured by nursing home days, hospital services, and medical costs, has
been reduced. The use of medications has improved, with fewer drugs being
prescribed (Hanlon et al. 2001; Reuben 2003; Spinewine et al. 2007; Stuck
et al. 1993). In-home geriatric assessment of older patients may postpone
the onset of disability, as well as reduce the number of patients requiring
permanent placement in nursing homes (Stuck et al. 1995). A multi-
institutional randomized controlled trial of geriatric evaluation and
management units in the Veterans Affairs Health Care System showed a
positive effect on functional status and quality of life for inpatients and on
mental health and quality of life for outpatients, with overall costs
equivalent to those for usual care, but had no effect on morbidity or
mortality (Cohen et al. 2002). As suggested by the evidence, comprehensive
geriatric assessment and management may serve as a useful tool for the
diagnosis and care of older patients, and the geriatrician has a valuable and
essential role in the evaluation and treatment of this population.
Conclusion
With longevity, all older adults face the prospect of progressive
physiological decline and increased vulnerability. This may result in
significant sensory deficits, increased susceptibility to illness, and less
robust intellectual and physical abilities. Those entering their seventh,
eighth, and ninth decades of life must confront many challenges to their
health and independence, but human beings are eminently adaptable and
resilient (Dubos 1965). Old age should not automatically lead to isolation
and infirmity; instead, it can be a time of happiness, creativity, and
fulfillment (Cohen et al. 2006). For many, the challenges of aging can be
met—insightfully and optimistically—by drawing on years of personal
experience and accumulated wisdom (Brooks 2014). Obtaining good
interdisciplinary geriatric care—care that enlists older individuals (and their
families) as equal partners and systematically addresses their medical,
social, psychological, and functional needs—may also be beneficial in
aging well and in ensuring that quality of life is as high as possible.
Key Points
• People older than age 65 years constitute one of the fastest-growing
segments of the U.S. population, and the average life span has
lengthened significantly.
• The hallmarks of physiological change in older adults are twofold:
impaired homeostasis (also called homeostenosis) and increased
vulnerability because of decreased reserve capacity.
• The time needed for older adults to learn new information increases, and
they may have more difficulty accessing data from long-term memory.
• Older persons develop significant age-related changes in vision—
including decreases in accommodation, ability to adapt to light, color
discrimination, and visual acuity—and in hearing, with loss of hearing
ability in both high and low frequencies.
• Aging results in arterial stiffening and subsequent increased systolic
blood pressure and aortic pulse pressure, with resultant susceptibility to
left ventricular hypertrophy, cardiac ischemia, left ventricular failure,
cerebrovascular ischemia, and renal dysfunction.
• The immune response of the older individual is less vigorous, with
decreased cell-mediated response, impaired humoral response, and
increased susceptibility to infection.
• The decline in renal clearance and the increase in the elimination half-
life in elderly people may require adjustment of the dosing interval of
medications; because older adults are more sensitive to medications,
drugs must often be given in lower doses.
• Some chronic diseases are more prevalent in older adults, predominantly
occurring as a result of usual aging; these include obesity, hypertension,
hypercholesterolemia, type 2 diabetes mellitus, osteoarthritis,
cerebrovascular disease, cardiovascular disease, and cancer.
• Several common syndromes—known generally as geriatric syndromes—
are found more frequently in older patients, including dementia, falls,
urinary incontinence, polypharmacy, and frailty.
• The effectiveness of comprehensive geriatric assessment and
management has been validated in several large randomized studies, and
the geriatrician has a valuable and essential role in the evaluation and
treatment of this population.
References
Administration on Aging: Department of Health and Human Services,
Aging Statistics: Projected Future Growth of the Older Population,
2012. Available at:
http://www.aoa.gov/AoARoot/Aging_Statistics/future_growth/future_g
rowth.aspx#age. Accessed April 8, 2014.
Alexander NB: Falls and gait disturbances, in Geriatrics Review Syllabus:
A Core Curriculum in Geriatric Medicine. Edited by Cobbs E, Duthie
EH, Murphy JB. Dubuque, IA, Kendall/Hunt, 1999, pp 145–149
Alzheimer’s Association: Proposed Revisions to Diagnostic Criteria for
Alzheimer’s Disease, July 2010. Available at:
http://www.alz.org/documents_custom/Alz_Diag_Criteria_FAQ.pdf.
Accessed April 8, 2014.
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 60(4):
616–631, 2012 22376048
Armbrecht HJ: The biology of aging. J Lab Clin Med 138(4):220–225,
2001 11574815
Aw D, Silva AB, Palmer DB: Immunosenescence: emerging challenges for
an ageing population. Immunology 120(4):435–446, 2007 17313487
Berry SD, Kiel DP: Falls, in Geriatrics Review Syllabus, 8th Edition.
Edited by Durso SC, Sullivan GM. New York, American Geriatrics
Society, 2013
Bhasin S, Cunningham GR, Hayes FJ, et al: Testosterone therapy in men
with androgen deficiency syndromes: an Endocrine Society clinical
practice guideline. J Clin Endocrinol Metab 95:2536–2559, 2010
Birks J, Cochrane Dementia and Cognitive Improvement Group:
Cholinesterase inhibitors for Alzheimer’s disease. The Cochrane
Library 2006. Available at:
http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD005593/abstra
ct;jsessionid=4FA2F34374EC93993955F67AD324F922.f03t02.
Accessed April 8, 2014.
Bischoff-Ferrari HA, Dawson-Hughes B, Willett WC, et al: Effect of
vitamin D on falls: a meta-analysis. JAMA 291(16):1999–2006, 2004
15113819
Bolland MJ, Avenelli A, Baron JA, et al: Effect of calcium supplements on
risk of myocardial infarction and cardiovascular events: meta-analysis.
BMJ 341:c3691, 2010
Brooks D: Why elders smile. New York Times, December 4, 2014.
Available at: http://www.nytimes.com/2014/12/05/opinion/david-
brooks-why-elders-smile.html?_r=0. Accessed December 11, 2014.
Centers for Disease Control and Prevention: Diabetes Public Health
Resource: Incidence of Diagnosed Diabetes Per 1,000 Population Aged
18–79 Years, By Age, 1980–2011 (updated October 2, 2014). 2014a.
Available at http://www.cdc.gov/diabetes/statistics/incidence/fig3.htm.
Accessed December 11, 2014.
Centers for Disease Control and Prevention: Home and Recreational Safety.
Falls Among Older Adults: An Overview (updated September 22,
2014). 2014b. Available at:
http://www.cdc.gov/HomeandRecreationalSafety/Falls/adultfalls.html.
Accessed December 11, 2014.
Chatta GS, Lipschitz DA: Aging of the hematopoietic system, in Principles
of Geriatric Medicine and Gerontology, 5th Edition. Edited by Hazzard
WR, Blass JP, Halter JB, et al. New York, McGraw-Hill, 2003, pp 763–
770
Clinical Trials.gov: Clinical Trial of Solanezumab for Older Individuals
Who May be at Risk for Memory Loss (A4) (updated December 4,
2014). 2014a. Available at:
http://clinicaltrials.gov/ct2/show/NCT02008357. Accessed December
11, 2014.
Clinical Trials.gov: The Study of Nasal Insulin in the Fight Against
Forgetfulness (SNIFF) (updated September 15, 2014). 2014b.
Available at: http://clinicaltrials.gov/ct2/show/NCT01767909.
Accessed December 11, 2014.
Cohen GD, Perlstein S, Chapline J, et al: The impact of professionally
conducted cultural programs on the physical health, mental health, and
social functioning of older adults. Gerontologist 46(6):726–734, 2006
17169928
Cohen HJ: In search of the underlying mechanisms of frailty. J Gerontol A
Biol Sci Med Sci 55(12):M706–M708, 2000 11129391
Cohen HJ, Feussner JR, Weinberger M, et al: A controlled trial of inpatient
and outpatient geriatric evaluation and management. N Engl J Med
346(12):905–912, 2002 11907291
Cornoni-Huntley J, Brock DB, Ostfield A, et al (eds): Established
Populations for the Epidemiologic Study of the Elderly: Resource Data
Book. Bethesda, MD, National Institutes of Health, 1986
Craft S, Cholerton B, Reger M: Aging and cognition: what is normal? In
Principles of Geriatric Medicine and Gerontology, 5th Edition. Edited
by Hazzard WR, Blass JP, Halter JB, et al. New York, McGraw-Hill,
2003, pp 1355–1372
Defilippi JL, Crismon ML: Antipsychotic agents in patients with dementia.
Pharmacotherapy 20(1):23–33, 2000 10641973
Doody RS, Raman R, Farlow M, et al: A phase 3 trial of semagacestat for
treatment of Alzheimer’s disease. N Engl J Med 369(4):341–350, 2013
23883379
Doody RS, Thomas RG, Farlow M, et al: Phase 3 trials of solanezumab for
mild-to-moderate Alzheimer’s disease. N Engl J Med 370:311–321,
2014
DuBeau CW: Urinary incontinence, in Geriatrics Review Syllabus: A Core
Curriculum in Geriatric Medicine, 6th Edition. Edited by Pompei P,
Murphy JB. New York, American Geriatrics Society, 2006, pp 184–195
Dubos R: Man Adapting. New Haven, CT, Yale University Press, 1965
Emanuel E: Reinventing American Health Care: How the Affordable Care
Act Will Improve Our Terribly Complex, Blatantly Unjust,
Outrageously Expensive, Grossly Inefficient, Error Prone System. New
York, Public Affairs, 2014
Farrimond LE, Roberts E, McShane R: Memantine and cholinesterase
inhibitor combination therapy for Alzheimer’s disease: a systematic
review. BMJ Open 2:e000917, 2012. Available at:
http://bmjopen.bmj.com/content/2/3/e000917.full. Accessed April 8,
2014.
Firth M, Prather CM: Gastrointestinal motility problems in the elderly
patient. Gastroenterology 122:1688–1700, 2002 12016432
Folstein MF, Folstein SE, McHugh PR: “Mini-Mental State.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Fried LP: Epidemiology of aging. Epidemiol Rev 22(1):95–106, 2000
10939013
Fried LP, Walston J: Frailty and failure to thrive, in Principles of Geriatric
Medicine and Gerontology, 5th Edition. Edited by Hazzard WR, Blass
JP, Halter JB, et al. New York, McGraw-Hill, 2003, pp 1487–1502
Gillespie LD, Gillespie WJ, Robertson MC, et al: Interventions for
preventing falls in elderly people. Cochrane Database Syst Rev
(4):CD000340, 2003 14583918
Gómez-Tortosa E, Ingraham AO, Irizarry MC, et al: Dementia with Lewy
bodies. J Am Geriatr Soc 46(11):1449–1458, 1998 9809770
Grimes DA, Lobo RA: Perspectives on the Women’s Health Initiative trial
of hormone replacement therapy. Obstet Gynecol 100(6):1344–1353,
2002 12468183
Gruenewald DA, Matsumoto AM: Aging of the endocrine system, in
Principles of Geriatric Medicine and Gerontology, 5th Edition. Edited
by Hazzard WR, Blass JP, Halter JB, et al. New York, McGraw-Hill,
2003, pp 819–836
Hall KE, Wiley JW: Age-associated changes in gastrointestinal function, in
Principles of Geriatric Medicine and Gerontology, 5th Edition. Edited
by Hazzard WR, Blass JP, Halter JB, et al. New York, McGraw-Hill,
2003, pp 593–600
Hall WJ: Update in geriatrics. Ann Intern Med 127(7):557–564, 1997
9313023
Halter JB: Diabetes mellitus, in Principles of Geriatric Medicine and
Gerontology, 5th Edition. Edited by Hazzard WR, Blass JP, Halter JB,
et al. New York, McGraw-Hill, 2003, pp 855–874
Hamerman D: Toward an understanding of frailty. Ann Intern Med
130(11):945–950, 1999 10375351
Hanlon JT, Schmader KE, Ruby CM, et al: Suboptimal prescribing in older
inpatients and outpatients. J Am Geriatr Soc 49(2):200–209, 2001
11207875
Harvey PT: Common eye diseases of elderly people: identifying and
treating causes of vision loss. Gerontology 49(1):1–11, 2003 12457044
Ho JC, Chan KN, Hu WH, et al: The effect of aging on nasal mucociliary
clearance, beat frequency, and ultrastructure of respiratory cilia. Am J
Respir Crit Care Med 163(4):983–988, 2001 11282777
Huang Q, Tang J: Age-related hearing loss or presbycusis. Eur Arch
Otorhinolaryngol 267(8):1179–1191, 2010 20464410
Hubschman JP, Reddy S, Schwartz SD: Age-related macular degeneration:
current treatments. Clin Ophthalmol 3:155–166, 2009 19668560
Hunink MG, Goldman L, Tosteson AN, et al: The recent decline in
mortality from coronary heart disease, 1980–1990: the effect of secular
trends in risk factors and treatment. JAMA 277(7):535–542, 1997
9032159
Janssens JP: Aging of the respiratory system: impact on pulmonary function
tests and adaptation to exertion. Clin Chest Med 26(3):469–484, vi–vii,
2005 16140139
Karlin BE, Visnic S, McGee JS, et al: Results from the multisite
implementation of STAR-VA: a multicomponent psychosocial
intervention for managing challenging dementia-related behaviors of
veterans. Psychol Serv 11(2):200–208, 2014 23937081
King MB, Tinetti ME: Falls in community-dwelling older persons. J Am
Geriatr Soc 43(10):1146–1154, 1995 7560708
Klafki HW, Staufenbiel S, Kornhuber J, et al: Therapeutic approaches to
Alzheimer’s disease. Brain 139:2840–2855, 2006 17018549
Lakatta EG: Cardiovascular aging research: the next horizons. J Am Geriatr
Soc 47(5):613–625, 1999 10323658
Loeser RF, Delbono O: Aging and the musculoskeletal system, in Principles
of Geriatric Medicine and Gerontology, 5th Edition. Edited by Hazzard
WR, Blass JP, Halter JB, et al. New York, McGraw-Hill, 2003, pp 905–
918
Logsdon RG, McCurry SM, Teri L: STAR-Care-givers: A community-
based approach for teaching family caregivers to use behavioral
strategies to reduce affective disturbances in persons with dementia.
Alzheimer’s Care Today 6(2):146–153, 2005
Majumdar AP, Jaszewski R, Dubick MA: Effect of aging on the
gastrointestinal tract and the pancreas. Proc Soc Exp Biol Med
215(2):134–144, 1997 9160041
Marin DB, Sewell MC, Schlechter A: Alzheimer’s disease: accurate and
early diagnosis in the primary care setting. Geriatrics 57(2):36–40, quiz
43, 2002 11851205
Matsumoto AM: Andropause: clinical implications of the decline in serum
testosterone levels with aging in men. J Gerontol A Biol Sci Med Sci
57(2):M76–M99, 2002 11818427
McKeown T: The role of medicine: dream, mirage, or nemesis. Princeton,
NJ, Princeton University Press, 1979
McNabney MK, Fedarko NS: Biology, in Geriatrics Review Syllabus, Book
I. Edited by Durse SC, Sullivan GM. New York, American Geriatrics
Society, 2013
McKhann GM, Knopman DS, Chertkow H, et al: The diagnosis of
dementia 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
7(3):263–269, 2011 21514250
Miller RA: The biology of aging and longevity, in Principles of Geriatric
Medicine and Gerontology. Edited by Hazzard WR, Blass JP, Ettinger
WH, et al. New York, McGraw-Hill, 1999, pp 3–19
Morley JE, Haren MT, Rolland Y, et al: Frailty. Med Clin North Am
90(5):837–847, 2006 16962845
Morris JC: Is Alzheimer’s disease inevitable with age?: Lessons from
clinicopathologic studies of healthy aging and very mild Alzheimer’s
disease. J Clin Invest 104(9):1171–1173, 1999 10545515
Nelson HD, Humphrey LL, Nygren P, et al: Postmenopausal hormone
replacement therapy: scientific review. JAMA 288(7): 872–881, 2002
12186605
Nyenhuis DL, Gorelick PB: Vascular dementia: a contemporary review of
epidemiology, diagnosis, prevention, and treatment. J Am Geriatr Soc
46(11):1437–1448, 1998 9809769
O’Connor SD, Prusiner S, Dychtwald K: The age of Alzheimer’s. New
York Times. October 27, 2010. Available at:
http://www.nytimes.com/2010/10/28/opinion/28oconnor.html.
Accessed April 8, 2014.
O’Rourke MF, Hashimoto J: Mechanical factors in arterial aging: a clinical
perspective. J Am Coll Cardiol 50:1–13, 2006 17601538
Oskvig RM: Special problems in the elderly. Chest 115(5 suppl):158S–
164S, 1999 10331350
Perls T: The different paths to age one hundred. Ann N Y Acad Sci
1055:13–25, 2005 16387714
Perry HM 3rd: The endocrinology of aging. Clin Chem 45(8 Pt 2):1369–
1376, 1999 10430820
Prestwood K, Duque G: Osteoporosis, in Principles of Geriatric Medicine
and Gerontology, 5th Edition. Edited by Hazzard WR, Blass JP, Halter
JB, et al. New York, McGraw-Hill, 2003, pp 973–985
Raina P, Santaguida P, Ismaila A, et al: Effectiveness of cholinesterase
inhibitors and memantine for treating dementia: evidence review for a
clinical practice guideline. Ann Intern Med 148(5):379–397, 2008
18316756
Reuben DB: Principles of geriatric assessment, in Principles of Geriatric
Medicine and Gerontology, 5th Edition. Edited by Hazzard WR, Blass
JP, Halter JB, et al. New York, McGraw-Hill, 2003, pp 99–110
Rizvi AA: Management of diabetes in older adults. Am J Med Sci
333(1):35–47, 2007 17220692
Rubenstein LZ, Josephson KR: Falls and their prevention in elderly people:
what does the evidence show? Med Clin North Am 90(5):807–824,
2006 16962843
Rubenstein LZ, Josephson KR, Robbins AS: Falls in the nursing home. Ann
Intern Med 121(6):442–451, 1994 8053619
Sadovsky R, Dhindsa S, Margo K: Testosterone deficiency: which patients
should you screen and treat? J Fam Pract 56 (5 suppl Testosterone):S1–
S20, 2007 17475159
Schwartz JB: Clinical pharmacology, in Principles of Geriatric Medicine
and Gerontology, 4th Edition. Edited by Hazzard WR, Blass JP,
Ettinger WH, et al. New York, McGraw-Hill, 1999, pp 303–332
Schwartz RS, Kohrt WM: Exercise in elderly people: physiological and
functional effects, in Principles of Geriatric Medicine and Gerontology,
5th Edition. Edited by Hazzard WR, Blass JP, Halter JB, et al. New
York, McGraw-Hill, 2003, pp 931–946
Seals DR, Esler MD: Human ageing and the sympathoadrenal system. J
Physiol 528 (Pt 3):407–417, 2000 11060120
Semla TP, Rochon PA: Pharmacotherapy, in Geriatrics Review Syllabus: A
Core Curriculum in Geriatric Medicine, 6th Edition. Edited by Pompei
P, Murphy JB. New York, American Geriatrics Society, 2006, pp 72–80
Shock NW: Normal Human Aging: The Baltimore Longitudinal Study of
Aging. Washington, DC, National Institute on Aging, U.S. Government
Printing Office, 1984
Sharma G, Goodwin J: Effect of aging on respiratory system physiology
and immunology. Clin Interv Aging 1(3):253–260, 2006 18046878
Smith CH, Briony B, Daureeawoo Y, et al: Effect of aging on stimulated
salivary flow in adults. J Am Geriatr Soc 61:805–808, 2013 23617733
Spinewine A, Schmader KE, Barber N, et al: Appropriate prescribing in
elderly people: how well can it be measured and optimised? Lancet
370:173–184, 2007 17630041
Stewart RB: Drug use in the elderly, in Therapeutics in the Elderly, 3rd
Edition. Edited by Delafuente JC, Stewart RB. Cincinnati, OH, Harvey
Whitney, 2001, pp 235–256
Stuck AE, Siu AL, Wieland GD, et al: Comprehensive geriatric assessment:
a meta-analysis of controlled trials. Lancet 342(8878): 1032–1036,
1993 8105269
Stuck AE, Aronow HU, Steiner A, et al: A trial of annual in-home
comprehensive geriatric assessments for elderly people living in the
community. N Engl J Med 333(18):1184–1189, 1995 7565974
Taffet GE: Age-related physiologic changes, in Geriatrics Review Syllabus:
A Core Curriculum in Geriatric Medicine, 5th Edition. Edited by
Cobbs E, Duthie EH, Murphy JB. Dubuque, IA, Kendall/Hunt, 1999,
pp 10–23
Tannenbaum C, Perrin L, DuBeau CE, et al: Diagnosis and management of
urinary incontinence in the older patient. Arch Phys Med Rehabil
82(1):134–138, 2001 11239300
Teri L, McCurry SM, Logsdon R, et al: Training community consultants to
help family members improve dementia care: a randomized controlled
trial. Gerontologist 45(6):802–811, 2005 16326662
Tune LE: Risperidone for the treatment of behavioral and psychological
symptoms of dementia. J Clin Psychiatry 62 (suppl 21): 29–32, 2001
11584986
Ungvari Z, Kaley G, de Cabo R, et al: Mechanisms of vascular aging: new
perspectives. J Gerontol A Biol Sci Med Sci 65(10): 1028–1041, 2010
20576649
Vaillant GE, Mukamal K: Successful aging. Am J Psychiatry 158(6):839–
847, 2001 11384887
Veldhuis JD: Changes in pituitary function with ageing and implications for
patient care. Nat Rev Endocrinol 9(4):205–215, 2013 23438832
Vigen R, O’Donnell CI, Barón AE, et al: Association of testosterone
therapy with mortality, myocardial infarction, and stroke in men with
low testosterone levels. JAMA 310(17):1829–1836, 2013 24193080
Weinstein JR, Anderson S: The aging kidney: physiological changes. Adv
Chronic Kidney Dis 17(4):302–307, 2010 20610357
Werner CA: The Older Population: 2010. 2010 Census Briefs (C2010BR-
09). Washington, DC, U.S. Census Bureau, November 2011. Available
at: http://www.census.gov/prod/cen2010/briefs/c2010br-09.pdf.
Accessed December 11, 2014.
Weymouth F: Effect of age on visual acuity, in Vision of the Aging Patient.
Edited by Hirsh MJ, Wick RE. Philadelphia, PA, Chilton, 1960, pp 37–
62
Whitaker M, Guo J, Kehoe T, et al: Bisphosphonates for osteoporosis—
where do we go from here? N Engl J Med 366(22):2048–2051, 2012
22571168
Yesavage JA, Brink TL, Rose TL, et al: Development and validation of a
geriatric depression screening scale: a preliminary report. J Psychiatr
Res 17:37–49, 1983 7183759
Suggested Readings
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 60:616–
631, 2012
Aw D, Silva AB, Palmer DB: Immunosenescence: emerging challenges for
an ageing population. Immunology 120:435–446, 2007
Bischoff-Ferrari HA, Dawson-Hughes B, Willett WC, et al: Effect of
vitamin D on falls: a meta-analysis. JAMA 291:1999–2006, 2004
Cohen HJ, Feussner JR, Weinberger M, et al: A controlled trial of inpatient
and outpatient geriatric evaluation and management. N Engl J Med
346:905–912, 2002
Fried LP: Epidemiology of aging. Epidemiol Rev 22:95–106, 2000
Patterson SM, Hughes C, Kerse N, et al: Interventions to improve the
appropriate use of polypharmacy for older people (review). Cochrane
Database Syst Rev (5): CD008165, 2012
Tinetti M, Kumar C: The patient who falls: “it’s always a trade-off.” JAMA
303(3): 258–266, 2010
Ungvari Z, Kaley G, de Cabo R, et al. Mechanisms of vascular aging: new
perspectives. J Gerontol A Biol Sci Med Sci 65A(10):1028–1041, 2010
Vaillant GE, Mukamal K: Successful aging. Am J Psychiatry 158:839–847,
2001
Whitaker M, Guo J, Kehoe T, et al. Bisphosphonates for osteoporosis:
where do we go from here? N Engl J Med 366:2048–2051, 2012
CHAPTER 3
Single-Nucleotide Polymorphisms
Until relatively recently, the majority of known disease-causing mutations
involved single DNA nucleotide changes (single-nucleotide polymorphisms
[SNPs]) in coding DNA that resulted in an altered amino acid sequence of
the translated protein. However, there is now greater recognition of the
contribution of SNPs in ncDNA sequences to disease risk and expression.
Koboldt and Miller (2007), for example, mapped more than 9 million SNPs
to gene sequences and ncDNA sequences of functional significance,
including transcription factor binding sites, exon splicing enhancers or
silencers, transcribed ncRNA, regulatory-potential sequences, and ncRNA
binding sites. They found that SNPs in coding DNA that altered protein
structure had significantly decreased allele frequencies and increased
population specificity (providing evidence of a functional effect, subject to
natural selection). Furthermore, they observed the same pattern among
SNPs in ncDNA that mapped to transcription factor binding sites, ncRNA
binding elements, regulatory-potential sequences, and splice sites, with
substitutions in ncRNA binding or splice sites being more likely to show
evidence of selection than coding SNPs. Subsequent discoveries have led
ncRNA to be implicated in several diseases that affect the brain, such as
schizophrenia, in which the strongest genetic findings to date are an
association of disease with genetic variation located within the gene for
microRNA 137 (Schizophrenia Psychiatric GWAS Consortium 2011).
Copy-Number Variation
Efforts to screen the human genome have shown that large segments of
DNA may also be deleted or duplicated (Inoue and Lupski 2002; Lupski
2007), resulting in changes in the number of genes within these regions and
disrupting gene structure at deletion or duplication breakpoints (Neumann
et al. 2006). Diseases caused by genomic rearrangements that result in an
altered number of gene copies (copy-number variation [CNV]) are often
referred to as genomic disorders. Efforts to map genomic variation in the
complete human genome have identified CNVs in 1,400 regions that
overlap with 14.5% of genes associated with human disease (Redon et al.
2006). CNV is known to be associated with a number of
neurodevelopmental and neurodegenerative disorders, including sporadic
and familial cases (Redon et al. 2006; Sebat et al. 2007). Familial early-
onset cases of AD and Parkinson’s disease (PD) due to CNV, for example,
are described later in this chapter (see “Genetics of Mental Disorders of
Aging and Later Life”). Too often, however, reports have been limited to
the discovery of regions containing CNVs, without identifying the specific
DNA sequence resulting in illness (the latter of which remains an important
area of study for the field).
De Novo Mutations
Although genetic risk is thought to be passed down through families, it is
also true that new (i.e., de novo) mutations or structural rearrangements
may occur in a parental gamete (sperm or egg) and thus first manifest in an
affected offspring. Rates of de novo mutations are increased, for example,
by increasing paternal age, whereas increasing maternal age is a risk factor
for de novo structural alterations such as trisomy 21, which results in Down
syndrome.
Before genetic testing is ordered, the patient, family, and health care
team must discuss a number of issues (Goldman 2012, p. 504):
Alzheimer’s Disease
AD—or, in DSM-5, neurocognitive disorder due to Alzheimer’s disease—is
the predominant cause of dementia in the United States, with over 5 million
people affected. The etiology of AD remains unclear, although significant
strides have been made using gene mapping efforts. Success has been most
notable for Mendelian familial early-onset AD (EOAD), which comprises a
minority (less than 5%) of all AD cases (Goldman et al. 2011). However,
recent success in identifying genetic variants in AD has led to a deeper
understanding of the biology of the disorder, and has potential therapeutic
implications for both EOAD and late-onset AD (LOAD).
Frontotemporal Dementia
Frontotemporal dementia (FTD)—or, in DSM-5, neurocognitive disorder
due to frontotemporal lobar degeneration—is a clinically and pathologically
heterogeneous group of non-Alzheimer dementias whose underlying
pathology includes progressive degeneration of the prefrontal and anterior
temporal lobes (Neary et al. 2005). There are three main clinical syndromes
of FTD, characterized by leading features at presentation (Warren et al.
2013). About half of FTD cases present with behavioral change, referred to
as behavioral variant FTD. The remainder present with language decline
(primary progressive aphasia) characterized either by impaired speech
production (progressive nonfluent aphasia) or impaired word
comprehension and semantic memory (semantic dementia). It should be
noted, however, that a fair amount of overlap exists across these syndromes
(Warren et al. 2013). In contrast to AD—the most common dementia of
later life—FTD often presents in later middle age, with memory being
initially well maintained. Behavioral or personality changes that occur in
individuals with FTD are often mistaken for a primary psychiatric disorder,
particularly if the changes are accompanied by psychotic features. Clues
that such features are harbingers of FTD include absence of prior
psychiatric history, as well as the emergence of more specific symptoms
such as changes in eating behavior or social faux pas (Warren et al. 2013).
Nearly all FTD cases result from one of two major types of
neurodegenerative lesions: 1) tau-positive inclusions and 2) ubiquitin-
positive inclusions, shown to contain transactive response (TAR) DNA-
binding protein 43 (TDP-43) (Neumann et al. 2006). The neuropathological
findings of FTD overlap with those of several other neurodegenerative
disorders; for example, tau-positive inclusions are also seen in progressive
supranuclear palsy and corticobasal degeneration, and TDP-43–positive
inclusions are often seen with motor neuron disease.
FTD has been the subject of extensive genetic investigation. A positive
family history is present in up to 50% of cases (Bird et al. 2003; Chow et al.
1999). Mutations in one of several genes can result in FTD, which typically
demonstrates an autosomal dominant pattern of transmission. Major causes
of FTD include mutations in the following genes: MAPT, PGRN or GRN,
and C9orf72 (Wang et al. 2013). A minority of cases are caused by
mutations in the following genes: TDP-43, VCP, CHMP2B, and FUS. In
addition, involvement of the APOE gene locus has been reported (Wang et
al. 2013). A significant discovery identified mutation in C9orf72 (DeJesus-
Hernandez et al. 2011) as perhaps the most common genetic abnormality in
both familial behavioral variant FTD (11.7% of cases) and motor neuron
disease (23.5%). This new mutation is an expansion of a noncoding
GGGGCC hexanucleotide repeat located in chromosome 9p (Dobson-Stone
et al. 2012).
Testing for frontotemporal dementia–associated genes. Definitive
diagnosis of FTD (and AD, for that matter) can be made only at autopsy,
but genetics may be used to increase diagnostic and prognostic accuracy
(Farlow and Foroud 2013). MAPT, GRN, and C9orf72 may be easily
sequenced for mutations that lead to FTD and together account for a
majority of familial cases. Families concerned about the possibility of FTD
developing in future generations frequently ask their physician to order
genetic testing (Chow and Alobaidy 2013). As always, a referral to a
genetic counselor should precede the collection of samples for testing
(Chow and Alobaidy 2013).
The first step in determining the order for genetic testing for FTD is to
take a detailed family history (Goldman 2012). As with AD, testing for
autosomal dominant genes is less likely to yield a mutational finding
without a known or suspected family history of FTD. However, PGRN and
C9orf72 mutations have been found in sporadic cases (Goldman 2012).
Genetic testing for FTD is also greatly facilitated by having a pathological
diagnosis on an affected family member, because autopsy results can guide
genetic testing by eliminating the need to test certain genes.
FTD genotyping may soon play a role in treatment selection for FTD.
Several preliminary reports have proposed the use of antisense
oligonucleotides (ASOs; short synthetic single DNA strands designed to
bind their complementary sequence) to reduce levels of RNA containing the
pathological hexanucleotide repeat in chromosome 9p (Lagier-Tourenne et
al. 2013). ASO therapies may treat gain-of-function disorders by silencing
the causative gene, either by acting on both alleles for nonessential genes or
by selectively acting on the mutant allele in the case of essential genes
(Fernandes et al. 2013). For certain loss-of-function diseases, there is also
the possibility of using ASOs for splice modulation, which can in certain
cases restore the gene function or otherwise compensate for its loss
(Fernandes et al. 2013).
Parkinson’s Disease
PD is a progressive neurodegenerative disorder that affects 1%–2% of the
population ages 65 years and older (de Rijk et al. 1997). It is characterized
by motor symptoms that include akinesia, rest tremor, rigidity, and
disturbance of postural reflexes. Many cases of PD are thought to arise
sporadically; as a result, the risk for first-degree family members of
individuals with PD is increased only two- to threefold, and only in families
with an early age at onset (Gasser 2005). Mutations in multiple genes have
been identified that appear to cause PD via a Mendelian mode of
inheritance. As with AD, however, these mutations account for a small
number of affected families and are primarily associated with earlier
lifetime onset (for reviews, see Gasser 2005; Hardy et al. 2006).
Autosomal dominant PD is robustly associated with mutations in the
following genes: SNCA, LRRK2, EIF4G1, and VPS35. Recessive PD is
associated with mutations in PARK2, PINK1, and DJ1/PARK7 (Puschmann
2013). Changes in a long list of additional genes have also been suggested,
including genes for hereditary ataxias, frontotemporal dementia, dopa-
responsive dystonia, and other conditions. Point mutations and
multiplications in SNCA, the gene encoding α-synuclein protein, cause
cognitive or psychiatric symptoms and parkinsonism, with widespread α-
synuclein pathology in the central and peripheral nervous system
(Puschmann 2013). LRRK2 mutations may lead to a clinical phenotype
closely resembling idiopathic PD, with a variety of neuropathology.
Mutations in PARK2, PINK1, and DJ1/PARK7, on the other hand, may
cause early-onset parkinsonism with limited risk for cognitive decline.
Carriers of mutations in the other genes may develop parkinsonism with or
without additional symptoms, but rarely a disease resembling PD. Although
some mutations occur with high frequency in specific populations, all are
very rare. Therefore, the genetic etiology of the majority of cases of
sporadic or hereditary PD remains unknown on a more global scale
(Puschmann 2013).
In a two-stage meta-analysis, the International Parkinson’s Disease
Genomics Consortium and Wellcome Trust Case Control Consortium 2
(2011) identified several new potential genetic loci for PD. The researchers
identified five additional PD risk loci—PARK16/1q32, FGF20/8p22,
STX1B/16p11, STBD1/4q21, and GPNMB/ 7p15—the first two of which
had been suggested by previous association studies. Using data from
postmortem brain samples assayed for gene expression and methylation, the
investigators were able to identify methylation and expression changes
associated with PD risk variants within PARK16/1q32, GPNMB/7p15, and
STX1B/16p11 loci, suggesting potential molecular mechanisms and
candidate genes at these locations.
Testing for Parkinson’s disease–associated genes. Currently, genetic
testing is available for a number of PD-related genes, including SNCA,
LRRK2 (PARK8), PARK2, PARK7, and PINK1 (Athena Diagnostics 2014).
Routine testing is not recommended at this time, however, due to lack of
disease-modifying therapies and still evolving research on the role and
utility of testing in clinical decision making. If an individual or family is
interested in pursuing testing, it is recommended that this is guided by a
genetic counselor with experience in working with patients with familial
history of PD.
Rare Mutations
Efforts to identify rare variants with high penetrance leading to Mendelian
forms of disease have proved successful for neurodegenerative disorders
but unfortunately have not yielded definitive results for schizophrenia,
bipolar disorders, or major depressive disorder. In recent years, efforts have
shifted to the detection of rare de novo variants, with most studies to date
having focused on schizophrenia. This shift has been motivated in part by
the recognition that many individuals with schizophrenia do not have
family histories of the disorder, as well as the fact that schizophrenia has
persisted in the population despite conferring lower reproductive success.
Both of these observations may be explained by an excess of de novo
mutations (Xu et al. 2011). Efforts in this area have met with some
preliminary successes, suggesting there may be enrichment of de novo
mutations that disrupt genes involved in brain development and/or synaptic
function (Gulsuner et al. 2013; Kenny et al. 2014; Xu et al. 2012).
However, these studies have been limited by the fact that most identified
mutations are both individually rare and not fully penetrant, meaning that
they occur to some extent in unaffected individuals as well. The result is
that substantially larger numbers of subjects will need to be genotyped for
many of these rare variants before clear evidence of causality for any of
these genes can be established. It is expected that such studies will be
forthcoming.
Structural Variants
A number of studies have established that individuals with schizophrenia
have a greater frequency of genetic structural variation than do individuals
without schizophrenia; data for bipolar disorders are less striking but also
consistent with a greater frequency of structural variation (Malhotra and
Sebat 2012; Sullivan et al. 2012). Similar findings have not emerged for
major depressive disorder. Not only is the overall burden of structural
variants increased in schizophrenia, but alterations at a number of specific
genomic loci have been replicated across studies (Malhotra and Sebat
2012). Such loci have included confirmation of a previously known
association of increased risk for schizophrenia in individuals with a large
deletion at chromosome (chr) 22q11.2, which is associated with the
developmental disorder velocardiofacial (i.e., DiGeorge) syndrome, in
which about 25% of individuals develop schizophrenia. Other loci with
replicated structural variations associated with increased risk of
schizophrenia, and in some cases with bipolar disorders and recurrent
depression, include deletions at chr1q21.1, chr3q29, and chr15q13.3.
However, the implications of these and other similar findings for a more
specific biological understanding of schizophrenia and bipolar disorders are
not entirely clear, because these deletions are also associated with increased
risk for intellectual disability, developmental disorders, and autism
spectrum disorders. In addition, these loci contain many genes within the
deleted regions, and therefore determining the actual genetic changes that
lead to neuropsychiatric impairment remains a challenge.
With regard to the latter issue, there have been several structural variants
that seem to implicate a single gene or a limited set of genes. Among the
most intensively studied of these has been a reciprocal translocation
between chr1 and chr11 that disrupts DISC1, as well as ncRNAs DISC2 and
DISC1FP1 (Thomson et al. 2013). The translocation between chr1 and
chr11 has been found in a single large Scottish family, in which carriers of
the translocation have increased rates of schizophrenia, recurrent major
depressive disorder, and bipolar disorders (Blackwood et al. 2001).
However, the translocation is not fully penetrant, because carriers are
unaffected. This finding has spurred extensive examination of the genetics
of DISC1 in other individuals and families. These studies, which have
encompassed assessments of structural variation, rare mutations, and
common variants, have provided supportive but not unequivocal evidence
for an association of DISC1 with schizophrenia, depressive disorders, and
autism spectrum disorders (Thomson et al. 2013). Nevertheless, as one of
the best single-gene candidates for causation of mental illness, DISC1 has
been the subject of increasingly expansive investigations of its functionality
(Thomson et al. 2013).
Deletions and duplications affecting the gene NRXN1 have likewise been
described in schizophrenia but are also known to occur in autism spectrum
disorder, intellectual disability, and seizure disorder (Doherty et al. 2012).
Similarly, a duplication affecting the vasoactive intestinal peptide receptor 2
gene (VIPR2)—which, despite its name, is expressed and functions in the
brain—has been associated with schizophrenia and autism spectrum
disorders but not with bipolar disorders (Vacic et al. 2011). The fact that
these duplications increase the amount of VIPR2 expressed raises the
question of whether antagonists could be developed as potential targeted
therapeutics for schizophrenia.
Common Variants
Until very recently, most studies of common variants focused on candidate
genes identified by their known function or position within chromosomal
regions showing linkage to disease in gene mapping studies. However, the
extent to which these earlier reports may represent true associations remains
unclear, due to many biases inherent in such approaches. The field has since
moved toward the use of GWASs, which, by controlling for multiple
hypothesis testing, increases the confidence that significant associations are
true positives. However, GWASs also require large samples to detect
significant associations. Therefore, recent international efforts to combine
all moderate-sized GWASs in psychiatric illness into a large analysis via the
Psychiatric Genomics Consortium are under way and have generated a
series of analyses with increasing sample size. The most recent analysis of
common variation in schizophrenia involved 36,989 subjects with
schizophrenia and 113,075 control subjects. Investigators found 108
significantly associated genomic loci, implicating multiple genes and
ncRNAs as possibly associated with increased risk for schizophrenia
(Schizophrenia Working Group of the Psychiatric Genomics Consortium
2014). A number of these findings have highlighted consistent trends
observed in earlier studies. First and foremost has been the significant
association of schizophrenia and bipolar disorders with genes involved in
calcium signaling, including the calcium channel, voltage-dependent, L
type, alpha 1C subunit gene (CACNA1C) and the calcium channel, voltage-
dependent, beta 2 subunit gene (CACNB2). These findings have clear
potential therapeutic implications (e.g., for agents active at these receptors),
although much work remains necessary to elucidate the biological
mechanisms underlying these associations. A second important and
consistent finding was the identification of associations with variants in the
DRD2 gene and in genes affecting glutamate signaling (e.g. GRM3,
GRIN2A, SRR, GRIA1), supporting prior hypothesized dopaminergic and
glutamatergic mechanisms in disease.
Genetic studies have also found schizophrenia to be associated with the
ncRNA miR-137, a microRNA that acts to reduce expression of multiple
gene targets and is also associated with increased risk for schizophrenia
(Ripke et al. 2013). In addition, many genes associated with schizophrenia
are themselves potentially regulated by miR-137. This publication also
found many potential associations of another class of RNA—long ncRNA
(lncRNA)—with risk for schizophrenia. The functions of lncRNA are still
being uncovered but are expected to have a role in the regulation of gene
transcription, further highlighting the extent to which genetic variation in
schizophrenia may act through complex regulation of gene expression
rather than through alterations in specific protein sequences (Cookson et al.
2009). Finally, as in previous studies, recent investigations have found the
major histocompatibility complex to be associated with schizophrenia,
although the complex genetics of the major histocompatibility complex
make the implications of this association somewhat unclear at this time.
The efforts of the Psychiatric Genomics Consortium have also led to the
aforementioned discovery of shared common genetic variation among
individuals with schizophrenia, bipolar disorders, major depressive
disorder, and other disorders (see beginning of section “Psychiatric
Disorders of Adulthood: Schizophrenia, Bipolar Disorders, and Major
Depressive Disorder”), and efforts to identify genetic variation that may
contribute to risk across disorders have since been under way. The largest of
these to date examined 33,332 individual cases diagnosed with bipolar
disorders, major depressive disorder, schizophrenia, autism spectrum
disorder, or ADHD, as well as 27,888 healthy controls of European ancestry
(Cross-Disorder Group of the Psychiatric Genomics Consortium and
Genetic Risk Outcome of Psychosis [GROUP] Consortium 2013).
Significant associations with SNPs in ITIH3, AS3MT, and CACNB2 were
found, with similar associations to risk for all five disorders. In addition, a
significant association with a SNP in CACNA1C was driven by the
schizophrenia and bipolar groups. In contrast to these shared genetic
associations, no studies have identified significant associations of common
variants with major depressive disorder alone.
The inability to detect an association of common variants with major
depressive disorder alone, as well as reports of the relatively lower
heritability of major depressive disorder in comparison with schizophrenia
or bipolar disorders, likely reflects a greater role of environmental
precipitants in this syndrome. Identifying genetic variants that contribute to
major depressive disorder may require the study of groups of individuals
whose major depressive disorder occurs in the context of a specific or a
limited set of environmental factors. For example, a recent finding that
attracted a great deal of attention in studies of major depressive disorder
was the report that short alleles of a 44-base-pair insertion/deletion
polymorphism in the serotonin transporter gene SLC6A4 moderated
environmental effects on depression, increasing the risk for depression
induced by stressful life events or childhood maltreatment (Caspi et al.
2003). The possibility that genetic variation in SLC6A4 might influence
stress reactivity has been supported by several smaller studies using
different approaches (see review in Levinson 2006). However, two
subsequent large-scale studies were unable to replicate these findings
(Gillespie et al. 2005; Surtees et al. 2006). More recent meta-analyses have
continued to provide conflicting results, adding uncertainty to the
relationship between genetic variation in SLC6A4, stressful life events, and
major depressive disorder (Karg et al. 2011). Despite the lack of
convergence on SLC6A4, this approach could be used to identify common
variants using GWASs, if sufficiently large samples characterized for
environmental exposure to stressful events could be identified.
The consideration of specified environmental factors may have particular
relevance for the study of the genetics of late-life depression. Whereas
some environmental stressors that are common in late life (e.g.,
bereavement) may share mechanisms with early life stressors, other
important environmental contributors to late-life depression, such as
cerebrovascular disease, are likely to interact with a different set of genetic
variants. There have been some efforts to detect common variants that may
interact with cerebrovascular disease to alter the risk for late-life depression
(reviewed in Chapter 9, “Depressive Disorders”). At present, these studies
have been limited in sample size and to the study of a subset of SNPs
occurring in a few candidate genes. Therefore, they must be interpreted
with caution, because these designs are prone to generate both false-positive
and false-negative results when common variants are considered.
Nevertheless, such work is accelerating, driven by advances in the
identification of genetic variants associated with ischemic and large artery
stroke (Dichgans et al. 2014).
Conclusion
Medical genetics has entered the postgenomics era: the pace at which
genetic variants are being discovered continues to accelerate, and such
discoveries are beginning to inform the development of novel and much-
needed interventions. With faster and increasingly inexpensive genomic
approaches becoming available, untangling the genetic underpinnings of
some of the more complex disorders in geriatric psychiatry will ideally soon
follow. Although the impact of these developments on practicing geriatric
psychiatrists has so far been limited, it will soon be felt. The potential for
genomics to contribute to individualized clinical care has long been
recognized, and many optimistic scenarios for its use have been proposed.
In fact, several academic medical centers and integrated health systems
have already embarked on the implementation of genomic medicine in
clinical practice (Manolio et al. 2013).
Although advancements in medical genomics continue, it is important
for us to emphasize the reasons why identifying genes that lead to the
development of complex disorders remains a challenging process. The flow
of events leading from variation in the sequence of a gene to the
development of multiple cognitive, emotional, and behavioral criteria that
define the complex neuropsychiatric disorders of later life is not linear or
straightforward. Genes encode proteins, which act within extensive
biochemical networks in which changes may be amplified or compensated
for by the effects of other proteins or environmental factors. The net result
is that the magnitude of an increase in disease risk for a complex disorder
due to any single genetic variation will be relatively small, suggesting that
the ability to predict clinical outcomes for individual patients will be quite
limited, even if one knows that a patient carries a genetic variation
associated with the disease of interest.
Given current limitations in the availability of disease-modifying
therapies for a number of neurodegenerative illnesses, as well as the
complex ethical considerations involved in genomic medicine, clinicians
must carefully consider the appropriateness of ordering genetic tests for
their patients. Because both positive and negative findings may have
significant implications for patients and their families, thorough genetic
counseling is advised before and after genetic testing is performed. Methods
for screening and diagnosing neurodegenerative illnesses will continue to
improve as additional genes are identified and correlated with increasingly
specific phenotypes, and such discoveries will hopefully inform the
development of preventive and disease-modifying therapies (Farlow and
Foroud 2013). Geriatric psychiatrists will need to be familiar with these
developments so as to effectively counsel their patients and continue to
provide state-of-the-art care.
Key Points
• The discovery of rare disease-causing mutations has been instrumental in
furthering the understanding of the neurobiology of a number of
neuropsychiatric disorders of later life, including Alzheimer’s disease,
frontotemporal dementia, and Parkinson’s disease. Furthermore, the pace
of discovery in these areas continues to accelerate.
• Many neuropsychiatric disorders of later life—including the most
common presentations of the dementias—are genetically complex
diseases, resulting from the interaction of multiple genes and
environmental factors. Such complexity limits the definitive
identification of contributory genes and reduces the ability to use genetic
information to predict clinical outcomes. Nevertheless, important strides
in this area are continuing to be made.
• Genetic testing may be useful for some of the aforementioned disorders,
including early-onset Alzheimer’s disease and frontotemporal dementia,
and the likelihood of identifying causal mutations in individuals affected
by these disorders increases in the presence of a positive family history.
References
Alagiakrishnan K, Gill SS, Fagarasanu A: Genetics and epigenetics of
Alzheimer’s disease. Postgrad Med J 88(1043):522–529, 2012
22543304
Alzheimer Disease and Frontotemporal Dementia Mutation Database, 2014.
Available at: http://www.molgen.ua.ac.be/ADMutations. Accessed
February 13, 2008.
Alzheimer’s Disease Education and Referral Center: Dominantly Inherited
Alzheimer Network Trial: An Opportunity to Prevent Dementia (DIAN
TU), 2014. Available at: http://www.nia.nih.gov/alzheimers/clinical-
trials/dominantly-inherited-alzheimer-network-trial-opportunity-
prevent-dementia. Accessed January 22, 2014.
Athena Diagnostics: Athena Diagnostics Test Catalog. Worchester, MA,
Athena Diagnostics, 2014. Available at:
http://www.athenadiagnostics.com. Accessed January 22, 2014.
Bacanu SA, Devlin B, Chowdari KV, et al: Heritability of psychosis in
Alzheimer disease. Am J Geriatr Psychiatry 13(7):624–627, 2005
16009739
Bienvenu OJ, Davydow DS, Kendler KS: Psychiatric “diseases” versus
behavioral disorders and degree of genetic influence. Psychol Med
41(1):33–40, 2011 20459884
Bird T, Knopman D, VanSwieten J, et al: Epidemiology and genetics of
frontotemporal dementia/Pick’s disease. Ann Neurol 54(suppl 5):S29–
S31, 2003 12833366
Blackwood DH, Fordyce A, Walker MT, et al: Schizophrenia and affective
disorders—cosegregation with a translocation at chromosome 1q42
that directly disrupts brain-expressed genes: clinical and P300 findings
in a family. Am J Hum Genet 69(2):428–433, 2001 11443544
Brousseau T, Legrain S, Berr C, et al: Confirmation of the epsilon 4 allele
of the apolipoprotein E gene as a risk factor for late-onset Alzheimer’s
disease. Neurology 44(2):342–344, 1994 8309588
Campion D, Flaman JM, Brice A, et al: Mutations of the presenilin I gene
in families with early onset Alzheimer’s disease. Hum Mol Genet
4(12):2373–2377, 1995 8634712
Campion D, Dumanchin C, Hannequin D, et al: Early onset autosomal
dominant Alzheimer disease: prevalence, genetic heterogeneity, and
mutation spectrum. Am J Hum Genet 65(3):664–670, 1999 10441572
Carninci P, Kasukawa T, Katayama S, et al: The transcriptional landscape of
the mammalian genome. Science 309(5740): 1559–1563, 2005
16141072
Caspi A, Sugden K, Moffitt TE, et al: Influence of life stress on depression:
moderation by a polymorphism in the 5-HTT gene. Science
301(5631):386–389, 2003 12869766
Cervilla J, Prince M, Joels S, et al: Premorbid cognitive testing predicts the
onset of dementia and Alzheimer’s disease better than and
independently of APOE genotype. J Neurol Neurosurg Psychiatry
75(8):1100–1106, 2004 15258208
Cheng Z, Ventura M, She X, et al: A genome-wide comparison of recent
chimpanzee and human segmental duplications. Nature 437(7055):88–
93, 2005 16136132
Chow TW, Alobaidy AA: Incorporating new diagnostic schemas, genetics,
and proteinopathy into the evaluation of frontotemporal degeneration.
Continuum (Minneap Minn) 19(2 Dementia):438–456, 2013 23558488
Chow TW, Miller BL, Hayashi VN, et al: Inheritance of frontotemporal
dementia. Arch Neurol 56(7):817–822, 1999 10404983
Cookson W, Liang L, Abecasis G, et al: Mapping complex disease traits
with global gene expression. Nat Rev Genet 10(3): 184–194, 2009
19223927
Coon KD, Dunckley TL, Stephan DA: A generic research paradigm for
identification and validation of early molecular diagnostics and new
therapeutics in common disorders. Mol Diagn Ther 11(1):1–14, 2007
17286446
Corder EH, Saunders AM, Strittmatter WJ, et al: Gene dose of
apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in
late onset families. Science 261(5123): 921–923, 1993 8346443
Cross-Disorder Group of the Psychiatric Genomics Consortium, Genetic
Risk Outcome of Psychosis (GROUP) Consortium: Identification of
risk loci with shared effects on five major psychiatric disorders: a
genome-wide analysis. Lancet 381(9875):1371–1379, 2013 23453885
Cross-Disorder Group of the Psychiatric Genomics Consortium, Lee SH,
Ripke S, et al: Genetic relationship between five psychiatric disorders
estimated from genome-wide SNPs. Nat Genet 45(9):984–994, 2013
23933821
Cruts M, van Duijn CM, Backhovens H, et al: Estimation of the genetic
contribution of presenilin-1 and -2 mutations in a population-based
study of presenile Alzheimer disease. Hum Mol Genet 7(1):43–51,
1998 9384602
DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al: Expanded
GGGGCC hexanucleotide repeat in noncoding region of C9ORF72
causes chromosome 9p-linked FTD and ALS. Neuron 72(2):245–256,
2011 21944778
de Rijk MC, Breteler MM, den Breeijen JH, et al: Dietary antioxidants and
Parkinson disease. The Rotterdam Study. Arch Neurol 54(6):762–765,
1997 9193212
Devanand DP, Jacobs DM, Tang MX, et al: The course of psychopathologic
features in mild to moderate Alzheimer disease. Arch Gen Psychiatry
54(3):257–263, 1997 9075466
Dichgans M, Malik R, König IR, et al: Shared genetic susceptibility to
ischemic stroke and coronary artery disease: a genome-wide analysis of
common variants. Stroke 45(1):24–36, 2014 24262325
Dobson-Stone C, Hallupp M, Bartley L, et al: C9ORF72 repeat expansion
in clinical and neuropathologic frontotemporal dementia cohorts.
Neurology 79(10):995–1001, 2012 22875086
Doherty JL, O’Donovan MC, Owen MJ: Recent genomic advances in
schizophrenia. Clin Genet 81(2):103–109, 2012 21895634
Eder J, Hommel U, Cumin F, et al: Aspartic proteases in drug discovery.
Curr Pharm Des 13(3):271–285, 2007 17313361
Ertekin-Taner N: Genetics of Alzheimer’s disease: a centennial review.
Neurol Clin 25(3):611–667, v, 2007 17659183
Farlow JL, Foroud T: The genetics of dementia. Semin Neurol 33(4):417–
422, 2013 24234360
Farrer LA, Cupples LA, Haines JL, et al; APOE and Alzheimer Disease
Meta Analysis Consortium: Effects of age, sex, and ethnicity on the
association between apolipoprotein E genotype and Alzheimer disease:
a meta-analysis. JAMA 278(16):1349–1356, 1997 9343467
Fernandes SA, Douglas AG, Varela MA, et al: Oligonucleotide-based
therapy for FTD/ALS caused by the C9orf72 repeat expansion: a
perspective. J Nucleic Acids 2013:208245. Published online Nov 17,
2013. 24349764
Gasser T: Genetics of Parkinson’s disease. Curr Opin Neurol 18(4):363–
369, 2005 16003110
Ghosh AK, Brindisi M, Tang J: Developing β-secretase inhibitors for
treatment of Alzheimer’s disease. J Neurochem 120 (suppl 1):71–83,
2012 22122681
Gillespie NA, Whitfield JB, Williams B, et al: The relationship between
stressful life events, the serotonin transporter (5-HTTLPR) genotype
and major depression. Psychol Med 35(1):101–111, 2005 15842033
Glenner GG, Wong CW: Alzheimer’s disease and Down’s syndrome:
sharing of a unique cerebrovascular amyloid fibril protein. Biochem
Biophys Res Commun 122(3):1131–1135, 1984 6236805
Goate A, Chartier-Harlin MC, Mullan M, et al: Segregation of a missense
mutation in the amyloid precursor protein gene with familial
Alzheimer’s disease. Nature 349(6311):704–706, 1991 1671712
Goldman JS: New approaches to genetic counseling and testing for
Alzheimer’s disease and frontotemporal degeneration. Curr Neurol
Neurosci Rep 12(5):502–510, 2012 22773362
Goldman JS, Hahn SE, Catania JW, et al: Genetic counseling and testing for
Alzheimer disease: joint practice guidelines of the American College of
Medical Genetics and the National Society of Genetic Counselors.
Genet Med 13(6):597–605, 2011 21577118
Guerreiro R, Wojtas A, Bras J, et al: TREM2 variants in Alzheimer’s
disease. N Engl J Med 368(2):117–127, 2013 23150934
Gulsuner S, Walsh T, Watts AC, et al: Spatial and temporal mapping of de
novo mutations in schizophrenia to a fetal prefrontal cortical network.
Cell 154(3):518–529, 2013 23911319
Guo T, Hobbs DW: Development of BACE1 inhibitors for Alzheimer’s
disease. Curr Med Chem 13(15):1811–1829, 2006 16787223
Hardy J: A hundred years of Alzheimer’s disease research. Neuron 52(1):3–
13, 2006 17015223
Hardy J, Cai H, Cookson MR, et al: Genetics of Parkinson’s disease and
parkinsonism. Ann Neurol 60(4):389–398, 2006 17068789
Henderson AS, Easteal S, Jorm AF, et al: Apolipoprotein E allele epsilon 4,
dementia, and cognitive decline in a population sample. Lancet
346(8987):1387–1390, 1995 7475820
Hollingworth P, Hamshere ML, Holmans PA, et al: Increased familial risk
and genomewide significant linkage for Alzheimer’s disease with
psychosis. Am J Med Genet B Neuropsychiatr Genet 144B(7):841–
848, 2007 17492769
Hollingworth P, Sweet R, Sims R, et al: Genome-wide association study of
Alzheimer’s disease with psychotic symptoms. Mol Psychiatry
17(12):1316–1327, 2012 22005930
Hudson TJ: Genome variation and personalized cancer medicine. J Intern
Med 274(5):440–450, 2013 23751076
Hutton M, Busfield F, Wragg M, et al: Complete analysis of the presenilin 1
gene in early onset Alzheimer’s disease. Neuroreport 7(3):801–805,
1996 8733749
Imbimbo BP, Giardina GA: γ-Secretase inhibitors and modulators for the
treatment of Alzheimer’s disease: disappointments and hopes. Curr Top
Med Chem 11(12):1555–1570, 2011 21510832
Inoue K, Lupski JR: Molecular mechanisms for genomic disorders. Annu
Rev Genomics Hum Genet 3:199–242, 2002 12142364
International HapMap Project: International HapMap Project Database,
2014. Available at: http://hapmap.ncbi.nlm.nih.gov/. Accessed
February 13, 2014.
International Parkinson’s Disease Genomics Consortium, Wellcome Trust
Case Control Consortium 2: A two-stage meta-analysis identifies
several new loci for Parkinson’s disease. PLoS Genet 7(6): e1002142,
2011 21738488
Jonsson T, Stefansson H, Steinberg S, et al: Variant of TREM2 associated
with the risk of Alzheimer’s disease. N Engl J Med 368(2):107–116,
2013 23150908
Kamboh MI, Demirci FY, Wang X, et al: Genome-wide association study of
Alzheimer’s disease. Transl Psychiatr 2:e117, 2012 22832961
Karg K, Burmeister M, Shedden K, et al: The serotonin transporter
promoter variant (5-HTTLPR), stress, and depression meta-analysis
revisited: evidence of genetic moderation. Arch Gen Psychiatry
68(5):444–454, 2011 21199959
Kaufer DI, Cummings JL, Christine D, et al: Assessing the impact of
neuropsychiatric symptoms in Alzheimer’s disease: the
Neuropsychiatric Inventory Caregiver Distress Scale. J Am Geriatr Soc
46(2):210–215, 1998 9475452
Kenny EM, Cormican P, Furlong S, et al: Excess of rare novel loss-of-
function variants in synaptic genes in schizophrenia and autism
spectrum disorders. Mol Psychiatry 19(8):872–879, 2014 24126926
Klages JD, Fisk JD, Rockwood K: APOE genotype, memory test
performance, and the risk of Alzheimer’s disease in the Canadian Study
of Health and Aging. Dement Geriatr Cogn Disord 15(1):1–5, 2003
12457072
Koboldt DC, Miller RD: Identification of functional SNPs in noncoding
regions of the human genome. 2007. Available at
http://snp.wustl.edu/snp-and-fp-tdi-resources/abstracts/Koboldt-
HUGO-Functional-SNPs.pdf. Accessed February 12, 2008.
Lagier-Tourenne C, Baughn M, Rigo F, et al: Targeted degradation of sense
and antisense C9orf72 RNA foci as therapy for ALS and
frontotemporal degeneration. Proc Natl Acad Sci USA 110(47):E4530–
E4539, 2013 24170860
Lambert JC, Ibrahim-Verbaas CA, Harold D, et al: Meta-analysis of 74,046
individuals identifies 11 new susceptibility loci for Alzheimer’s
disease. Nat Genet 45(12): 1452–1458, 2013 24162737
Levinson DF: The genetics of depression: a review. Biol Psychiatry
60(2):84–92, 2006 16300747
Levy S, Sutton G, Ng PC, et al: The diploid genome sequence of an
individual human. PLoS Biol 5(10):e254, 2007 17803354
Levy-Lahad E, Wasco W, Poorkaj P, et al: Candidate gene for the
chromosome 1 familial Alzheimer’s disease locus. Science
269(5226):973–977, 1995a 7638622
Levy-Lahad E, Wijsman EM, Nemens E, et al: A familial Alzheimer’s
disease locus on chromosome 1. Science 269(5226):970–973, 1995b
7638621
Lupski JR: Structural variation in the human genome. N Engl J Med
356(11):1169–1171, 2007 17360997
Malhotra D, Sebat J: CNVs: harbingers of a rare variant revolution in
psychiatric genetics. Cell 148(6):1223–1241, 2012 22424231
Manolio TA, Chisholm RL, Ozenberger B, et al: Implementing genomic
medicine in the clinic: the future is here. Genet Med 15(4):258–267,
2013 23306799
Mayeux R, Stern Y, Ottman R, et al: The apolipoprotein epsilon 4 allele in
patients with Alzheimer’s disease. Ann Neurol 34(5):752–754, 1993
8239575
Murray PS, Kumar S, Demichele-Sweet MA, et al: Psychosis in
Alzheimer’s Disease. Biol Psychiatry 75(7):542–552, 2014 24103379
Neary D, Snowden J, Mann D: Frontotemporal dementia. Lancet Neurol
4(11):771–780, 2005 16239184
Neumann M, Sampathu DM, Kwong LK, et al: Ubiquitinated TDP-43 in
frontotemporal lobar degeneration and amyotrophic lateral sclerosis.
Science 314(5796):130–133, 2006 17023659
Parkes M, Barrett JC, Prescott NJ, et al: Sequence variants in the autophagy
gene IRGM and multiple other replicating loci contribute to Crohn’s
disease susceptibility. Nat Genet 39(7):830–832, 2007 17554261
Patel L, Parker B, Yang D, et al: Translational genomics in cancer research:
converting profiles into personalized cancer medicine. Cancer Biol
Med 10(4):214–220, 2013 24349831
Pericak-Vance MA, Haines JL: Genetic susceptibility to Alzheimer disease.
Trends Genet 11(12):504–508, 1995 8533168
Prasher VP, Farrer MJ, Kessling AM, et al: Molecular mapping of
Alzheimer-type dementia in Down’s syndrome. Ann Neurol 43(3):380–
383, 1998 9506555
Puschmann A: Monogenic Parkinson’s disease and parkinsonism: clinical
phenotypes and frequencies of known mutations. Parkinsonism Relat
Disord 19(4): 407–415, 2013 23462481
Rebeck GW, Reiter JS, Strickland DK, et al: Apolipoprotein E in sporadic
Alzheimer’s disease: allelic variation and receptor interactions. Neuron
11(4):575–580, 1993 8398148
Redon R, Ishikawa S, Fitch KR, et al: Global variation in copy number in
the human genome. Nature 444(7118):444–454, 2006 17122850
Ripke S, O’Dushlaine C, Chambert K, et al: Genome-wide association
analysis identifies 13 new risk loci for schizophrenia. Nat Genet
45(10):1150–1159, 2013 23974872
Rossor MN, Fox NC, Beck J, et al: Incomplete penetrance of familial
Alzheimer’s disease in a pedigree with a novel presenilin-1 gene
mutation. Lancet 347(9014): 1560, 1996 8684135
Rovelet-Lecrux A, Hannequin D, Raux G, et al: APP locus duplication
causes autosomal dominant early onset Alzheimer disease with cerebral
amyloid angiopathy. Nat Genet 38(1):24–26, 2006 16369530
Samonte RV, Eichler EE: Segmental duplications and the evolution of the
primate genome. Nat Rev Genet 3(1):65–72, 2002 11823792
Saunders AM, Strittmatter WJ, Schmechel D, et al: Association of
apolipoprotein E allele epsilon 4 with late-onset familial and sporadic
Alzheimer’s disease. Neurology 43(8):1467–1472, 1993 8350998
Schellenberg GD, Bird TD, Wijsman EM, et al: Genetic linkage evidence
for a familial Alzheimer’s disease locus on chromosome 14. Science
258(5082):668–671, 1992 1411576
Schizophrenia Psychiatric GWAS Consortium: Genome-wide association
study identifies five new schizophrenia loci. Nat Genet 43(10):969–
976, 2011 21926974
Schizophrenia Working Group of the Psychiatric Genomics Consortium:
Biological insights from 108 schizophrenia-associated genetic loci.
Nature 511(7510):421–427, 2014 25056061
Schmechel DE, Saunders AM, Strittmatter WJ, et al: Increased amyloid
beta-peptide deposition in cerebral cortex as a consequence of
apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl
Acad Sci USA 90(20):9649–9653, 1993 8415756
Sebat J, Lakshmi B, Malhotra D, et al: Strong association of de novo copy
number mutations with autism. Science 316(5823):445–449, 2007
17363630
Selkoe DJ, American College of Physicians, American Physiological
Society: Alzheimer disease: mechanistic understanding predicts novel
therapies. Ann Intern Med 140(8):627–638, 2004 15096334
Seshadri S, Wolf PA, Beiser A, et al: Lifetime risk of dementia and
Alzheimer’s disease: the impact of mortality on risk estimates in the
Framingham Study. Neurology 49(6):1498–1504, 1997 9409336
Sherrington R, Rogaev EI, Liang Y, et al: Cloning of a gene bearing
missense mutations in early-onset familial Alzheimer’s disease. Nature
375(6534):754–760, 1995 7596406
Sleegers K, Brouwers N, Gijselinck I, et al: APP duplication is sufficient to
cause early onset Alzheimer’s dementia with cerebral amyloid
angiopathy. Brain 129(Pt 11):2977–2983, 2006 16921174
St George-Hyslop P, Haines J, Rogaev E, et al: Genetic evidence for a novel
familial Alzheimer’s disease locus on chromosome 14. Nat Genet
2(4):330–334, 1992 1303289
Sullivan PF, Daly MJ, O’Donovan M: Genetic architectures of psychiatric
disorders: the emerging picture and its implications. Nat Rev Genet
13(8):537–551, 2012 22777127
Surtees PG, Wainwright NW, Willis-Owen SA, et al: Social adversity, the
serotonin transporter (5-HTTLPR) polymorphism and major depressive
disorder. Biol Psychiatry 59(3):224–229, 2006 16154545
Swearer JM, Drachman DA, O’Donnell BF, et al: Troublesome and
disruptive behaviors in dementia: relationships to diagnosis and disease
severity. J Am Geriatr Soc 36(9):784–790, 1988 3411060
Sweet RA, Nimgaonkar VL, Devlin B, et al: Increased familial risk of the
psychotic phenotype of Alzheimer disease. Neurology 58(6):907–911,
2002 11914406
Sweet RA, Bennett DA, Graff-Radford NR, et al: Assessment and familial
aggregation of psychosis in Alzheimer’s disease from the National
Institute on Aging Late Onset Alzheimer’s Disease Family Study.
Brain 133(Pt 4):1155–1162, 2010 20147454
Taft RJ, Pang KC, Mercer TR, et al: Non-coding RNAs: regulators of
disease. J Pathol 220(2):126–139, 2010 19882673
Theuns J, Marjaux E, Vandenbulcke M, et al: Alzheimer dementia caused
by a novel mutation located in the APP C-terminal intracytosolic
fragment. Hum Mutat 27(9):888–896, 2006 16917905
Thomson PA, Malavasi EL, Grünewald E, et al: DISC1 genetics, biology
and psychiatric illness. Front Biol (Beijing) 8(1):1–31, 2013 23550053
Todd JA, Walker NM, Cooper JD, et al: Robust associations of four new
chromosome regions from genome-wide analyses of type 1 diabetes.
Nat Genet 39(7):857–864, 2007 17554260
Tunstall N, Owen MJ, Williams J, et al: Familial influence on variation in
age of onset and behavioural phenotype in Alzheimer’s disease. Br J
Psychiatry 176:156–159, 2000 10755053
Vacic V, McCarthy S, Malhotra D, et al: Duplications of the neuropeptide
receptor gene VIPR2 confer significant risk for schizophrenia. Nature
471(7339):499–503, 2011 21346763
Vassar R, Bennett BD, Babu-Khan S, et al: Beta-secretase cleavage of
Alzheimer’s amyloid precursor protein by the transmembrane aspartic
protease BACE. Science 286(5440):735–741, 1999 10531052
Wang X, Shen Y, Chen W: Progress in frontotemporal dementia research.
Am J Alzheimers Dis Other Demen 28(1):15–23, 2013 23221030
Warren JD, Rohrer JD, Rossor MN: Clinical review: frontotemporal
dementia. BMJ 347:f4827, 2013 23920254
Wellcome Trust Case Control Consortium: Genome-wide association study
of 14,000 cases of seven common diseases and 3,000 shared controls.
Nature 447(7145): 661–678, 2007 17554300
Wragg RE, Jeste DV: Overview of depression and psychosis in Alzheimer’s
disease. Am J Psychiatry 146(5):577–587, 1989 2653053
Xu B, Roos JL, Dexheimer P, et al: Exome sequencing supports a de novo
mutational paradigm for schizophrenia. Nat Genet 43(9):864–868,
2011 21822266
Xu B, Ionita-Laza I, Roos JL, et al: De novo gene mutations highlight
patterns of genetic and neural complexity in schizophrenia. Nat Genet
44(12):1365–1369, 2012 23042115
Zeggini E, Weedon MN, Lindgren CM, et al: Replication of genome-wide
association signals in U.K. samples reveals risk loci for type 2 diabetes.
Science 316(5829): 1336–1341, 2007 17463249
Suggested Resource
GeneCards® (www.genecards.org)—Searchable, integrated database
providing comprehensive information on all known and predicted
human genes
PART II
History
The elements of a diagnostic workup of the elderly patient are presented in
Table 4–1. To obtain historical information, the clinician should first
interview the patient, if that is feasible, and then ask the patient’s
permission to interview family members. Members from at least two
generations, if available for interview, can expand the perspective on the
older adult’s impairment. If the patient has difficulty providing an accurate
or understandable history, the clinician should concentrate especially on
eliciting the symptoms or problems that the patient perceives as being most
disabling, then fill the historical gap with data from the family.
Present Illness
DSM-5 (American Psychiatric Association 2013) provides the clinician
with a useful catalog of symptoms and behaviors of psychiatric interest that
are relevant to the diagnosis of the present illness. Symptoms are bits of
data—the most visible part of the clinical picture and generally the part
most easily agreed on among clinicians. Symptoms should be defined in
such a way that if multiple clinicians independently obtain equivalent
information, they would have minimal disagreement about the presence or
absence of a symptom. The decision about whether those symptoms form a
syndrome or derive from a particular etiology must be determined
independently of the data collection on symptoms.
Past History
Next, the clinician must review the past history of symptoms and episodes.
Has the individual had similar episodes in the past? How long did the
episodes last? When did they occur? How many times in the patient’s
lifetime have such episodes occurred? Unfortunately, the older adult may
not equate present distress with past episodes that are symptomatically
similar, so the perspective of the family is especially valuable in the attempt
to link current and past episodes.
Other psychiatric and medical problems should be reviewed as well,
especially medical illnesses that have led to hospitalization and the use of
medication. Not infrequently, an older adult has experienced a major illness
or trauma in childhood or as a younger adult but views this information as
being of no relevance to the present episode and therefore dismisses it.
Probes to elicit these data are essential. Older adults may ignore or even
forget past psychiatric difficulties, especially if these difficulties were
disguised. For example, mood swings in early or middle life may have
occurred during periods of excessive and productive activity, episodes of
excessive alcohol intake, or periods of vague, undiagnosed physical
problems. Previous periods of overt disability in usual activities may flag
those episodes. An older person sometimes becomes angry or irritated when
the clinician continues to probe. Reassurance regarding the importance of
obtaining this information will generally suffice, except when dealing with
a patient who cannot tolerate the discomfort and distress, even for brief
periods. Older persons who have chronic and moderately severe anxiety or
a histrionic personality style, as well as distressed Alzheimer’s patients,
tolerate their symptoms poorly.
Family History
The distribution of psychiatric symptoms and illnesses in the family should
be determined next. The older person with symptoms consistent with major
neurocognitive impairment may have a family history of a disorder such as
major neurocognitive disorder due to Alzheimer’s disease. The genogram
can be valuable for charting the distribution of mental illness and other
relevant behaviors throughout the family tree. This genogram should
include parents, blood-related aunts and uncles, brothers and sisters,
spouse(s), children, grandchildren, and great-grandchildren (recognizing
that data from many family members will not be complete). A history
should be obtained about institutionalization, significant memory problems
in family members, hospitalization for a nervous breakdown or depressive
disorder, suicide, alcohol abuse and dependence, electroconvulsive therapy,
long-term residence in a mental health facility (and possibly a diagnosis of
schizophrenia), and use of mental health services by family members
(Blazer 1984).
Of relevance to the pharmacological treatment of certain disorders—
especially depression—in older adults is the tendency of individuals in a
family to respond therapeutically to the same pharmacological agent. If the
older adult has a depressive disorder and if biological relatives have been
treated effectively for depression, the clinician should determine what
pharmacological agent was used to treat the depression. For example, a
positive response to sertraline in a family member of the depressed older
patient could make sertraline the drug of choice in treating that patient,
assuming side effects are not at issue (Ayd 1975).
Accurate genetic information can be better obtained when family
members from more than one generation are interviewed. Many psychiatric
disorders are characterized by a variety of symptoms, so asking the patient
or one family member for a history of depression is insufficient. Research
on the genetic expression of psychiatric disorders in families requires the
psychiatric investigator to interview directly as many family members as
possible to determine accurately the distribution of disorders throughout the
family. Such detailed family assessment is not feasible for clinicians, yet a
telephone call to a relative with permission from the patient may become a
standard of clinical assessment as the genetics of psychiatric disorders are
clarified.
Context
Psychiatric disorders occur in a biomedical and psychosocial context.
Although the clinician will try to determine what medical problems the
patient has experienced, it is possible to overlook a variation in the relative
contribution of these medical disorders to psychopathology or to overlook
the psychosocial contribution to the onset and continuance of the problem.
Has the spouse of the older adult undergone a change? Are the middle-aged
children managing high stress, such as simultaneously caring for an
emotionally disturbed child and the loss of employment? Are the
grandchildren placing emotional stress on the elderly patient, perhaps by
requesting money? Has the economic status of the older adult deteriorated?
Has the availability of medical care changed? Although many psychiatric
disorders are biologically driven, they do not occur in a psychosocial
vacuum. Environmental precipitants remain important in the web of
causation leading to the onset of an episode of emotional distress and are
critical to the assessment of the older adult.
Medication History
It is essential to evaluate the medication history of the older adult. A careful
review of current and past medications by the clinician, a nurse, or a
physician’s assistant is essential. The older person should be asked to bring
to the appointment all pill bottles, a list of medications taken, and the
dosage schedule. A comparison between the written schedule and the pill
containers will frequently expose some discrepancy. Both prescription and
over-the-counter drugs, such as laxatives and vitamins, should be recorded.
The clinician can then identify the medications that potentially lead to drug-
drug interactions and ask about them during subsequent patient visits.
Most elderly persons take a variety of medicines simultaneously, and the
potential for drug-drug interaction is high. For example, concomitant use of
fluoxetine and warfarin has been associated with an increase in the half-life
of warfarin, which could lead to severe bruising (although this finding is not
well documented). Some medications prescribed for older persons—such as
the β-blocker propranolol and calcium channel blockers—can exacerbate or
produce depressive symptoms.
Older persons are less likely than younger persons to have a substance
use disorder, but a careful history of alcohol and drug intake (especially
nonprescription use of prescription medications) is essential to the
diagnostic workup. Although older persons do not usually volunteer
information about their substance intake, they are generally forthcoming
when asked about their drinking habits.
Medical History
Given the high likelihood of comorbid medical problems associated with
psychiatric disorders in late life, a comprehensive medical history is
essential. Most older persons see a primary care physician regularly
(although decreasing payments from Medicare render this assumption less
accurate each year). The geriatric psychiatrist should obtain medical
records, if possible. Major illnesses should be recorded. A brief phone call
to the primary care physician can be extremely useful.
Family Assessment
Clinicians working with older adults must be equipped to evaluate the
family—both its functionality and its potential as a resource for the older
adult. Geriatric psychiatry, almost by definition, is family psychiatry. The
family assessment is best done, if possible, in conjunction with a social
worker. Just as an elevated white blood cell count is not pathognomonic for
a particular infectious agent yet is critical to the diagnosis, the complaint
that “my family no longer loves me” does not reveal the specific problems
in the family yet does highlight the need to assess the potential of that
family for providing care and support for the older adult (Blazer 1984). The
purpose of a comprehensive diagnostic family workup is to determine the
nature of the family structure in interaction, the presence or absence of a
crisis in the family, and the type and amount of support available to the
older adult.
A primary goal of the clinician, as advocate for the older adult with
psychiatric disturbance, is to facilitate family support for the elder during a
time of disability. At least four parameters of support are important for the
clinician to evaluate as the treatment plan evolves: 1) the availability of
family members to the older person over time; 2) the tangible services
provided by the family to the older person; 3) the perception of family
support by the older patient (and therefore the willingness of the patient to
cooperate and accept support); and 4) tolerance by the family of specific
behaviors that derive from the psychiatric disorder.
The clinician should ask the older person, “If you become ill, is there a
family member who will take care of you for a short period of time?” Next,
the availability of family members who can care for the older adult over an
extended period should be determined. If a particular member is designated
as the primary caregiver, plans for respite care should be discussed. Given
the increased focus on short hospital stays and the documented higher levels
of impairment on discharge, the availability of family members becomes
essential to the effective care of the older adult after hospitalization for a
psychiatric disorder or a combined medical and psychiatric disorder.
What specific, tangible services can be provided to the older adult by
family members? Even the most devoted spouse can be limited in the
delivery of certain services because, for example, he or she does not drive a
car, and therefore cannot provide transportation, or is not physically strong
enough to provide certain types of nursing care. Generic services of special
importance in at-home support of the older adult with psychiatric
impairment include transportation; nursing services (e.g., administering
medications at home); physical therapy; checking on or continuous
supervision of the patient; homemaker and household services; meal
preparation; administrative, legal, and protective services; financial
assistance; living quarters; and coordination of the delivery of services.
These services are considered generic because they can be defined in terms
of their activities, regardless of who provides each service. Assessing the
range and extent of service delivery by the family to the older person with
functional impairment provides a convenient barometer of the economic,
social, and emotional burdens placed on the family.
Regardless of the level and types of services provided by the family to
the older person, if these services are to be effective, it is beneficial for the
older individual to perceive that he or she lives in a supportive environment.
Intangible supports include the perception of a dependable network,
participation or interaction in the network, a sense of belonging to the
network, intimacy with network members, and a sense of usefulness to the
family (Blazer and Kaplan 1983). The sense of usefulness may be of less
importance to some older adults who believe they have contributed to the
family for many years and therefore deserve reciprocal services in their
waning years. Unfortunately, family members, frequently stressed across
generations, may not recognize this reciprocal responsibility.
Family tolerance of specific behaviors may not correlate with overall
support. Every person has a level of tolerance for specific behaviors that are
especially difficult to manage. Sanford (1975) found that the following
behaviors were tolerated by families of older persons with impairments (in
decreasing percentages): incontinence of urine (81%), personality conflicts
(54%), falls (52%), physically aggressive behavior (44%), inability to walk
unaided (33%), daytime wandering (33%), and sleep disturbance (16%).
These relative frequencies may appear counterintuitive, because
incontinence is generally considered particularly aversive to family
members; however, although the outcome of incontinence can be corrected
easily enough, a few nights of no sleep can easily extend family members
beyond their capabilities for serving a parent, sibling, or spouse.
Key Points
• The diagnostic interview is the cornerstone of assessment and treatment
assignment for the older adult with psychiatric impairment.
• A thorough medication history, although it takes time to obtain, saves
valuable time and complications in the treatment of psychiatric disorders
in older adults.
• Functional status (i.e., the ability to perform usual activities of daily
living) is often as important as diagnosis in tracking the progress of
treatment of psychiatric disorders in older adults.
• Geriatric psychiatry is family psychiatry.
• What is gained in reliability by using a structured diagnostic interview is
offset by the loss of valuable information about the subjective feelings of
the older adult and the context of the emergence of symptoms.
• Speak clearly and slowly but not in a patronizing way to the older adult,
who might have a hearing impairment.
References
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Ayd FJ: Treatment-resistant patients: a moral, legal and therapeutic
challenge, in Rational Psychopharmacotherapy and the Right to
Treatment. Edited by Ayd FJ. Baltimore, MD, Ayd Medical
Communications, 1975, pp 3–15
Blazer DG: The OARS Durham surveys: description and application, in
Multidimensional Functional Assessment: The OARS Methodology—
A Manual, 2nd Edition. Durham, NC, Duke University Center for the
Study of Aging and Human Development, 1978a, pp 75–88
Blazer DG: Techniques for communicating with your elderly patient.
Geriatrics 33:79–80, 83–84, 1978b 710895
Blazer DG: Depression in Late Life. St Louis, MO, CV Mosby, 1982
Blazer DG: Evaluating the family of the elderly patient, in A Family
Approach to Health Care in the Elderly. Edited by Blazer D, Siegler IC.
Menlo Park, CA, Addison-Wesley, 1984, pp 13–32
Blazer DG, Kaplan BH: The assessment of social support in an elderly
community population. Am J Soc Psychiatry 3:29–36, 1983
Copeland JRM, Kelleher MJ, Kellett JM, et al: A semi-structured clinical
interview for the assessment of diagnosis and mental state in the
elderly: the Geriatric Mental State Schedule, I: development and
reliability. Psychol Med 6(3):439–449, 1976 996204
Copeland JRM, Dewey ME, Griffiths-Jones HM: A computerized
psychiatric diagnostic system and case nomenclature for elderly
subjects: GMS and AGECAT. Psychol Med 16(1):89–99, 1986
3515380
Duke University Center for the Study of Aging and Human Development:
Multidimensional Functional Assessment: The OARS Methodology—
A Manual, 2nd Edition. Durham, NC, Duke University Center for the
Study of Aging and Human Development, 1978
Eisenberg L: Disease and illness: distinctions between professional and
popular ideas of sickness. Cult Med Psychiatry 1(1):9–23, 1977
756356
Feighner JP, Robins E, Guze SB, et al: Diagnostic criteria for use in
psychiatric research. Arch Gen Psychiatry 26(1):57–63, 1972 5009428
First MB, Spitzer RL, Gibbon M: Structured Clinical Interview for DSM-
IV. Washington, DC, American Psychiatric Press, 1997
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Hachinski VC, Iliff LD, Zilhka E, et al: Cerebral blood flow in dementia.
Arch Neurol 32(9):632–637, 1975 1164215
Inouye SK, van Dyck CH, Alessi CA, et al: Clarifying confusion: the
confusion assessment method: a new method for detection of delirium.
Ann Intern Med 113(12):941–948, 1990 2240918
Kessler RC, Ustün TB: The World Mental Health (WMH) Survey Initiative
Version of the World Health Organization (WHO) Composite
International Diagnostic Interview (CIDI). Int J Methods Psychiatr Res
13(2):93–121, 2004 15297906
Kessler RC, Berglund P, Demler O, et al: Lifetime prevalence and age-of-
onset distributions of DSM-IV disorders in the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):593–602, 2005
15939837
Linn L: Clinical manifestations of psychiatric disorders, in Comprehensive
Textbook of Psychiatry, 3rd Edition, Vol 1. Edited by Kaplan HI,
Freedman AM, Sadock BJ. Baltimore, MD, Williams & Wilkins, 1980,
pp 990–1034
Meyers BS, Greenberg R: Late-life delusional depression. J Affect Disord
11(2):133–137, 1986 2948986
Montgomery SA, Åsberg M: A new depression scale designed to be
sensitive to change. Br J Psychiatry 134:382–389, 1979 444788
Myers JK, Weissman MM, Tischler GL, et al: Six-month prevalence of
psychiatric disorders in three communities 1980 to 1982. Arch Gen
Psychiatry 41(10):959–967, 1984 6332591
Nasreddine ZS, Phillips NA, Bédirian V, et al: The Montreal Cognitive
Assessment, MoCA: a brief screening tool for mild cognitive
impairment. J Am Geriatr Soc 53(4):695–699, 2005 15817019
National Institute of Mental Health: Development of a Dyskinetic
Movement Scale (Publ No 4). Rockville, MD, National Institute of
Mental Health, Psychopharmacology Research Branch, 1975
Radloff LS: The CES-D Scale: a self-report depression scale for research in
the general population. Appl Psychol Meas 1:385–401, 1977
Regier DA, Myers JK, Kramer M, et al: The NIMH Epidemiologic
Catchment Area program: historical context, major objectives, and
study population characteristics. Arch Gen Psychiatry 41(10):934–941,
1984 6089692
Robins LN, Cottler L, Bucholz K, et al: Diagnostic Interview Schedule for
the DSM-IV (DIS-IV). St. Louis, MO, Washington University School
of Medicine, 2000
Ross CE, Mirowsky J: Components of depressed mood in married men and
women: the Center for Epidemiologic Studies’ Depression Scale. Am J
Epidemiol 119:997–1004, 1984 6731436
Sanford JRA: Tolerance of debility in elderly dependants by supporters at
home: its significance for hospital practice. BMJ 3(5981):471–473,
1975 1156826
Spitzer RL, Endicott J, Cohen GM: Psychiatric Status Schedule, 2nd
Edition. New York, New York State Department of Mental Hygiene,
Evaluation Unit, Biometrics Research, 1968
Wing JK, Cooper JE, Sartorius N: The Measurement and Classification of
Psychiatric Symptoms. London, Cambridge University Press, 1974
World Health Organization: Measuring Health and Disability: Manual for
WHO Disability Assessment Schedule (WHODAS 2.0). Edited by
Üstün TB, Kostanjsek N, Chatterji S, Rehm J. Geneva, Switzerland,
World Health Organization, 2010. Available at:
http://whqlibdoc.who.int/publications/2010/9789241547598_eng.pdf.
Accessed October 2014.
Yesavage JA, Brink TL, Rose TL, et al: Development and validation of a
geriatric depression screening scale: a preliminary report. J Psychiatr
Res 17(1):37–49, 1982–1983 7183759
Suggested Readings
Blazer DG: Techniques for communicating with your elderly patient.
Geriatrics 33:79–80, 83–84, 1978
Inouye SK: Clarifying confusion: the Confusion Assessment Method: a new
method for detection of delirium. Ann Intern Med 113:941–950, 1990
Nasreddine ZS, Phillips NA, Bédirian V, et al: The Montreal Cognitive
Assessment (MoCA): a brief screening tool for mild cognitive
impairment. J Am Geriatr Soc 53:695–699, 2005
Othmer E, Othmer SC, Othmer JP: Psychiatric interview, history and
mental status examination, in Kaplan and Sadock’s Comprehensive
Textbook of Psychiatry, Vol 1. Edited by Sadock BJ, Sadock VA.
Philadelphia, PA, Lippincott Williams and Wilkins, 2005, pp 794–826
CHAPTER 5
Hematologic Tests
A complete blood cell count (CBC) is a standard part of any evaluation. It
screens for multiple problems, including infections and anemia. It also
provides a platelet count, a value important to monitor in psychiatric
medications associated with thrombocytopenia, such as divalproex sodium
or carbamazepine. This concern is particularly important in elderly patients,
because the risk of drug-induced thrombocytopenia may increase with age.
Lithium, in contrast, may result in mild leukocytosis. Because of the risk of
agranulocytosis, CBC testing is required weekly or biweekly for patients
taking clozapine and may be needed more frequently if the patient develops
signs of infection. Mirtazapine can also lead to agranulocytosis in rare
cases, and although routine CBC monitoring is not indicated, it should be
pursued if a patient develops sore throat, fever, stomatitis, or other signs of
infection.
Chemistry Tests
Most general chemistry panels have a variety of values that may be helpful
in medical evaluations. Blood glucose values may reveal hyperinsulinemia
and hypoglycemia, which may produce anxiety and weakness; more
commonly these tests show hyperglycemia, which may be associated with
diabetes and result in lethargy or, in severe cases, delirium, diabetic coma,
or ketoacidosis. This testing is critical for the diagnosis of diabetes, which
can be diagnosed with 1) an overnight fasting glucose greater than 126
mg/dL, 2) a random plasma glucose greater than 200 mg/dL with symptoms
of diabetes, or 3) an oral glucose tolerance test resulting in a plasma glucose
over 200 mg/dL 2 hours after a 75-g glucose load.
Kidney function tests are equally important. Blood urea nitrogen and
creatinine will be elevated in kidney failure and in hypovolemic states such
as dehydration. These tests also must be performed before initiating lithium
therapy because of lithium’s potential for nephrotoxicity.
General chemistry panels also measure serum sodium, potassium, and
other electrolytes. Hyponatremia—commonly defined as a serum sodium
concentration less than 135 mEq/L—has been reported with selective
serotonin reuptake inhibitors (SSRIs), particularly in the elderly. The signs
and symptoms of hyponatremia result from neurological dysfunction
secondary to cerebral edema. Acute hyponatremia can start with nausea and
malaise when the plasma sodium concentration falls below 125–130 mEq/L
and progresses rapidly to coma and respiratory arrest if the plasma sodium
concentration falls below 115–120 mEq/L. In chronic hyponatremia, the
brain cells adapt to the edema, and symptoms are much less severe. Patients
may be asymptomatic despite a plasma sodium concentration that is
persistently as low as 115–120 mEq/L. When symptoms do occur in
individuals with such low sodium concentrations, they are relatively
nonspecific (e.g., fatigue, nausea, dizziness, gait disturbances, forgetfulness,
confusion, lethargy, muscle cramps). The clinician should be vigilant to this
risk in older adults when they begin taking SSRIs. Of all the electrolyte
abnormalities, potassium disorders may be the most crucial to identify.
These rarely cause psychiatric symptoms but may result in severe cardiac
arrhythmias. Although not always included in routine chemistry screens,
calcium and magnesium levels are also important to consider, because
abnormal levels may result in paranoid ideation or frank psychosis. Any or
all of these results may be abnormal in patients undergoing hemodialysis.
Urinalysis
A urinalysis is an inexpensive, noninvasive test that provides a significant
amount of information. It determines the urine’s specific gravity, which
may indicate dehydration, and also tests for glucose and ketones, important
in the evaluation of diabetic patients. In the elderly population, the most
important use of urinalysis may be as a screening tool for urinary tract
infections (UTIs). A UTI is suggested when a microscopic examination
shows high levels of white blood cells, bacteria, positive leukocyte esterase
and nitrite, and possibly red blood cells; high numbers of epithelial cells
make the results difficult to interpret, because their presence suggests
contamination. A urine culture is a definitive means of diagnosing a UTI
and will identify the infecting organism and its susceptibility to
antimicrobial treatments. Approximately 20% of admissions from the
community to geropsychiatry units may have UTIs, and many cases of UTI
result in a delirium that improves with appropriate antibiotic treatment
(Levkoff et al. 1991; Manepalli et al. 1990).
Electrocardiogram
An electrocardiogram (ECG) provides a graphic representation of the
heart’s electrical activity, obtained via surface electrodes placed in specific
locations on the patient’s chest. This placement makes possible a graph of
electrical activity from a variety of spatial perspectives. In psychiatry, the
most important roles of the ECG include screening for cardiovascular
disease that may preclude the use of specific medications and monitoring
for drug-induced electrocardiographic changes either from standard doses
or from overdose. Electrocardiographic changes associated with specific
psychotropic medications are summarized in Table 5–3.
The TCAs are well known to be cardiotoxic in overdose; even at
therapeutic doses, their use is considered unsafe in patients with
cardiovascular disease, particularly ischemic disease. Although the most
common cardiovascular complication of TCAs is orthostatic hypotension,
TCAs have the same pharmacological properties as type IA
antiarrhythmics, such as quinidine and procainamide. TCAs slow
conduction at the bundle of His; individuals with preexisting bundle branch
block who take TCAs are at increased risk for atrioventricular block. Even
therapeutic levels are associated with prolonged PR intervals and QRS
complexes; these results may be more pronounced in elderly individuals
because the incidence and severity of adverse drug reactions increase with
age. If TCAs are used, baseline and frequent follow-up ECGs should be
obtained.
Imaging Studies
Plain film radiographs remain an integral piece of the diagnostic imaging
performed in geriatric psychiatry. Such techniques are most commonly used
to detect lung pathology that may contribute to mental status changes or to
detect bone fractures. Plain film radiographs are critical for individuals who
have both severe dementia and either a recent history of falls or newly
developed limb immobility.
A number of more recently developed imaging techniques have greatly
enhanced diagnostic abilities. These techniques are costly, so they should
not be used without a good rationale that includes why they are needed and
how the specific findings may affect a patient’s treatment plan. The
following discussion focuses on two commonly used structural imaging
techniques: computed tomography (CT) and MRI. Because these techniques
are also discussed in other chapters of this book, we focus on the scientific
basis behind these tools and provide information to support their clinical
use, particularly in brain imaging, and to facilitate providing informed
consent.
Other imaging techniques such as single-photon emission computed
tomography (SPECT) and functional MRI are used mostly in research and
have limited clinical use, and therefore are not discussed here. Amyloid
imaging agents involving positron emission tomography (PET) are also
used primarily in research, but because of the number of ongoing clinical
research trials being done to establish their utility, a brief discussion is
included later.
Computed Tomography
Computed tomography is a general term for several radiographic techniques
that result in the computer-assisted generation of a series of images showing
slices of an organ or body region, such as the brain or abdomen. The CT
scanner uses a small X-ray device that rotates around the body region of
interest in a fixed plane; these signals are sent to a computer that produces
the corresponding cross-sectional slice for that plane. The computer can
create sections in axial, coronal, and sagittal alignments. More recent
advances in software and display systems have led to many useful clinical
applications, including virtual CT colonoscopy or angiography.
When used to examine brain structure, CT can allow for the ready
identification of many structures, although it does have limitations. By
measuring differences in density, it can distinguish among CSF, blood,
bone, gray matter, and white matter. CT is particularly useful for
demonstrating bone abnormalities (such as skull fractures), areas of
hemorrhage (such as a subdural hematoma), and the mass effect from
various lesions. It can also display atrophy or ventricular enlargement.
However, CT is not very useful for visualizing posterior fossa or brain stem
structures because of surrounding bone.
A typical concern of patients is radiation exposure. CT scans require the
use of a limited amount of radiation; any given CT procedure results in a
radiation exposure, but that exposure is well below governmental
recommendations for individuals who work around radiation. However,
these recommendations do not consider multiple CT scans (thus multiple
radiation exposures) or CT studies that overlap scanned regions, a technique
that increases the radiation dose. CT imaging should be used when
appropriate, but other assessment techniques that may result in lower
radiation exposure should also be considered.
MRI has advantages and disadvantages when compared with CT. MRI
produces higher-resolution images and can obtain good detail in regions
(such as the posterior fossa) that are poorly visualized on CT. Additionally,
no radiation is involved in MRI. Unfortunately, the procedure is more
grueling than CT because the patient must remain motionless for a longer
period of time in a smaller, enclosed space. This may be difficult for
individuals who are claustrophobic. Additionally, the magnetic device must
be housed in an area devoid of iron, and staff and patients must not carry or
wear certain metals or have them embedded in their bodies. Moreover, MRI
tends to be more costly than CT imaging in most institutions.
In the psychiatric workup of a geriatric patient (Table 5–4), MRI should
be considered when the clinician suspects small lesions in regions difficult
to visualize—for example, to obtain evidence of midbrain hemorrhage in a
patient with suspected Wernicke’s encephalopathy, or to confirm a
suspected pituitary tumor in an individual with hyperprolactinemia, which
may be seen in association with risperidone and other high-potency
antipsychotic agents. Hyperprolactinemia carries the risks of osteopenia,
sexual dysfunction, amenorrhea, breast enlargement, and possibly cardiac
disease and breast cancer. Switching from high-potency to low-potency
antipsychotic drugs such as quetiapine or aripiprazole has been shown not
only to normalize prolactin but also in some instances to reverse menstrual
function or other symptoms (Shim et al. 2007). MRI can also easily identify
vascular pathology, including lacunar infarcts, and it is better than CT for
defining exact anatomical localization.
A limitation of CT and MRI is that neither can differentiate between
acute and chronic lesions. Diffusion-weighted imaging (DWI) overcomes
this difficulty. DWI is based on the capacity of fast MRI to detect a signal
related to the movement of water molecules between two closely spaced
radiofrequency pulses (diffusion). This technique can detect abnormalities
due to ischemia within 3–30 minutes of onset, whereas conventional MRI
and CT images would still appear normal. Therefore, DWI is helpful in
defining the clinically appropriate infarct when multiple subcortical infarcts
of various ages are present.
The American Academy of Neurology recommends routine use of
structural neuroimaging (noncontrast head CT or MRI) in the initial
evaluation of all patients with dementia (Knopman et al. 2001). Recent
studies also suggest novel uses for MRI in dementia. Schuff et al. (2009)
showed that in the Alzheimer’s Disease Neuroimaging Initiative cohort,
patients with MCI and AD show progressive hippocampal loss over 6
months and then accelerated loss over 1 year. NeuroQuant, an FDA-
approved software program, automatically quantifies volumes of brain
structures and may be useful for measuring progressive hippocampal loss in
patients. MRI measurements of cortical thickness, ventricular volume, and
mean diffusivity may also help to distinguish normal pressure
hydrocephalus from other neurodegenerative disorders (Ivkovic et al. 2013;
Moore et al. 2012).
Positron Emission Tomography Imaging
PET imaging has recently been incorporated into dementia workup, and
imaging agents that specifically bind amyloid or tau are being studied to
determine their clinical utility. The main risk of PET imaging is exposure
from radioactive imaging agents, which currently are mostly
18fluorodeoxyglucose (FDG) based and have a half-life of about 110
minutes. PET has two important advantages: high sensitivity and
quantification of distribution of the radioactive tracer; its main disadvantage
is the lack of spatial resolution. MRI has several important advantages:
high-resolution imaging of structures, excellent anatomical contrast for soft
tissue structures, and other useful measurements such as perfusion,
diffusivity, and spectroscopy. PET-MRI hybrid imaging combines the
advantages of each imaging modality but is currently limited by lack of
availability to many patients.
FDG-PET Imaging
FDG-PET imaging can be useful in distinguishing AD from frontotemporal
dementia (FTD) when the clinical diagnosis is unclear. On FDG-PET
imaging, AD causes hypometabolism predominantly in posterior
temporoparietal association and posterior cingulate cortices, whereas FTD
causes hypometabolism in the frontal lobes and anterior temporal and
anterior cingulate cortices. In one study, visual interpretation of FDG-PET
metabolic and statistical maps was superior to clinical assessment, with a
diagnostic accuracy of 89.6% (Foster et al. 2007). Medicare has approved
payment for FDG-PET imaging in patients who meet the criteria for both
AD and FTD and need clarification of their diagnosis.
Genetic Testing
Genetics in geriatric psychiatry is covered in more detail in Chapter 3,
“Genomics in Geriatric Psychiatry.” In this section, intended to serve as an
introduction to genetic testing, we briefly discuss a well-researched test
examining for APOE alleles, pharmacogenomics, and ethical and
psychological concerns related to genetic testing.
APOE Testing
Extensive research has attempted to identify genetic markers for AD.
Mutations on chromosomes 1, 14, and 21 have been linked to rare forms of
early-onset familial AD; such findings may help families make decisions
about pregnancies. One of the most studied genes for AD is APOE. This
gene encodes for an astrocyte-secreted plasma protein that is involved in
cholesterol transport. APOE may also play a role in the regeneration of
injured nerve tissue. There are three possible alleles (ε2, ε3, ε4) of the
APOE gene that may be combined in a heterozygous (ε2/ε3, ε2/ε4, ε3/ε4) or
homozygous (ε2/ε2, ε3/ε3, ε4/ε4) fashion.
Multiple epidemiological studies have documented that the presence of
the ε4 allele is a risk factor for AD. Additionally, the presence of ε4 alleles
increases the specificity of the diagnosis of AD. Despite these associations,
the presence of an ε4 allele, even a homozygous ε4/ε4 genotype, is not
diagnostic for AD. The APOE story may be even more complicated; Roses
et al. (2010) found that another factor, TOMM40 poly-T polymorphism,
may be linked to APOE and that together these may affect the mean age at
onset of AD. A clinical trial is under way to further validate this finding.
APOE testing is not currently recommended to predict dementia risk in
asymptomatic individuals. Arguments against routine testing include the
lack of an effective treatment to modify the disease course and the lack of
evidence that APOE status may influence current supportive treatments.
Pharmacogenomics
Most pharmacogenetic tests are designed to detect certain alleles that may
be correlated with psychotropic response or serious psychotropic-related
adverse events. The first FDA-approved pharmacogenetics test, the
AmpliChipR CYP450 Test made by Roche Diagnostics, includes the
assessment of 27 alleles in cytochrome P450, family 2, subfamily D,
polypeptide 6 gene CYP2D6 (Malhotra et al. 2012). This assessment of the
CYP2D6 allele is purportedly helpful for clinicians who are trying to
determine an appropriate starting dose for numerous psychotropic drugs
(including several SSRIs, TCAs, and antipsychotics) that are metabolized
by CYP2D6. Ultra-rapid metabolizers (about 1% of the population) may
require higher dosages to have therapeutic levels, whereas poor
metabolizers (about 7%–10% of Caucasians and 1% of Asians) may require
lower dosages to achieve similar levels. A number of studies have also
examined various alleles to try to detect those individuals who are at higher
risk for serious adverse events, such as metabolic syndrome, from taking
antipsychotics (Malhotra et al. 2012). Currently, based on the available
studies, the FDA recommends performing the HLA-B*1502 allele test for
carbamazepine-induced Stevens-Johnson syndrome only for individuals of
Asian descent.
Psychiatrists should ask themselves a few questions before ordering the
results of a pharmacogenetics test for clinical purposes. First, is the patient
from the specific population in which the test has been shown to be valid?
For example, the HLA-B*1502 allele test was validated in Asian but not
Caucasian populations. Second, what is the evidence for predictive power
of the test in the clinical (vs. the laboratory) setting? Demonstration of
biologically accurate results and quality control is an essential part of any
laboratory test but does not necessarily equate to clinical validity. Third,
what is the likelihood that the pharmacogenomic test will sufficiently
explain the observed phenotype? Although an allele may be associated with
a high risk of an adverse outcome, it may not explain the majority of cases
seen.
Additionally, as discussed in the next section, ethical and psychological
concerns must be considered. Does the patient fully appreciate the
implications of undergoing such tests in the clinical setting? For example, a
patient may agree to the release of test results to a health insurance
company or another third party, believing at the time that the test is
designed for a particular purpose (e.g., detection of risk of weight gain from
anti-psychotics). However, future studies may show that this allele is
associated with development of an unrelated disease process such as cancer,
and this discovery may have consequences that the patient did not anticipate
if proper counseling was not received. Therefore, collaboration with a
genetics counselor is essential before any genetic testing is performed in the
clinical—and arguably in the research—setting.
Omics Technologies
Since the inception of the Human Genome Project, high-throughput omics
technologies have shown immense promise to revolutionize medicine in the
decades to come. There is, however, an absence of widespread use and
acceptance of these tests in the clinical setting for various reasons,
including need for replication, need for demonstration of validity in the
general population, and costs. Nevertheless, given the high likelihood that
psychiatrists will have patients who will ask to have these tests performed
or will bring in results of these tests from elsewhere, it is important that
clinicians have a basic conceptual understanding of the general purpose and
challenges that may arise in the interpretation of these omics technologies.
Given the rapid development of this field, an exhaustive review is not
possible. Nevertheless, many of the same questions regarding the clinical
interpretation of pharmacogenetic tests (see earlier section
“Pharmacogenomics”) can also be applied to the other omics technologies.
The purpose of the omics technologies is to examine the changes that
can occur at different levels within an organism due to both physiological
and pathophysiological processes. Table 5–5 briefly describes various
omics technologies (Valdes et al. 2013). The overall concept is that these
technologies sample the process of how the organism is functioning from
the beginning product (the genome) to its end product (its metabolites), as
well as in between (epigenomics, transcriptomics, and proteomics). These
findings can be used to determine at which stage(s) critical differences or
modifications associated with certain types of outcomes (particularly in
diseases or adverse medication effects) may arise.
Conclusion
Laboratory testing can provide invaluable information in both the diagnosis
and the treatment of geriatric psychiatry patients. Clinicians should
carefully interpret laboratory values in the context of patients’ histories and
other available data. The role of neuroimaging, CSF biomarkers, and
various “omics” in clinical care is the focus of future research and will
revolutionize the care of the geriatric psychiatric patient. Future directions
for laboratory values include detection of populations who may be at risk
for certain psychiatric disorders (e.g., dementia) and prediction of which
individuals may respond or not respond to certain treatments for psychiatric
disorders.
Key Points
• Laboratory testing is an essential component of the psychiatric
evaluation for elderly individuals, who often present with comorbid
medical illnesses.
• Laboratory tests are also useful in monitoring medication side effects.
New guidelines have been proposed to monitor patients taking atypical
antipsychotics.
• Neuroimaging is useful in evaluation of a variety of neuropsychiatric
illnesses including, but not limited to, dementia.
• Genetic testing has great potential in geriatric psychiatry but currently
has limited clinical utility. Important ethical issues should be considered
when using genetic testing.
References
American Diabetes Association, American Psychiatric Association,
American Association of Clinical Endocrinologists, et al: Consensus
development conference on antipsychotic drugs and obesity and
diabetes. Diabetes Care 27(2):596–601, 2004 14747245
Blennow K, Hampel H, Weiner M, et al: Cerebrospinal fluid and plasma
biomarkers in Alzheimer disease. Nat Rev Neurol 6(3):131–144, 2010
20157306
Boushey CJ, Beresford SA, Omenn GS, et al: A quantitative assessment of
plasma homocysteine as a risk factor for vascular disease. Probable
benefits of increasing folic acid intakes. JAMA 274(13):1049–1057,
1995 7563456
Castro VM, Clements CC, Murphy SN, et al: QT interval and
antidepressant use: a cross sectional study of electronic health records.
BMJ 346:f288, 2013 23360890
Centers for Disease Control and Prevention: HIV Among Older Americans.
Page last updated December 20, 2013. Available at
http://www.cdc.gov/hiv/risk/age/olderamericans/. Accessed November
13, 2013.
Centers for Disease Control and Prevention: Reported STDs in the United
States. 2012 National Data for Chlamydia, Gonorrhea, and Syphilis
(CDC Fact Sheet). January 2014. Available at:
http://www.cdc.gov/nchhstp/newsroom/docs/std-trends-508.pdf.
Accessed November 9, 2014.
Chien DT, Szardenings AK, Bahri S, et al: Early clinical PET imaging
results with the novel PHF-tau radioligand [F18]-T808. J Alzheimers
Dis 38(1):171–184, 2014 23948934
Doraiswamy PM, Sperling RA, Coleman RE, et al: Amyloid-β assessed by
florbetapir F 18 PET and 18-month cognitive decline: a multicenter
study. Neurology 79(16):1636–1644, 2012 22786606
Durga J, van Boxtel MP, Schouten EG, et al: Effect of 3-year folic acid
supplementation on cognitive function in older adults in the FACIT
trial: a randomised, double blind, controlled trial. Lancet
369(9557):208–216, 2007 17240287
Finch N, Baker M, Crook R, et al: Plasma progranulin levels predict
progranulin mutation status in frontotemporal dementia patients and
asymptomatic family members. Brain 132(Pt 3):583–591, 2009
19158106
Forman MS, Farmer J, Johnson JK, et al: Frontotemporal dementia:
clinicopathological correlations. Ann Neurol 59(6):952–962, 2006
16718704
Foster NL, Heidebrink JL, Clark CM, et al: FDG-PET improves accuracy in
distinguishing frontotemporal dementia and Alzheimer’s disease. Brain
130(Pt 10): 2616–2635, 2007 17704526
Foulds PG, Davidson Y, Mishra M, et al: Plasma phosphorylated-TDP-43
protein levels correlate with brain pathology in frontotemporal lobar
degeneration. Acta Neuropathol 118(5):647–658, 2009 19823856
Grundman M, Pontecorvo MJ, Salloway SP, et al: Potential impact of
amyloid imaging on diagnosis and intended management in patients
with progressive cognitive decline. Alzheimer Dis Assoc Disord 27(1):
4–15, 2013 23203162
Grunwald M, Busse F, Hensel A, et al: Correlation between cortical theta
activity and hippocampal volumes in health, mild cognitive
impairment, and mild dementia. J Clin Neurophysiol 18(2):178–184,
2001 11435810
Hansson O, Zetterberg H, Buchhave P, et al: Association between CSF
biomarkers and incipient Alzheimer’s disease in patients with mild
cognitive impairment: a follow-up study. Lancet Neurol 5(3):228–234,
2006 16488378
Hsich G, Kenney K, Gibbs CJ, et al: The 14–3–3 brain protein in
cerebrospinal fluid as a marker for transmissible spongiform
encephalopathies. N Engl J Med 335(13): 924–930, 1996 8782499
Ivkovic M, Liu B, Ahmed F, et al: Differential diagnosis of normal pressure
hydrocephalus by MRI mean diffusivity histogram analysis. AJNR Am
J Neuroradiol 34(6):1168–1174, 2013 23257611
Jack CR Jr, Albert MS, Knopman DS, et al: Introduction to the
recommendations from the National Institute on Aging–Alzheimer’s
Association workgroups on diagnostic guidelines for Alzheimer’s
disease. Alzheimer’s and Dementia. The Journal of the Alzheimer’s
Association 7:257–262, 2011 21514247
Klee GG, Hay ID: Biochemical testing of thyroid function. Endocrinol
Metab Clin North Am 26(4):763–775, 1997 9429859
Knopman DS, DeKosky ST, Cummings JL, et al: Practice parameter:
diagnosis of dementia (an evidence-based review). Neurology
56(9):1143–1153, 2001 11342678
Kogan EA, Korczyn AD, Virchovsky RG, et al: EEG changes during long-
term treatment with donepezil in Alzheimer’s disease patients. J Neural
Transm 108(10):1167–1173, 2001 11725819
Kowalski JW, Gawel M, Pfeffer A, et al: The diagnostic value of EEG in
Alzheimer disease: correlation with the severity of mental impairment.
J Clin Neurophysiol 18(6):570–575, 2001 11779971
Lampert EJ, Roy Choudhury K, Hostage CA, et al: Prevalence of
Alzheimer’s pathologic endophenotypes in asymptomatic and mildly
impaired first-degree relatives. PLoS ONE 8(4):e60747, 2013
23613741
Leblhuber F, Walli J, Artner-Dworzak E, et al: Hyperhomocysteinemia in
dementia. J Neural Transm 107(12):1469–1474, 2000 11458999
Levkoff S, Cleary P, Liptzin B, et al: Epidemiology of delirium: an
overview of research issues and findings. Int Psychogeriatr 3(2):149–
167, 1991 1811770
Luchsinger JA, Tang MX, Miller J, et al: Relation of higher folate intake to
lower risk of Alzheimer disease in the elderly. Arch Neurol 64(1):86–
92, 2007 17210813
Maddalena A, Papassotiropoulos A, Müller-Tillmanns B, et al: Biochemical
diagnosis of Alzheimer disease by measuring the cerebrospinal fluid
ratio of phosphorylated tau protein to beta-amyloid peptide42. Arch
Neurol 60(9):1202–1206, 2003 12975284
Malhotra AK, Zhang JP, Lencz T: Pharmacogenetics in psychiatry:
translating research into clinical practice. Mol Psychiatry 17(8):760–
769, 2012 22083729
Manepalli J, Grossberg GT, Mueller C: Prevalence of delirium and urinary
tract infection in a psychogeriatric unit. J Geriatr Psychiatry Neurol
3(4):198–202, 1990 2073307
McMahon JA, Green TJ, Skeaff CM, et al: A controlled trial of
homocysteine lowering and cognitive performance. N Engl J Med
354(26):2764–2772, 2006 16807413
Meins W, Müller-Thomsen T, Meier-Baumgartner H-P: Subnormal serum
vitamin B12 and behavioural and psychological symptoms in
Alzheimer’s disease. Int J Geriatr Psychiatry 15(5):415–418, 2000
10822240
Moore DW, Kovanlikaya I, Heier LA, et al: A pilot study of quantitative
MRI measurements of ventricular volume and cortical atrophy for the
differential diagnosis of normal pressure hydrocephalus. Neurol Res Int
2012:6 pages, 2012 21860791
National Human Genome Research Institute: Genetic discrimination. Last
Updated July 31, 2014. Available at
http://www.genome.gov/10002077#al-3. Accessed on November 9,
2014.
National Institute on Aging: COGNISION as a Useful Cognitive Biomarker
for Alzheimer’s Disease. 2013. Available at:
www.nia.nih.gov/alzheimers/clinical-trials/cognisiontm-useful-
cognitive-biomarker-alzheimers-disease. Accessed November 13,
2013.
Neumann M, Sampathu DM, Kwong LK, et al: Ubiquitinated TDP-43 in
frontotemporal lobar degeneration and amyotrophic lateral sclerosis.
Science 314(5796):130–133, 2006 17023659
Nilsson K, Gustafson L, Hultberg B: Improvement of cognitive functions
after cobalamin/folate supplementation in elderly patients with
dementia and elevated plasma homocysteine. Int J Geriatr Psychiatry
16(6):609–614, 2001 11424170
Poser S, Mollenhauer B, Kraubeta A, et al: How to improve the clinical
diagnosis of Creutzfeldt-Jakob disease. Brain 122(Pt 12):2345–2351,
1999 10581227
Ravaglia G, Forti P, Maioli F, et al: Elevated plasma homocysteine levels in
centenarians are not associated with cognitive impairment. Mech
Ageing Dev 121(1–3): 251–261, 2000 11164478
Robins Wahlin TB, Wahlin A, Winblad B, et al: The influence of serum
vitamin B12 and folate status on cognitive functioning in very old age.
Biol Psychol 56(3):247–265, 2001 11399353
Roses AD, Lutz MW, Amrine-Madsen H, et al: A TOMM40 variable-length
polymorphism predicts the age of late-onset Alzheimer’s disease.
Pharmacogenomics J 10(5):375–384, 2010 20029386
Sabin TD: AIDS: the new “great imitator.” J Am Geriatr Soc 35(5):460–
464, 1987 3571794
Schuff N, Woerner N, Boreta L, et al: MRI of hippocampal volume loss in
early Alzheimer’s disease in relation to ApoE genotype and
biomarkers. Brain 132(Pt 4): 1067–1077, 2009 19251758
Selhub J, Bagley LC, Miller J, et al: B vitamins, homocysteine, and
neurocognitive function in the elderly. Am J Clin Nutr 71(2):614S–
620S, 2000 10681269
Shim JC, Shin JG, Kelly DL, et al: Adjunctive treatment with a dopamine
partial agonist, aripiprazole, for antipsychotic-induced
hyperprolactinemia: a placebo-controlled trial. Am J Psychiatry 164(9):
1404–1410, 2007 17728426
Steinhoff BJ, Räcker S, Herrendorf G, et al: Accuracy and reliability of
periodic sharp wave complexes in Creutzfeldt-Jakob disease. Arch
Neurol 53(2):162–166, 1996 8639066
Thomas SHL: Drugs, QT interval abnormalities and ventricular
arrhythmias. Adverse Drug React Toxicol Rev 13(2):77–102, 1994
7918900
U.S. Food and Drug Administration: Clinical Evaluation of QT/QTc
Interval Prolongation and Proarrhythmic Potential for Non-
Antiarrhythmic Drugs, 2005. Available at:
www.fda.gov/downloads/RegulatoryInformation/Guidances/ucm12935
7.pdf. Accessed November 13, 2013.
Valdes AM, Glass D, Spector TD: Omics technologies and the study of
human ageing. Nat Rev Genet 14(9):601–607, 2013 23938363
Zannas AS, Wong TZ, Doraiswamy PM: How much is a picture worth?
Putting amyloid imaging to the test. Dement Geriatr Cogn Dis Extra
2(1):649–651, 2012 23341830
Zivin K, Pfeiffer PN, Bohnert AS, et al: Evaluation of the FDA warning
against prescribing citalopram at doses exceeding 40 mg. Am J
Psychiatry 170(6):642–650, 2013 23640689
Suggested Readings
Knopman DS, DeKosky ST, Cummings JL, et al: Practice parameter:
diagnosis of dementia (an evidence-based review). Report of the
Quality Standards Subcommittee of the American Academy of
Neurology. Neurology 56:1143–1153, 2001
Townsend BA, Petrella, JR, Murali Doraiswamy P: The role of
neuroimaging in geriatric psychiatry. Curr Opin Psychiatry 15:427–
432, 2002
CHAPTER 6
Alzheimer’s Disease
The presentation of AD neurocognitive disorder is dominated by a
pronounced impairment in recent memory processing, which remains the
most affected area of mentation in the majority of cases. This difficulty is
now understood to arise from the selective involvement of the medial
temporal lobe early in the illness (Braak and Braak 1991; Hyman et al.
1984), giving rise to impaired consolidation of newly learned information
into more permanent memory stores located across interconnected
neocortical structures. On formal neuropsychological testing, the memory
problem of AD is manifest as a rapid forgetting of new information after
very brief delays of 5 minutes or more (Welsh et al. 1991). Patients in the
mild prodrome of the illness often show the characteristic memory disorder
of more fully expressed disease and may show other mild deficits in
executive function, language expression, visuoperception, and attention
(Bäckman et al. 2005; Hayden et al. 2005; Twamley et al. 2006). At this
early symptomatic stage, a diagnosis of mild cognitive impairment due to
AD can be made (Albert et al. 2011). A diagnosis of mild cognitive
impairment due to AD is synonymous with prodromal AD, a diagnostic
label used in Europe (Dubois et al. 2010), and with DSM-5 mild
neurocognitive disorder due to AD (American Psychiatric Association
2013).
As AD progresses, other areas of cognition become progressively more
involved, reflecting the specific spread of neuropathological involvement to
the lateral temporal areas, parietal cortex, and frontal neocortical areas
(Small et al. 2000; Storandt et al. 2006; Welsh et al. 1992). With the
advancement of symptomatic disease and as decline in independent
function becomes apparent, a diagnosis of major neurocognitive disorder
due to AD can be made (McKhann et al. 2011). At this stage, the
prototypical changes appear in expressive language, visuospatial function,
higher executive control, and semantic knowledge (Weintraub et al. 2012).
At later stages of the illness, anomia with impaired semantic fluency (e.g.,
generation of names of animals) is generally seen on examination. Word
search and circumlocution tendencies are common in conversational
speech, whereas speech comprehension itself is better preserved, as are all
other fundamental elements of communication (Weintraub et al. 2012).
Visuospatial problems become more prominent in later stages of illness,
resulting in dressing apraxia, difficulty in recognizing objects or people, and
problems in performing familiar motor acts (Cronin-Golomb and Amick
2001). Subtle problems in spatial processing and visual motion detection
can occur early and may be detectable only on formal examination
(Mapstone et al. 2003). Visuospatial problems can be illuminated by tests of
spatial judgment and visual organization (Rizzo et al. 2000). In everyday
settings, visuospatial problems may manifest as intermittent topographical
dis-orientation, leading to difficulties in finding familiar routes while
driving (Rizzo et al. 1997). Figure 6–1 compares the memory loss
characterizing AD and mild cognitive impairment with that characterizing
normal aging.
Cerebrovascular Disease
The neuropsychological profile of vascular dementia differs in many
respects from that of AD, the largest difference being the absence of the
profound memory impairment that is a hallmark of AD (Tierney et al.
2001). The presentation of vascular neurocognitive disorders will vary
according to the type and extent of the vascular compromise—multiple
infarctions, a single strategic stroke, microvascular disease, cerebral
hypoperfusion, hemorrhage, or combinations of these etiologies (Cohen et
al. 2002). Multi-infarct dementia, arising from multiple large- and small-
vessel strokes, will demonstrate a pattern of multifocal impairments on
testing that respect the cerebral territories involved by the infarctions (Chui
et al. 1992; Román et al. 1993). In disorders attributed to diffuse small-
vessel disease (e.g., Binswanger’s disease), the pattern shown on testing
reflects the disruption in the dorsolateral prefrontal and subcortical circuitry
(Kramer et al. 2002). Memory is involved, but the deficits are often patchy
in nature. Patients may show impaired recollection of some recent event but
show a surprising memory of some other occurrence transpiring within the
same time frame. On formal neuropsychological testing, the pattern on
memory testing is one of inefficient acquisition of new information leading
to a flattened learning curve over repeating trials (Looi and Sachdev 1999;
Padovani et al. 1995). Recall performance can be quite low, similar to that
seen in AD and mild cognitive impairment, but rapid forgetting is not a
typical feature (Hayden et al. 2005; Matsuda et al. 1998). The information
acquired, albeit limited, is generally retained, so that savings scores
between a final learning trial and a later delayed recall trial generally are
high. Finally, recognition improves dramatically with a recognition format,
suggesting a primary difficulty in retrieval rather than in storage or
consolidation of new information (Hayden et al. 2005). Beyond memory,
dysexecutive functions are typically involved, leading to slowed
sequencing, cognitive inflexibility, and decreased verbal fluency (Mathias
and Burke 2009). Asymmetries on sensory motor function or deficits in
coordination also are frequently demonstrated.
Conclusion
The neuropsychological evaluation provides a useful and cost-effective
management approach for diagnosis and management of the growing
geriatric population with memory complaints. A neuropsychological
evaluation is not needed in most major neurocognitive disorders in which
symptoms are obvious and the diagnosis is secure. It can be enormously
useful, however, in more complex, less straightforward diagnostic
situations, such as in early AD detection or in geriatric depression. By its
objective nature, the neuropsychological examination has strong
applications in medical management, providing information regarding
patient capacities and deficits that is important for choosing intervention
approaches and for guiding future decision making with respect to
competency and safety.
Key Points
• The neuropsychological presentation of Alzheimer’s disease is
characterized by a pronounced deficit in the consolidation of new
information from short-term, immediate memory to a more permanent
store. Thus, the deficit early in the disorder is a problem of rapid
forgetting of newly learned information.
• The profile of normal cognitive aging is characterized by modest
declines on executive function tests, in large measure because of
inefficiencies in multi-task processing and declines in perceptual motor
speed.
• Frontotemporal lobar degeneration is characterized by profound
functional and behavioral changes. The neurocognitive deficits
associated with the disorder, particularly in the early stages, may be
difficult to discern with mental status instruments. Neuropsychological
testing targeting executive functions can tease out the impairments in
behavioral regulation, disinhibition, perseveration, judgment, and
abstraction.
• The neuropsychological deficits associated with neurocognitive disorder
due to Parkinson’s disease are clinically very similar to those of a closely
aligned condition, neurocognitive disorder with Lewy bodies. However,
the neuropsychological profiles of these conditions can be distinguished
from that of neurocognitive disorder due to Alzheimer’s disease.
Visuospatial deficits are common early in neurocognitive disorder due
Parkinson’s disease and neurocognitive disorder with Lewy bodies, and
the memory disorders are less severe than those of Alzheimer’s disease.
• Geriatric depression can cause significant impairments in the efficiency
of cognitive processing, leading to selective problems in sustained
attention, concentration, and memory. It is a risk factor for cognitive
decline to dementia. When co-occurring with progressive neurological
disorders, such as Alzheimer’s disease or vascular neurocognitive
disorder, depression can lead to excess disability and an overall
reduction in the quality of life that might otherwise be achieved.
References
Aarsland D, Cummings JL, Larsen JP: Neuropsychiatric differences
between Parkinson’s disease with dementia and Alzheimer’s disease.
Int J Geriatr Psychiatry 16(2):184–191, 2001 11241724
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 work-groups on
diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement
7(3):270–279, 2011 21514249
Alexopoulos GS, Kiosses DN, Klimstra S, et al: Clinical presentation of the
“depression-executive dysfunction syndrome” of late life. Am J Geriatr
Psychiatry 10(1):98–106, 2002 11790640
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Attix DK, Welsh-Bohmer KA: Geriatric Neuropsychology: Assessment and
Intervention. Edited by Attix DK, Welsh-Bohmer KA. New York,
Guilford, 2006
Bäckman L, Jones S, Berger AK, et al: Cognitive impairment in preclinical
Alzheimer’s disease: a meta-analysis. Neuropsychology 19(4):520–
531, 2005 16060827
Ballard CG, Ayre G, O’Brien J, et al: Simple standardised
neuropsychological assessments aid in the differential diagnosis of
dementia with Lewy bodies from Alzheimer’s disease and vascular
dementia. Dement Geriatr Cogn Disord 10(2):104–108, 1999
10026383
Beats BC, Sahakian BJ, Levy R: Cognitive performance in tests sensitive to
frontal lobe dysfunction in the elderly depressed. Psychol Med
26(3):591–603, 1996 8733217
Beck AT, Steer RA: Beck Anxiety Inventory. San Antonio, TX,
Psychological Corporation, 1993
Beck AT, Steer RA, Brown GK: Beck Depression Inventory, II. San
Antonio, TX, Psychological Corporation, 1996
Benedict RHB: Brief Visuospatial Memory Test—Revised. Lutz, FL,
Psychological Assessment Resources, 1997
Benton AL, Hamsher K: Multilingual Aphasia Examination. Iowa City, IA,
AJA Associates, 1983
Benton AL, Eslinger PJ, Damasio AR: Normative observations on
neuropsychological test performances in old age. J Clin Neuropsychol
3(1):33–42, 1981 7276195
Benton AL, Hamsher K, Varney NR, et al: Contributions to
Neuropsychological Assessment. New York, Oxford University Press,
1983
Berg EA: A simple objective technique for measuring flexibility in
thinking. J Gen Psychol 39:15–22, 1948 18889466
Boone KB, Lesser I, Miller B, et al: Cognitive functioning in a mildly to
moderately depressed geriatric sample: relationship to chronological
age. J Neuropsychiatry Clin Neurosci 6(3):267–272, 1994 7950350
Boone KB, Lesser I, Miller B, et al: Cognitive functioning in older
depressed outpatients: relationship of presence and severity of
depression on neuropsychological test scores. Neuropsychology 9:390–
398, 1995
Botwinick J: Intellectual abilities, in The Handbook of the Psychology of
Aging. Edited by Birren JE, Schaie KW. New York, Van Nostrand
Reinhold, 1977, pp 508–605
Braak H, Braak E: Neuropathological stageing of Alzheimer-related
changes. Acta Neuropathol 82(4):239–259, 1991 1759558
Brandt J, Benedict RHB: Hopkins Verbal Learning Test—Revised. Lutz,
FL, Psychological Assessment Resources, 2001
Breitner JCS: Dementia—epidemiological considerations, nomenclature,
and a tacit consensus definition. J Geriatr Psychiatry Neurol 19(3):129–
136, 2006 16880354
Breitner JCS, Welsh KA: Diagnosis and management of memory loss and
cognitive disorders among elderly persons. Psychiatr Serv 46:29–35,
1995 7895118
Brun A, Englund B, Gustafson L, et al: Consensus statement: clinical and
neuropathological criteria for frontotemporal dementia: the Lund and
Manchester groups. J Neurol Neurosurg Psychiatry 57(4):416–418,
1994 8163988
Butters MA, Becker JT, Nebes RD, et al: Changes in cognitive functioning
following treatment of late-life depression. Am J Psychiatry
157(12):1949–1954, 2000 11097959
Butters MA, Young JB, Lopez O, et al: Pathways linking late-life
depression to persistent cognitive impairment and dementia. Dialogues
Clin Neurosci 10(3):345–357, 2008 18979948
Cahn-Weiner DA, Grace J, Ott BR, et al: Cognitive and behavioral features
discriminate between Alzheimer’s and Parkinson’s disease.
Neuropsychiatry Neuropsychol Behav Neurol 15(2):79–87, 2002
12050470
Cairns NJ, Bigio EH, Mackenzie IR, et al: Neuropathologic diagnostic and
nosologic criteria for frontotemporal lobar degeneration: consensus of
the Consortium for Frontotemporal Lobar Degeneration. Acta
Neuropathol 114(1):5–22, 2007 17579875
Chui HC, Victoroff JI, Margolin D, et al: Criteria for the diagnosis of
ischemic vascular dementia proposed by the State of California
Alzheimer’s Disease Diagnostic and Treatment Centers. Neurology
42(3 Pt 1):473–480, 1992 1549205
Coffey CE, Wilkinson WE, Parashos IA, et al: Quantitative cerebral
anatomy of the aging human brain: a cross-sectional study using
magnetic resonance imaging. Neurology 42(3 Pt 1):527–536, 1992
1549213
Cohen OS, Vakil E, Tanne D, et al: The Frontal Assessment Battery as a
tool for evaluation of frontal lobe dysfunction in patients with
Parkinson’s disease. J Geriatr Psychiatry Neurol 25(2):71–77, 2012
22689698
Cohen RA, Paul RH, Ott BR, et al: The relationship of subcortical MRI
hyperintensities and brain volume to cognitive function in vascular
dementia. J Int Neuropsychol Soc 8(6):743–752, 2002 12240738
Craik FIM, Rose NS: Memory encoding and aging: a neurocognitive
perspective. Neurosci Biobehav Rev 36(7):1729–1739, 2012 22155274
Cronin-Golomb A, Amick M: Spatial abilities in aging, Alzheimer’s
disease, and Parkinson’s disease, in Handbook of Neuropsychology,
Vol 6: Aging and Dementia, 2nd Edition. Edited by Boller F, Cappa S.
Amsterdam, Elsevier, 2001, pp 119–143
Daigneault S, Braun CM: Working memory and the Self-Ordered Pointing
Task: further evidence of early prefrontal decline in normal aging. J
Clin Exp Neuropsychol 15(6):881–895, 1993 8120125
D’Elia LF, Satz P, Uchiyama CL, et al: Color Trails Test. Lutz, FL,
Psychological Assessment Resources, 1996
Delis DC, Kramer JH, Kaplan E, et al: California Verbal Learning Test,
Adult Version, 2nd Edition. San Antonio, TX, Psychological
Corporation, 2000
Diniz BS, Butters MA, Albert SM, et al: Late-life depression and risk of
vascular dementia and Alzheimer’s disease: systematic review and
meta-analysis of community-based cohort studies. Br J Psychiatry
202(5):329–335, 2013 23637108
Drachman DA: Aging of the brain, entropy, and Alzheimer disease.
Neurology 67(8): 1340–1352, 2006 17060558
Dubois B, Feldman HH, Jacova C, et al: Revising the definition of
Alzheimer’s disease: a new lexicon. Lancet Neurol 9(11):1118–1127,
2010 20934914
Ebly EM, Parhad IM, Hogan DB, Fung TS: Prevalence and types of
dementia in the very old: results from the Canadian Study of Health
and Aging. Neurology 44(9):1593–1600, 1994 7936280
Eslinger PJ, Damasio AR, Benton AL, Van Allen M: Neuropsychologic
detection of abnormal mental decline in older persons. JAMA
253(5):670–674, 1985 3968802
Finch CE: The neurobiology of middle-age has arrived. Neurobiol Aging
30(4):515–520, discussion 530–533, 2009 19231030
Finkel D, Reynolds CA, McArdle JJ, et al: Age changes in processing speed
as a leading indicator of cognitive aging. Psychol Aging 22(3):558–
568, 2007 17874954
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Gascón-Bayarri J, Reñé R, Del Barrio JL, et al: Prevalence of dementia
subtypes in El Prat de Llobregat, Catalonia, Spain: the PRATICON
study. Neuroepidemiology 28(4):224–234, 2007 17878737
Geerlings MI, Jonker C, Bouter LM, et al: Association between memory
complaints and incident Alzheimer’s disease in elderly people with
normal baseline cognition. Am J Psychiatry 156(4):531–537, 1999
10200730
Geser F, Wenning GK, Poewe W, McKeith I: How to diagnose dementia
with Lewy bodies: state of the art. Mov Disord 20(suppl 12):S11–S20,
2005 16092075
Golden CJ, Freshwater SM: Stroop Color and Word Test: Revised
Examiners Manual. Woodale, IL, Stoeling, 2002
Gollan TH, Weissberger GH, Runnqvist E, et al: Self-ratings of spoken
language dominance: a Multilingual Naming Test (MINT) and
preliminary norms for young and aging Spanish-English bilinguals.
Bilingualism Language and Cognition 15(3):594–615, 2012
Gomar JJ, Bobes-Bascaran MT, Conejero-Goldberg C, et al: Utility of
combinations of biomarkers, cognitive markers, and risk factors to
predict conversion from mild cognitive impairment to Alzheimer
disease in patients in the Alzheimer’s Disease Neuroimaging Initiative.
Arch Gen Psychiatry 68(9):961–969, 2011 21893661
Gur RC, Gur RE, Obrist WD, et al: Age and regional cerebral blood flow at
rest and during cognitive activity. Arch Gen Psychiatry 44(7):617–621,
1987 3606327
Hamilton JM, Salmon DP, Galasko D, et al: A comparison of episodic
memory deficits in neuropathologically confirmed dementia with Lewy
bodies and Alzheimer’s disease. J Int Neuropsychol Soc 10(5):689–
697, 2004 15327716
Hanson L, Salmon D, Galasko D, et al: The Lewy body variant of
Alzheimer’s disease: a clinical and pathologic entity. Neurology
40(1):1–8, 1990 2153271
Hart RP, Kwentus JA, Taylor JR, Harkins SW: Rate of forgetting in
dementia and depression. J Consult Clin Psychol 55(1): 101–105, 1987
3571646
Hathaway SR, McKinley JC: Minnesota Multiphasic Personality Inventory:
Manual (Revised). San Antonio, TX, Psychological Corporation, 1951
Haug H, Barmwater U, Eggers R, et al: Anatomical changes in aging brain:
morphometric analysis of the human proscencephalon, in
Neuropharmacology, Vol 21: Aging. Edited by Cervos-Navarro J,
Sarkander HI. New York, Raven, 1983, pp 1–12
Hayden KM, Warren LH, Pieper CF, et al: Identification of VaD and AD
prodromes: the Cache County Study. Alzheimers Dement 1(1):19–29,
2005 19595812
Heaton RK, Grant I, Matthews CG: Comprehensive Norms for an
Expanded Halstead-Reitan Battery: Demographic Corrections,
Research Findings, and Clinical Applications. Odessa, FL,
Psychological Assessment Resources, 1991
Hebert LE, Weuve J, Scherr PA, et al: Alzheimer disease in the United
States (2010-2050) estimated using the 2010 census. Neurology
80(19):1778–1783, 2013 23390181
Heyman A, Fillenbaum GG, Gearing M, et al: Comparison of Lewy body
variant of Alzheimer’s disease with pure Alzheimer’s disease:
Consortium to Establish a Registry for Alzheimer’s Disease, Part XIX.
Neurology 52(9):1839–1844, 1999 10371532
Holman RC, Khan AS, Kent J, et al: Epidemiology of Creutzfeldt-Jakob
disease in the United States, 1979–1990: analysis of national mortality
data. Neuroepidemiology 14(4):174–181, 1995 7643951
Horn J: The theory of fluid and crystallized intelligence in relation to
concepts of cognitive psychology and aging in adulthood, in Aging and
Cognitive Processes. Edited by Craik F, Trehub S. New York, Plenum,
1982, pp 237–278
Howieson DB, Holm LA, Kaye JA, et al: Neurologic function in the
optimally healthy oldest old: neuropsychological evaluation.
Neurology 43(10):1882–1886, 1993 8413942
Hughes CP, Berg L, Danziger WL, et al: A new clinical scale for the staging
of dementia. Br J Psychiatry 140:566–572, 1982 7104545
Hyman BT, Van Hoesen GW, Damasio AR, et al: Alzheimer’s disease: cell-
specific pathology isolates the hippocampal formation. Science
225(4667):1168–1170, 1984 6474172
Ivnik RJ, Malec JF, Smith GE, et al: Mayo’s older Americans normative
studies: updated AVLT norms for ages 56–97. Clin Neuropsychol 6:83–
104, 1992
Jackson M, Lowe J: The new neuropathology of degenerative
frontotemporal dementias. Acta Neuropathol 91(2):127–134, 1996
8787144
Johnson JK, Diehl J, Mendez MF, et al: Frontotemporal lobar degeneration:
demographic characteristics of 353 patients. Arch Neurol 62(6):925–
930, 2005 15956163
Kaplan EF, Goodglass H, Weintraub S: The Boston Naming Test, 2nd
Edition. Philadelphia, PA, Lea and Febiger, 1978
Knopman DS, DeKosky ST, Cummings JL, et al: Practice parameter:
diagnosis of dementia (an evidence-based review). Neurology
56(9):1143–1153, 2001 11342678
Koltai DC, Welsh-Bohmer KA: Geriatric neuropsychological assessment, in
Clinician’s Guide to Neuropsychological Assessment, 2nd Edition.
Edited by Vanderploeg RD. Mahwah, NJ, Erlbaum Associates, 2000,
pp 383–415
Kramer JH, Reed BR, Mungas D, et al: Executive dysfunction in
subcortical ischaemic vascular disease. J Neurol Neurosurg Psychiatry
72(2):217–220, 2002 11796772
Krishnan KR: Depression as a contributing factor in cerebrovascular
disease. Am Heart J 140(4 suppl):70–76, 2000 11011351
Langley LK, Madden DJ: Functional neuroimaging of memory:
implications for cognitive aging. Microsc Res Tech 51(1):75–84, 2000
11002355
Lezak MD, Howieson DB, Biegler ED, et al: Neuropsychological
Assessment, 5th Edition. New York, Oxford University Press, 2012
Lippa CF, Duda JE, Grossman M, et al: DLB and PDD boundary issues:
diagnosis, treatment, molecular pathology, and biomarkers. Neurology
68(11):812–819, 2007 17353469
Litvan I, Goldman JG, Tröster AI, et al: Diagnostic criteria for mild
cognitive impairment in Parkinson’s disease: Movement Disorder
Society Task Force guidelines. Mov Disord 27(3):349–356, 2012
22275317
Lobo A, Launer LJ, Fratiglioni L, et al: Prevalence of dementia and major
subtypes in Europe: a collaborative study of population-based cohorts.
Neurology 54(11 suppl 5):S4–S9, 2000 10854354
Lockwood KA, Alexopoulos GS, van Gorp WG: Executive dysfunction in
geriatric depression. Am J Psychiatry 159(7):1119–1126, 2002
12091189
Looi JC, Sachdev PS: Differentiation of vascular dementia from AD on
neuropsychological tests. Neurology 53(4):670–678, 1999 10489025
Lyketsos CG, Sheppard JM, Steinberg M, et al: Neuropsychiatric
disturbance in Alzheimer’s disease clusters into three groups: the
Cache County study. Int J Geriatr Psychiatry 16(11):1043–1053, 2001
11746650
Mapstone M, Steffenella TM, Duffy CJ: A visuospatial variant of mild
cognitive impairment: getting lost between aging and AD. Neurology
60(5):802–808, 2003 12629237
Mathias JL, Burke J: Cognitive functioning in Alzheimer’s and vascular
dementia: a meta-analysis. Neuropsychology 23(4): 411–423, 2009
19586206
Matsuda O, Saito M, Sugishita M: Cognitive deficits of mild dementia: a
comparison between dementia of the Alzheimer’s type and vascular
dementia. Psychiatry Clin Neurosci 52(1):87–91, 1998 9682939
Mattis S, Jurica PJ, Leitten CL: Dementia Rating Scale-2TM (DRS-2TM).
Professional Manual. Lutz, FL, Psychological Assessment Resources,
2002
McKeith IG, Dickson DW, Lowe J, et al: Diagnosis and management of
dementia with Lewy bodies: third report of the DLB Consortium.
Neurology 65(12):1863–1872, 2005 16237129
McKhann GM, Knopman DS, Chertkow H, et al: The diagnosis of
dementia 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
7(3):263–269, 2011 21514250
Migliorelli R, Tesón A, Sabe L, et al: Prevalence and correlates of
dysthymia and major depression among patients with Alzheimer’s
disease. Am J Psychiatry 152(1):37–44, 1995 7802118
Mohs RC, Cohen L: Alzheimer’s Disease Assessment Scale (ADAS).
Psychopharmacol Bull 24(4):627–628, 1988 3249763
Monza D, Soliveri P, Radice D, et al: Cognitive dysfunction and impaired
organization of complex motility in degenerative parkinsonian
syndromes. Arch Neurol 55(3):372–378, 1998 9520011
Morris JC, Heyman A, Mohs RC, et al: The Consortium to Establish a
Registry for Alzheimer’s Disease (CERAD), Part I: clinical and
neuropsychological assessment of Alzheimer’s disease. Neurology
39(9):1159–1165, 1989 2771064
Osterrieth PA: Le test de copie d’une figure complexe. Archives de
Psychologie 30:206–356, 1944
Pachana NA, Boone KB, Miller BL, et al: Comparison of
neuropsychological functioning in Alzheimer’s disease and
frontotemporal dementia. J Int Neuropsychol Soc 2(6):505–510, 1996
9375154
Padovani A, Di Piero V, Bragoni M, et al: Patterns of neuropsychological
impairment in mild dementia: a comparison between Alzheimer’s
disease and multi-infarct dementia. Acta Neurol Scand 92(6):433–442,
1995 8750107
Park DC, Schwarz N: Cognitive Aging: A Primer. Philadelphia, PA,
Psychology Press, 1999
Pillon B, Dubois B, Agid Y: Severity and specificity of cognitive
impairment in Alzheimer’s, Huntington’s, and Parkinson’s diseases and
progressive supranuclear palsy. Ann N Y Acad Sci 640:224–227, 1991
1837977
Potter GG, Attix DK: An integrated model for geriatric neuropsychological
assessment, in Geriatric Neuropsychology: Assessment and
Intervention. Edited by Attix DK, Welsh-Bohmer KA. New York,
Guilford, 2006, pp 5–26
Rankin KP, Baldwin E, Pace-Savitsky C, et al: Self awareness and
personality change in dementia. J Neurol Neurosurg Psychiatry
76(5):632–639, 2005 15834018
Reichman WE, Coyne AC: Depressive symptoms in Alzheimer’s disease
and multi-infarct dementia. J Geriatr Psychiatry Neurol 8(2):96–99,
1995 7794481
Reid WG, Hely MA, Morris JG, et al: A longitudinal of Parkinson’s
disease: clinical and neuropsychological correlates of dementia. J Clin
Neurosci 3(4):327–333, 1996 18638897
Reitan RM: Validity of the Trail Making Test as an indicator of organic
brain damage. Percept Mot Skills 8:271–276, 1958
Reitan RM, Wolfson D: The Halstead-Reitan neuropsychological test
battery for adults: theoretical, methodological, and validational bases,
in Comprehensive Handbook of Psychological Assessment, Vol 1:
Intellectual and Neuropsychological Assessment. Edited by Goldstein
G, Beers S. Hoboken, NJ, Wiley, 2004, pp 105–131
Reuter-Lorenz PA, Park DC: Human neuroscience and the aging mind: a
new look at old problems. J Gerontol B Psychol Sci Soc Sci 65(4):405–
415, 2010 20478901
Rizzo M, Reinach S, McGehee D, et al: Simulated car crashes and crash
predictors in drivers with Alzheimer disease. Arch Neurol 54(5):545–
551, 1997 9152111
Rizzo M, Anderson SW, Dawson J, et al: Vision and cognition in
Alzheimer’s disease. Neuropsychologia 38(8):1157–1169, 2000
10838150
Román GC: Vascular dementia: distinguishing characteristics, treatment,
and prevention. J Am Geriatr Soc 51(5 suppl Dementia):S296–S304,
2003 12801386
Román GC, Tatemichi TK, Erkinjuntti T, et al: Vascular dementia:
diagnostic criteria for research studies. Report of the NINDS-AIREN
International Workshop. Neurology 43(2):250–260, 1993 8094895
Salmon DP, Galasko D, Hansen LA, et al: Neuropsychological deficits
associated with diffuse Lewy body disease. Brain Cogn 31(2):148–165,
1996 8811990
Salmon E, Perani D, Collette F, et al: A comparison of unawareness in
frontotemporal dementia and Alzheimer’s disease. J Neurol Neurosurg
Psychiatry 79(2):176–179, 2008 17898032
Salthouse TA: The processing-speed theory of adult age differences in
cognition. Psychol Rev 103(3):403–428, 1996 8759042
Salthouse TA: Relations between cognitive abilities and measures of
executive functioning. Neuropsychology 19(4):532–545, 2005
16060828
Salthouse TA: When does age-related cognitive decline begin? Neurobiol
Aging 30(4):507–514, 2009 19231028
Salthouse TA: Selective review of cognitive aging. J Int Neuropsychol Soc
16(5):754–760, 2010 20673381
Samson RD, Barnes CA: Impact of aging brain circuits on cognition. Eur J
Neurosci 37(12):1903–1915, 2013 23773059
Savolainen S, Paljärvi L, Vapalahti M: Prevalence of Alzheimer’s disease in
patients investigated for presumed normal pressure hydrocephalus: a
clinical and neuropathological study. Acta Neurochir (Wien)
141(8):849–853, 1999 10536721
Seeley WW, Bauer AM, Miller BL, et al: The natural history of temporal
variant frontotemporal dementia. Neurology 64(8): 1384–1390, 2005
15851728
Seltman RE, Matthews BR: Frontotemporal lobar degeneration:
epidemiology, pathology, diagnosis and management. CNS Drugs
26(10):841–870, 2012 22950490
Small BJ, Fratiglioni L, Viitanen M, et al: The course of cognitive
impairment in preclinical Alzheimer disease: three- and 6-year follow-
up of a population-based sample. Arch Neurol 57(6):839–844, 2000
10867781
Smith A: The Symbol Digit Modalities Test: a neuropsychologic test for
economic screening of learning and other cerebral disorders. Learning
Disorders 3:83–91, 1968
Soliveri P, Monza D, Paridi D, et al: Neuropsychological follow up in
patients with Parkinson’s disease, striatonigral degeneration-type
multisystem atrophy, and progressive supranuclear palsy. J Neurol
Neurosurg Psychiatry 69(3):313–318, 2000 10945805
Sperling RA, Aisen PS, Beckett LA, et al: Toward defining the preclinical
stages of Alzheimer’s disease: recommendations from the National
Institute on Aging–Alzheimer’s Association workgroups on diagnostic
guidelines for Alzheimer’s disease. Alzheimers Dement 7(3):280–292,
2011 21514248
Steffens DC, Otey E, Alexopoulos GS, et al: Perspectives on depression,
mild cognitive impairment, and cognitive decline. Arch Gen Psychiatry
63(2):130–138, 2006 16461855
Steinberg B, Bieliauskas L: Introduction to the special edition: IQ-based
MOANS norms for multiple neuropsychological instruments. Clin
Neuropsychol 19(3–4):277–279, 2005 16120533
Stern RA, White T: Neuropsychological Assessment Battery (NAB). Lutz,
FL, Psychological Assessment Resources, 2003
Storandt M, Grant EA, Miller JP, Morris JC: Longitudinal course and
neuropathologic outcomes in original vs revised MCI and in pre-MCI.
Neurology 67(3):467–473, 2006 16894109
Strauss E, Sherman EM, Spreen O: A Compendium of Neuropsychological
Tests: Administration, Norms, and Commentary, 3rd Edition. New
York, Oxford University Press, 2006
Tariot PN: CERAD behavior rating scale for dementia. Int Psychogeriatr 8
(suppl 3): 317–320, discussion 351–354, 1996 9154581
Testa D, Monza D, Ferrarini M, et al: Comparison of natural histories of
progressive supranuclear palsy and multiple system atrophy. Neurol Sci
22(3):247–251, 2001 11731878
Tierney MC, Black SE, Szalai JP, et al: Recognition memory and verbal
fluency differentiate probable Alzheimer disease from subcortical
ischemic vascular dementia. Arch Neurol 58(10):1654–1659, 2001
11594925
Tisserand DJ: Structural and Functional Changes Underlying Cognitive
Aging. Maastricht, The Netherlands, Maastricht University, 2003
Tröster AI: Neuropsychological characteristics of dementia with Lewy
bodies and Parkinson’s disease with dementia: differentiation, early
detection, and implications for “mild cognitive impairment” and
biomarkers. Neuropsychol Rev 18(1):103–119, 2008 18322801
Tschanz JT, Welsh-Bohmer KA, West N, et al: Identification of dementia
cases derived from a neuropsychological algorithm: comparisons with
clinically derived diagnoses. Neurology 54(6):1290–1296, 2000
10746600
Twamley EW, Ropacki SA, Bondi MW: Neuropsychological and
neuroimaging changes in preclinical Alzheimer’s disease. J Int
Neuropsychol Soc 12(5):707–735, 2006 16961952
van Hooren SA, Valentijn AM, Bosma H, et al: Cognitive functioning in
healthy older adults aged 64–81: a cohort study into the effects of age,
sex, and education. Neuropsychol Dev Cogn B Aging Neuropsychol
Cogn 14(1):40–54, 2007 17164189
Warren JD, Rohrer JD, Rossor MN: Clinical review: frontotemporal
dementia. BMJ 347:14827 2013 23920254
Wechsler D: Wechsler Intelligence Scale, 4th Edition. San Antonio, TX,
Psychological Corporation, 2009a
Wechsler D: Wechsler Memory Scale, 4thd Edition. San Antonio, TX,
Psychological Corporation, 2009b
Weintraub S, Wicklund AH, Salmon DP: The neuropsychological profile of
Alzheimer disease. Cold Spring Harb Perspect Med 2(4):a006171,
2012 22474609
Welsh K, Butters N, Hughes J, et al: Detection of abnormal memory decline
in mild cases of Alzheimer’s disease using CERAD
neuropsychological measures. Arch Neurol 48(3):278–281, 1991
2001185
Welsh KA, Butters N, Hughes JP, et al: Detection and staging of dementia
in Alzheimer’s disease. Use of the neuropsychological measures
developed for the Consortium to Establish a Registry for Alzheimer’s
Disease. Arch Neurol 49(5): 448–452, 1992 1580805
Welsh-Bohmer KA, Mohs RC: Neuropsychological assessment of
Alzheimer’s disease. Neurology 49(3 suppl 3):S11–S13, 1997 9310507
Welsh-Bohmer KA, Warren LH: Neurodegenerative dementias, in Geriatric
Neuropsychology: Assessment and Intervention. Edited by Attix DK,
Welsh-Bohmer KA. New York, Guilford, 2006, pp 56–88
Welsh-Bohmer KA, Koltai DC, Mason DJ: The clinical utility of
neuropsychological evaluation of patients with known or suspected
dementia, in Demonstrating Utility and Cost Effectiveness in Clinical
Neuropsychology. Edited by Prigatano G, Pliskin N. Philadelphia, PA,
Psychology Press-Taylor and Francis Group, 2003, pp 177–200
Wetzel L, Boll TJ: Short Category Test, Booklet Format. Los Angeles, CA,
Western Psychological Services, 1987
Williams-Gray CH, Foltynie T, Brayne CE, et al: Evolution of cognitive
dysfunction in an incident Parkinson’s disease cohort. Brain 130(Pt
7):1787–1798, 2007 17535834
Yesavage J, Brink TL, Rose TL, et al: Development and validation of a
geriatric depression scale: a preliminary report. J Psychiatr Res
17(1):37–49, 1983 7183759
Zaccai J, McCracken C, Brayne C: A systematic review of prevalence and
incidence studies of dementia with Lewy bodies. Age Ageing
34(6):561–566, 2005 16267179
Suggested Readings
Attix DK, Welsh-Bohmer KA (eds): Geriatric Neuropsychology:
Assessment and Intervention. New York, Guilford, 2006
Strauss E, Sherman EM, Spreen O: A Compendium of Neuropsychological
Tests: Administration, Norms, and Commentary, 3rd Edition. New
York, Oxford University Press, 2006
Tröster AI, Woods SP: Neuropsychological aspects, in Handbook of
Parkinson’s Disease, 4th Edition. Edited by Pahwa R, Lyons KE. New
York, Informa, 2007, pp 109–131
PART III
Delirium
Jane S. Saczynski, Ph.D.
Sharon K. Inouye, M.D., M.P.H.
Epidemiology
Delirium is often the only sign of an acute and serious medical condition
affecting a patient, and it most commonly occurs in frail older persons with
multiple comorbidities. Table 7–1 presents the prevalence rates (present on
admission) and incidence rates (new onset) of delirium in various patient
populations. The highest incidence rates were observed in the intensive care
unit (ICU) and postoperative and palliative care settings. Although high,
these rates likely represent underestimates of true incidence rates, because
many studies of delirium exclude patients with cognitive impairment or
dementia at baseline. In general medical and geriatric wards, the rates of
prevalence of delirium (present on admission) must be added to the
incidence rates to yield the overall occurrence rates of delirium in these
populations of 29%–64% (see Table 7–1). The prevalence of delirium in the
community setting is relatively low (1%–2%), but its onset usually brings
the patient to emergency care (Inouye et al. 2014b). On presentation to the
emergency department, delirium is present in 8%–17% of all seniors and in
40% of nursing home residents (Inouye et al. 2014b).
Delirium is consistently associated with higher mortality rates across all
nonsurgical populations, including general medical, geriatric, intensive
care, stroke, dementia, nursing home, and emergency department (see Table
7–1). Delirium in the ICU is associated with a 2- to 4-fold increased risk of
death both in and out of the hospital and with cognitive impairment for up
to 12 months after discharge; patients who develop delirium while on
general medical or geriatric wards are at 1.5-fold increased risk for death in
the year following hospitalization; and delirium in the emergency
department is associated with a 70% increased risk of death during the first
6 months after the visit (Inouye et al. 2014b; Pandharipande et al. 2013).
Postoperative cognitive impairment is common among surgical patients
who develop delirium, with impairments lasting up to 1 year
postoperatively, and physical function is impaired for 30 days or more after
discharge among surgical and nonsurgical patients who develop delirium
(Rudolph et al. 2009; Saczynski et al. 2012). Prevalent delirium at
admission to post-acute care is associated with a 5-fold increased risk of 6-
month mortality (Marcantonio et al. 2005).
20%–22%
Emergency department 8%–17% Mortality (RR=1.7)
—
Note. LOS=length of stay; RR=relative risk.
*The sum of both prevalence (present on admission) and incidence (new onset) yields the overall
occurrence rates of delirium in each setting.
†Adjusted relative risks were derived from studies that provided adjustment for at least one
covariable.
Source. Reprinted (with modifications) from Inouye SK, Westendorp RG, Saczynski JS, et al:
“Delirium in Elderly People.” The Lancet 383(9920):911–922, 2014 with permission from Elsevier.
No specific laboratory tests currently exist that will aid in the definitive
identification of delirium. The laboratory evaluation for delirium is
intended to identify contributing factors that will need to be addressed, and
the approach should be guided by astute clinical judgment and tailored to
the individual situation. Laboratory tests that should be considered in the
delirium evaluation include complete blood count, electrolytes, kidney and
liver function, oxygen saturation, and glucose levels. Evaluation of occult
infection can be obtained through blood cultures, urinalysis, and urine
culture. Other laboratory tests, such as thyroid function, arterial blood gas,
vitamin B12 level, cortisol level, drug levels, toxicology screen, and
ammonia levels, may be helpful in identifying factors that contribute to
delirium. An electrocardiogram and/or chest radiograph may prove useful
in patients with cardiac or respiratory diseases.
In general, the routine use of neuroimaging in delirium is not
recommended, because the overall diagnostic yield is low, and the findings
from neuroimaging change the management of patients in less than 10% of
cases (Hirao et al. 2006). Brain imaging techniques—computed
tomography (CT), positron emission tomography (PET), single-photon
emission computed tomography (SPECT), and magnetic resonance imaging
(MRI)—have low diagnostic yield in unselected patients but are
recommended in cases of head trauma or injury, for evaluation of new focal
neurological symptoms, for assessment for suspected encephalitis, or for
evaluation of fever of unknown origin. Cerebrospinal fluid examination,
accomplished through lumbar puncture, may be useful in cases where the
suspicion of meningitis, encephalitis, or subarachnoid hemorrhage is high
(Marcantonio 2011). It may also be indicated in cases where delirium is
persistent or where no etiology of delirium can be identified.
Pathophysiology of Delirium
The fundamental pathophysiological mechanisms of delirium remain
unclear, and because each episode of delirium has a unique set of
contributors representing different causal mechanisms, it is unlikely that a
single cause or pathophysiological mechanism for delirium will be
identified. Increasing evidence suggests that several sets of biological
factors interact and result in disruption of large-scale neuronal networks in
the brain, leading to acute confusion, cognitive dysfunction, and delirium
(Watt et al. 2013). Table 7–6 presents the mechanisms potentially
contributing to delirium and their level of evidence.
Although many neurotransmitters are implicated in delirium (Gaudreau
and Gagnon 2005), the most frequently considered mechanism of delirium
is dysfunction in the cholinergic system. Acetylcholine plays a key role in
mediating consciousness and attentional processes. Given that delirium is
manifested by an acute confusional state, often with alterations of
consciousness, it is likely to have a cholinergic basis. Evidence for the
cholinergic connection includes findings that anticholinergic drugs can
induce delirium in humans and animals and that serum anticholinergic
activity is increased in patients with delirium (Hshieh et al. 2008; Lauretani
et al. 2010). Also, cholinesterase inhibitors have been found to reduce
symptoms of delirium in some studies (Gleason 2003; Wengel et al. 1998).
Chronic stress induced by severe illness, trauma, or surgery involves
sympathetic and immune system activation that may lead to delirium; this
activation may include increased activity of the hypothalamic-pituitary-
adrenal axis with hypercortisolism, release of cerebral cytokines that alter
neurotransmitter systems, alterations in the thyroid axis, and modification
of blood-brain barrier permeability (Hughes et al. 2012). Neuroimaging
studies, using either CT or MRI, have demonstrated structural abnormalities
in the brains of patients with delirium, especially in the splenium of the
corpus callosum, thalamus, and right temporal lobe (Bogousslavsky et al.
1988; Doherty et al. 2005; Naughton et al. 1997; Ogasawara et al. 2005;
Takanashi et al. 2006). Advanced neuroimaging techniques find overall and
regional perfusion abnormalities in the brains of people with delirium (Fong
et al. 2006; Pfister et al. 2008). Functional imaging may help to distinguish
structural damage resulting from an episode of delirium from preexisting
changes (Choi et al. 2012).
Physiological stressors
Cortisol, S100B, neopterin, hypoxia O No
Metabolic disorders
Lactic acidosis E, O No
Hypo- or hyperglycemia O No
Insulin-like growth factor 1 (IGF-1) O Yes
Hypercapnia H Yes
Electrolyte disorders
Sodium, calcium, magnesium E, O No
Genetic factors
Apolipoprotein E (ApoE), dopamine transporter receptor O Yes
Glucocorticoid receptor O No
Toll-like receptor 4 H No
Note. E=Experimental—Controlled data available in humans (e.g., clinical trials and/or inference
from unintended side effects of medications); O=Observational—Only observational data available
in humans; H=Hypothetical—Studies not yet available in humans to support the mechanism.
Source. Reprinted from Inouye SK, Westendorp RG, Saczynski JS, et al: “Delirium in Elderly
People.” The Lancet 383(9920):911–922, 2014, with permission from Elsevier.
Precipitating factors
Medications
Multiple medications added 2.9 — — —
Psychoactive medication use 4.5 — — —
Sedative-hypnotics — — — 4.5
Use of physical restraints 3.2–4.4 — — —
Use of bladder catheter 2.4 — — —
Physiological
Elevated serum urea 5.1 — — 1.1
Elevated BUN/creatinine ratio 2.0 2.9 — —
Abnormal serum albumin — — 1.4 —
Abnormal sodium, glucose, or — 3.4 — —
potassium
Metabolic acidosis — — — 1.4
Infection — — — 3.1
Any iatrogenic event 1.9 — — —
Surgery
Aortic aneurysm — 8.3 — —
Noncardiac thoracic — 3.5 — —
Neurosurgery — — — 4.5
Trauma admission — — — 3.4
Urgent admission — — — 1.5
Coma — — — 1.8–21.3
Note. Data are relative risks. Some data are reported as ranges. BUN=blood urea nitrogen.
Source. Reprinted from Inouye SK, Westendorp RG, Saczynski JS, et al: “Delirium in Elderly
People.” The Lancet 383(9920):911–922, 2014, with permission from Elsevier.
Key Points
• Delirium is a common problem for older hospitalized persons.
• Delirium is the most frequent complication affecting surgical and
nonsurgical older patient populations and often goes unrecognized.
• Patients with delirium have a worse prognosis than comparable patients
without delirium and an increased risk of developing long-term cognitive
and functional decline.
• It is important to establish a patient’s level of baseline cognitive
functioning and course of cognitive change when evaluating for
delirium.
• The Confusion Assessment Method provides a simple diagnostic
algorithm and has become widely used for identification of delirium.
• Although delirium can be caused by a single factor, it is usually
multifactorial.
• Existing cognitive impairment and dementia are the leading risk factors
for development of delirium.
• Nonpharmacological approaches should be implemented as the first line
of treatment for delirium.
• Certain medications can induce or exacerbate an episode of delirium.
• Pharmacological management of delirium should be reserved for patients
with severe agitation.
References
Adamis D, Sharma N, Whelan PJ, et al: Delirium scales: a review of current
evidence. Aging Ment Health 14(5):543–555, 2010 20480420
Agostini JV, Inouye SK: Delirium, in Principles of Geriatric Medicine and
Gerontology, 5th Edition. New York, McGraw-Hill, 2003, pp 647–658
American Geriatrics Society: American Geriatrics Society updated Beers
Criteria for potentially inappropriate medication use in older adults. J
Am Geriatr Soc 60:616–631, 2012 22376048
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Barr J, Fraser GL, Puntillo K, et al: Clinical practice guidelines for the
management of pain, agitation, and delirium in adult patients in the
intensive care unit. Crit Care Med 41(1):263–306, 2013 23269131
Bergmann MA, Murphy KM, Kiely DK, et al: A model for management of
delirious postacute care patients. J Am Geriatr Soc 53(10):1817–1825,
2005 16181185
Bogousslavsky J, Ferrazzini M, Regli F, et al: Manic delirium and frontal-
like syndrome with paramedian infarction of the right thalamus. J
Neurol Neurosurg Psychiatry 51(1):116–119, 1988 3258356
Borson S, Scanlan J, Brush M, et al: The Mini-Cog: a cognitive “vital
signs” measure for dementia screening in multi-lingual elderly. Int J
Geriatr Psychiatry 15(11):1021–1027, 2000 11113982
Breitbart W, Rosenfeld B, Roth A, et al: The Memorial Delirium
Assessment Scale. J Pain Symptom Manage 13(3):128–137, 1997
9114631
Caplan GA, Coconis J, Board N, et al: Does home treatment affect
delirium? A randomised controlled trial of rehabilitation of elderly and
care at home or usual treatment (the REACH-OUT trial). Age Ageing
35(1):53–60, 2006 16239239
Centers for Medicare and Medicaid Services: Minimum Data Set, Version
3.0. Washington, DC, Centers for Medicare and Medicaid Services,
2010
Choi SH, Lee H, Chung TS, et al: Neural network functional connectivity
during and after an episode of delirium. Am J Psychiatry 169(5):498–
507, 2012 22549209
Clegg A, Young JB: Which medications to avoid in people at risk of
delirium: a systematic review. Age Ageing 40(1):23–29, 2011
21068014
Deschodt M, Braes T, Flamaing J, et al: Preventing delirium in older adults
with recent hip fracture through multidisciplinary geriatric
consultation. J Am Geriatr Soc 60(4):733–739, 2012 22429099
Doherty MJ, Jayadev S, Watson NF, et al: Clinical implications of splenium
magnetic resonance imaging signal changes. Arch Neurol 62(3):433–
437, 2005 15767508
Ely EW, Margolin R, Francis J, et al: Evaluation of delirium in critically ill
patients: validation of the Confusion Assessment Method for the
Intensive Care Unit (CAM-ICU). Crit Care Med 29(7):1370–1379,
2001 11445689
Fick D, Foreman M: Consequences of not recognizing delirium
superimposed on dementia in hospitalized elderly individuals. J
Gerontol Nurs 26(1):30–40, 2000 10776167
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Fong TG, Bogardus ST Jr, Daftary A, et al: Cerebral perfusion changes in
older delirious patients using 99mTc HMPAO SPECT. J Gerontol A
Biol Sci Med Sci 61(12):1294–1299, 2006 17234823
Foy A, O’Connell D, Henry D, et al: Benzodiazepine use as a cause of
cognitive impairment in elderly hospital inpatients. J Gerontol A Biol
Sci Med Sci 50(2):M99–M106, 1995 7874596
Gaudreau JD, Gagnon P: Psychotogenic drugs and delirium pathogenesis:
the central role of the thalamus. Med Hypotheses 64(3):471–475, 2005
15617851
Gaudreau JD, Gagnon P, Harel F, et al: Fast, systematic, and continuous
delirium assessment in hospitalized patients: the Nursing Delirium
Screening Scale. J Pain Symptom Manage 29(4):368–375, 2005
15857740
Gleason OC: Donepezil for postoperative delirium. Psychosomatics
44(5):437–438, 2003 12954923
Han JH, Zimmerman EE, Cutler N, et al: Delirium in older emergency
department patients: recognition, risk factors, and psychomotor
subtypes. Acad Emerg Med 16(3):193–200, 2009 19154565
Hirano LA, Bogardus ST Jr, Saluja S, et al: Clinical yield of computed
tomography brain scans in older general medical patients. J Am Geriatr
Soc 54(4):587–592, 2006 16686867
Hirao K, Ohnishi T, Matsuda H, et al: Functional interactions between
entorhinal cortex and posterior cingulate cortex at the very early stage
of Alzheimer’s disease using brain perfusion single-photon emission
computed tomography. Nucl Med Commun 27(2):151–156, 2006
16404228
Hshieh TT, Fong TG, Marcantonio ER, et al: Cholinergic deficiency
hypothesis in delirium: a synthesis of current evidence. J Gerontol A
Biol Sci Med Sci 63(7):764–772, 2008 18693233
Hufschmidt A, Shabarin V: Diagnostic yield of cerebral imaging in patients
with acute confusion. Acta Neurol Scand 118(4):245–250, 2008
18336626
Hughes CG, Patel MB, Pandharipande PP: Pathophysiology of acute brain
dysfunction: what’s the cause of all this confusion? Curr Opin Crit
Care 18(5):518–526, 2012 22941208
Inouye SK: The dilemma of delirium: clinical and research controversies
regarding diagnosis and evaluation of delirium in hospitalized elderly
medical patients. Am J Med 97(3):278–288, 1994 8092177
Inouye SK, Charpentier PA: Precipitating factors for delirium in
hospitalized elderly persons: predictive model and interrelationship
with baseline vulnerability. JAMA 275(11):852–857, 1996 8596223
Inouye SK, van Dyck CH, Alessi CA, et al: Clarifying confusion: the
confusion assessment method: a new method for detection of delirium.
Ann Intern Med 113(12):941–948, 1990 2240918
Inouye SK, Bogardus ST Jr, Charpentier PA, et al: A multicomponent
intervention to prevent delirium in hospitalized older patients. N Engl J
Med 340(9):669–676, 1999 10053175
Inouye SK, Foreman MD, Mion LC, et al: Nurses’ recognition of delirium
and its symptoms: comparison of nurse and researcher ratings. Arch
Intern Med 161(20): 2467–2473, 2001 11700159
Inouye SK, Leo-Summers L, Zhang Y, et al: A chart-based method for
identification of delirium: validation compared with interviewer ratings
using the Confusion Assessment Method. J Am Geriatr Soc 53(2):312–
318, 2005 15673358
Inouye SK, Baker DI, Fugal P, et al: Dissemination of the hospital elder life
program: implementation, adaptation, and successes. J Am Geriatr Soc
54(10):1492–1499, 2006 17038065
Inouye SK, Zhang Y, Jones RN, et al: Risk factors for delirium at discharge:
development and validation of a predictive model. Arch Intern Med
167(13):1406–1413, 2007 17620535
Inouye SK, Kosar CM, Tommet D, et al: The CAM-S: development and
validation of a new scoring system for delirium severity in 2 cohorts.
Ann Intern Med 160(8):526–533, 2014a 24733193
Inouye SK, Westendorp RG, Saczynski JS, et al: Delirium in elderly people.
Lancet 383(9920):911–922, 2014b 23992774
Jackson JC, Gordon SM, Hart RP, et al: The association between delirium
and cognitive decline: a review of the empirical literature.
Neuropsychol Rev 14(2):87–98, 2004 15264710
Jacobson S, Jerrier H: EEG in delirium. Semin Clin Neuropsychiatry
5(2):86–92, 2000 10837097
Jenssen S: Electroencephalogram in the dementia workup. Am J
Alzheimers Dis Other Demen 20(3):159–166, 2005 16003931
Larsen KA, Kelly SE, Stern TA, et al: Administration of olanzapine to
prevent postoperative delirium in elderly joint-replacement patients: a
randomized, controlled trial. Psychosomatics 51(5):409–418, 2010
20833940
Lauretani F, Ceda GP, Maggio M, et al: Capturing side-effect of medication
to identify persons at risk of delirium. Aging Clin Exp Res 22(5–
6):456–458, 2010 21422797
Leslie DL, Marcantonio ER, Zhang Y, et al: One-year health care costs
associated with delirium in the elderly population. Arch Intern Med
168(1):27–32, 2008 18195192
Lundström M, Edlund A, Karlsson S, et al: A multifactorial intervention
program reduces the duration of delirium, length of hospitalization, and
mortality in delirious patients. J Am Geriatr Soc 53(4):622–628, 2005
15817008
Marcantonio ER: In the clinic. Delirium. Ann Intern Med 154(11):ITC6-1–
ITC6-15; quiz ITC6–16, 2011 21646553
Marcantonio ER, Flacker JM, Wright RJ, et al: Reducing delirium after hip
fracture: a randomized trial. J Am Geriatr Soc 49(5): 516–522, 2001
11380742
Marcantonio ER, Kiely DK, Simon SE, et al: Outcomes of older people
admitted to postacute facilities with delirium. J Am Geriatr Soc
53(6):963–969, 2005 15935018
Marcantonio ER, Bergmann MA, Kiely DK, et al: Randomized trial of a
delirium abatement program for postacute skilled nursing facilities. J
Am Geriatr Soc 58(6):1019–1026, 2010 20487083
Martinez FT, Tobar C, Beddings CI, et al: Preventing delirium in an acute
hospital using a non-pharmacological intervention. Age Ageing
41(5):629–634, 2012 22589080
Milisen K, Foreman MD, Abraham IL, et al: A nurse-led interdisciplinary
intervention program for delirium in elderly hip-fracture patients. J Am
Geriatr Soc 49(5):523–532, 2001 11380743
Nasreddine ZS, Phillips NA, Bédirian V, et al: The Montreal Cognitive
Assessment, MoCA: a brief screening tool for mild cognitive
impairment. J Am Geriatr Soc 53(4):695–699, 2005 15817019
National Quality Measures: Delirium: proportion of patients meeting
diagnostic criteria on the Confusion Assessment Method (CAM).
February 27, 2003. Available at:
http://www.qualitymeasures.ahrq.gov/content.aspx?id=27635.
Accessed December 28, 2013.
Naughton BJ, Moran M, Ghaly Y, et al: Computed tomography scanning
and delirium in elder patients. Acad Emerg Med 4(12):1107–1110,
1997 9408423
Naughton BJ, Saltzman S, Ramadan F, et al: A multifactorial intervention to
reduce prevalence of delirium and shorten hospital length of stay. J Am
Geriatr Soc 53(1):18–23, 2005 15667371
Neelon VJ, Champagne MT, Carlson JR, et al: The NEECHAM Confusion
Scale: construction, validation, and clinical testing. Nurs Res
45(6):324–330, 1996 8941300
Ogasawara K, Komoribayashi N, Kobayashi M, et al: Neural damage
caused by cerebral hyperperfusion after arterial bypass surgery in a
patient with Moyamoya disease: case report. Neurosurgery 56(6):
E1380; discussion E1380, 2005 15918959
O’Mahony R, Murthy L, Akunne A, et al: Synopsis of the National Institute
for Health and Clinical Excellence guideline for prevention of delirium.
Ann Intern Med 154(11):746–751, 2011 21646557
Pandharipande PP, Girard TD, Jackson JC, et al; BRAIN-ICU Study
Investigators: Long-term cognitive impairment after critical illness. N
Engl J Med 369(14):1306–1316, 2013 24088092
Pfeiffer E: A short portable mental status questionnaire for the assessment
of organic brain deficit in elderly patients. J Am Geriatr Soc
23(10):433–441, 1975 1159263
Pfister D, Siegemund M, Dell-Kuster S, et al: Cerebral perfusion in sepsis-
associated delirium. Crit Care 12(3):R63, 2008 18457586
Pitkälä KH, Laurila JV, Strandberg TE, et al: Multicomponent geriatric
intervention for elderly inpatients with delirium: a randomized,
controlled trial. J Gerontol A Biol Sci Med Sci 61(2):176–181, 2006
16510862
Rudolph JL, Jones RN, Levkoff SE, et al: Derivation and validation of a
preoperative prediction rule for delirium after cardiac surgery.
Circulation 119(2):229–236, 2009 19118253
Saczynski J, Marcantonio ER, Quach L, et al: Cognitive trajectories after
postoperative delirium. N Engl J Med 367(1):30–39, 2012 22762316
Saczynski J, Kosar CM, Xu G, et al: A tale of two methods: chart and
interview methods for identifying delirium. J Am Geriatr Soc
62(3):518–524, 2014 24512042
Schor JD, Levkoff SE, Lipsitz LA, et al: Risk factors for delirium in
hospitalized elderly. JAMA 267(6):827–831, 1992 1732655
Schuurmans MJ, Shortridge-Baggett LM, Duursma SA: The Delirium
Observation Screening Scale: a screening instrument for delirium. Res
Theory Nurs Pract 17(1):31–50, 2003 12751884
Schweickert WD, Pohlman MC, Pohlman AS, et al: Early physical and
occupational therapy in mechanically ventilated, critically ill patients: a
randomised controlled trial. Lancet 373(9678):1874–1882, 2009
19446324
Steis MR, Evans L, Hirschman KB, et al: Screening for delirium using
family caregivers: convergent validity of the Family Confusion
Assessment Method and interviewer-rated Confusion Assessment
Method. J Am Geriatr Soc 60(11):2121–2126, 2012 23039310
Tabet N, Hudson S, Sweeney V, et al: An educational intervention can
prevent delirium on acute medical wards. Age Ageing 34(2):152–156,
2005 15713859
Takanashi J, Barkovich AJ, Shiihara T, et al: Widening spectrum of a
reversible splenial lesion with transiently reduced diffusion. AJNR Am
J Neuroradiol 27(4):836–838, 2006 16611774
Trzepacz PT, Baker RW, Greenhouse J: A symptom rating scale for
delirium. Psychiatry Res 23(1):89–97, 1988 3363018
Trzepacz PT, Mittal D, Torres R, et al: Validation of the Delirium Rating
Scale–Revised–98: comparison with the delirium rating scale and the
cognitive test for delirium. J Neuropsychiatry Clin Neurosci
13(2):229–242, 2001 11449030
van Eijk MM, Roes KC, Honing ML, et al: Effect of rivastigmine as an
adjunct to usual care with haloperidol on duration of delirium and
mortality in critically ill patients: a multicentre, double-blind, placebo-
controlled randomised trial. Lancet 376(9755):1829–1837, 2010
21056464
Wachter RM: Understanding Patient Safety, 2nd Edition. New York,
McGraw-Hill Medical, 2012
Wang W, Li HL, Wang DX, et al: Haloperidol prophylaxis decreases
delirium incidence in elderly patients after noncardiac surgery: a
randomized controlled trial. Crit Care Med 40(3):731–739, 2012
22067628
Watt D, Budding DE, Koziol LF: Delirium, in The Neuropsychology of
Psychopathology. Edited by Noggle CA, Dean RS. New York,
Springer, 2013, pp 425–440
Wei LA, Fearing MA, Sternberg EJ, et al: The Confusion Assessment
Method: a systematic review of current usage. J Am Geriatr Soc
56(5):823–830, 2008 18384586
Wengel SP, Roccaforte WH, Burke WJ: Donepezil improves symptoms of
delirium in dementia: implications for future research. J Geriatr
Psychiatry Neurol 11(3): 159–161, 1998 9894735
Wong CL, Holroyd-Leduc J, Simel DL, et al: Does this patient have
delirium? Value of bedside instruments. JAMA 304(7):779–786, 2010
20716741
World Health Organization: The ICD-10 Classification of Mental and
Behavioural Disorders: Diagnostic Criteria for Research. Geneva,
Switzerland, World Health Organization, 1993
Suggested Readings
American Geriatrics Society: American Geriatrics Society updated Beers
Criteria for potentially inappropriate medication use in older adults. J
Am Geriatr Soc 60:616–631, 2012 22376048
Inouye SK, Bogardus ST Jr, Charpentier PA, et al: A multicomponent
intervention to prevent delirium in hospitalized older patients. N Engl J
Med 340(9):669–676, 1999 10053175
Inouye SK, Baker DI, Fugal P, et al: Dissemination of the hospital elder life
program: implementation, adaptation, and successes. J Am Geriatr Soc
54(10):1492–1499, 2006 17038065
Inouye SK, Westendorp RG, Saczynski JS, et al: Delirium in elderly people.
Lancet 383(9920):911–922, 2014 23992774
Wong CL, Holroyd-Leduc J, Simel DL, et al: Does this patient have
delirium? Value of bedside instruments. JAMA 304(7):779–786, 2010
20716741
CHAPTER 8
Definitions
Table 8–1 provides definitions espoused by the American Association for
Geriatric Psychiatry (Lyketsos et al. 2006). Clinical syndromes such as dementia
and cognitive impairment not dementia (CIND) are differentiated from clinical
subsyndromes such as mild cognitive impairment (MCI). The table also clarifies
that AD refers to a process of characteristic pathological changes in the brain
that presumably causes (or influences) the observed dementia syndrome.
Although these definitions are important to the clinical world, one should
recognize that uncertainty remains about linking cognitive decline to brain
pathology. Many studies demonstrate relationships between specific anatomical
distributions of neuropathological markers and specific domains of cognitive
function (e.g., the presence of medial temporal neurofibrillary tangles affects
primarily episodic memory) (Dowling et al. 2011). Other studies show that even
older individuals without cognitive impairment can accumulate significant
neuropathological changes of AD, cerebral infarctions, and Lewy bodies
(Bennett et al. 2012; Negash et al. 2011). In fact, in a study undertaken to
examine the relationship of AD pathology, cerebral infarcts, and Lewy body
pathology to cognition in individuals without cognitive impairment, nearly all
individuals had AD pathology (more than 75% exhibiting amyloid) and a
significant percentage had macroscopic infarctions (22%), microscopic
infarctions (24%), and Lewy body pathology (13%) (Bennett et al. 2012).
Furthermore, up to one-third of individuals without dementia can have AD
lesions that meet criteria for intermediate or even high likelihood of AD (Negash
et al. 2011). In community settings, up to 88% of individuals with dementia or
cognitive impairment who come to autopsy have mixed rather than unitary brain
pathologies, including a combination of AD lesions (neocortical neurofibrillary
tangles and neuritic plaques), microvascular infarcts (microinfarcts and lacunar
infarcts), neocortical Lewy bodies, hippocampal sclerosis, and generalized brain
atrophy (White 2009). Although microvascular infarcts predominate as the sole
or dominant lesion in 33.8% of patients with dementia or cognitive impairment,
AD lesions predominate in 18.6% of these individuals and codominant lesions
(most often AD and microvascular) predominate in 14.2% (White 2009).
Functional Impairments
Patients with dementia have problems in their social and interpersonal
functioning and in their ability to live independently. Patients with milder
dementia have difficulties with instrumental activities of daily living (IADLs),
such as preparing a meal, balancing a checkbook, shopping, and driving.
Patients with more severe dementia develop impairments in their basic ADLs,
such as eating, toileting, dressing, and transferring. Changes in functional
abilities are associated with cognitive decline (Royall et al. 2007). Furthermore,
they have prognostic significance because declining functional abilities impact
caregiver burden and are associated with institutionalization (Massoud 2007).
Neuropsychiatric Impairments
Dementia has also been associated with neuropsychiatric symptoms (NPSs).
These are generally grouped into four types: 1) affective and motivational
symptoms, such as depression (affecting up to 50% of individuals with
dementia), apathy, anxiety, and irritability; 2) psychotic symptoms, such as
delusions or perceptual disturbances; 3) disturbances of basic drives, including
feeding, sleep, and sexuality; and 4) unexpected, socially inappropriate, or
disinhibited behaviors. The latter behaviors, such as spontaneous violence,
uncharacteristic vocalizations, intrusiveness, and wandering, typically arise in
more severe dementia; they represent behavioral manifestations of loss of
executive control, sometimes referred to as executive dysfunction syndrome
(Lyketsos et al. 2004). Research has found that over the course of a progressive
dementia, essentially all patients develop one or more of the NPSs (Okura et al.
2010; Steinberg et al. 2008). These symptoms are associated with a decline in
function and cognition among patients that may ultimately lead to increased
mortality (Levy et al. 2012). Furthermore, the severity of NPSs is a strong
predictor of caregiver burden (Bergvall et al. 2011), with increased rates of
depression, anxiety, stress, loss of sleep, medical illness, and mortality among
caregivers (Levy et al. 2012). Caregiver burden, in turn, results in greater usage
of institutionalization for patients with dementia (Levy et al. 2012).
Source. DSM-5 criteria for major neurocognitive disorder reprinted from Diagnostic and Statistical Manual
of Mental Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, pp. 602–605.
Used with permission. Copyright © 2013 American Psychiatric Association.
Neurological Impairments
Patients with dementia develop a range of neurological impairments. Depending
on the cause of dementia and specifically on the parts of the brain affected over
time, a range of neurological symptoms may occur. Most common are gait
disorders, especially unstable, ataxic, or labored gait. Other symptoms include
incontinence, focal findings, seizures, and, less commonly, cranial nerve
findings.
Source. DSM-5 criteria for mild neurocognitive disorder reprinted from Diagnostic and Statistical Manual
of Mental Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, pp. 605–606.
Used with permission. Copyright © 2013 American Psychiatric Association.
Conducting an Evaluation
Although a detailed discussion of how to evaluate a patient with suspected
dementia is beyond the scope of this chapter, we highlight critical aspects of the
evaluation, with a focus on taking a history, conducting a cognitive assessment,
and using diagnostic tests. A thorough discussion of the evaluation of the patient
with suspected dementia, including reasons for doing an evaluation, the setting
for the evaluation, and ways to communicate the diagnosis to the patient and
caregiver, is provided in Practical Dementia Care (Rabins et al. 2006).
History Taking
Because dementia is diagnosed clinically, a thorough medical history is
essential. A patient with suspected dementia may have difficulty providing
history due to language problems, memory disturbance, or anosognosia (lack of
insight). Therefore, it is critical to involve a reliable informant during history
taking. Informants need to be people who know the patient well, such as family
members. Because informants themselves can be influenced by their own mental
states, such as depression or denial of the situation, it is often useful to speak
with more than one informant to confirm or challenge discrepancies between the
history and the evaluation of the patient.
It is critical to date and elucidate the type of onset of cognitive symptoms
(e.g., insidious onset, abrupt onset following traumatic brain injury [TBI]), as
well as establish the progression of symptoms over time (e.g., gradual, stepwise,
or nonprogressive decline). Comparing the severity of the patient’s current
impairment with its duration often influences the differential diagnosis. For
instance, a slowly progressive dementia over years with insidious onset may
point to Alzheimer’s dementia, whereas a dementia that progresses rapidly over
months may point to dementia due to prion disease. Clinicians need to resolve
discrepancies in reports of severity and duration because they portend different
prognoses and recommendations. It is often more feasible to determine when the
patient was last well rather than when symptoms first started. Many times,
informants minimize early symptoms by attributing them to “normal aging.” It is
also important to remember that the history should systematically evaluate for
the presence or absence of the broader dementia syndrome presentation, as
discussed in the earlier section “Clinical Presentation of the Dementia
Syndrome.” Therefore, the history should assess for cortical and subcortical
cognitive symptoms; functional losses in social, interpersonal, and daily
functioning; the full range of NPSs; and neurological deficits.
Cognitive Assessment
Conducting a cognitive assessment is the central aspect of the evaluation. Many
specialists tend to use the Mini-Mental State Examination (MMSE; Folstein et
al. 1975) as their primary tool because it is brief, easy to use, and well known.
The MMSE, however, is inefficient in evaluating patients with milder cognitive
symptoms or mild dementia, especially those with subcortical features. This is
because the MMSE has ceiling effects, especially for premorbidly well-educated
and intelligent individuals, and has limitations in evaluating executive control
function. Furthermore, the MMSE is unable to discriminate subtle degrees of
impairment in severe dementia. We recommend that specialists in geriatric
psychiatry and other clinicians who work with patients broaden their use of
bedside standardized assessments. The Modified Mini-Mental State (3MS; Teng
and Chui 1987) and the Montreal Cognitive Assessment (MoCA; Nasreddine et
al. 2005) are two bedside cognitive tests that provide a more comprehensive
assessment of cognition. The 3MS and MoCA have many advantages: several
translations exist, and they have been validated in various languages; they assess
abstract thinking, delayed recall, and verbal fluency better than the MMSE; and
they have well-known population norms. In addition, for closer assessments of
executive functioning, geriatric psychiatrists should consider incorporating the
following three measures in every dementia evaluation: the Clock Drawing test
(van der Burg et al. 2004), the Frontal Assessment Battery (Dubois et al. 2000),
and the Mental Alternation Test (Jones et al. 1993).
Neuropsychological testing is often useful for differentiating dementia from
milder cognitive syndromes or normal aging or for clarifying the etiology of the
cognitive disorder. However, neuropsychological testing is not needed in every
case, assuming that the clinician conducts a standardized assessment using tools
similar to those discussed above. If neuropsychological testing is needed, the
clinician should have in mind specific questions that he or she wishes to address,
such as how to clarify the differential diagnosis or how to set the stage for
monitoring prognosis or response to treatment.
The diagnostic criteria for possible DLB require the presence of dementia
plus one core or suggestive feature. The DLB Consortium also provided a long
list of supportive features which lack diagnostic specificity but can support the
clinical diagnosis of DLB (McKeith et al. 2005). Examples of such supportive
features include falls, syncope, transient and unexplained loss of consciousness,
severe autonomic dysfunction, hallucinations (other than visual) or delusions,
depression, or brain imaging or EEG findings consistent with the diagnosis.
Fluctuating cognition occurs in about 60%–80% of people with DLB
(McKeith et al. 1996) and is often mistaken for delirium. The symptoms and
their duration vary between individuals and even within the same person.
Symptoms range widely from episodes resembling blackouts of absence seizures
to speech changes seen in strokes. They can last from seconds to days. Recurrent
visual hallucinations occur in over half of DLB cases (Simard and van Reekum
2004) and are the most frequently reported psychotic symptom. Hallucinations
tend to be images of people, animals, or objects (Ballard et al. 2013) but can be
as simple as shapes in the corner of one’s eye. They can occur at any time but
are more common at night and are not typically distressing unless accompanied
by delusions or occurring in severely demented individuals (Ballard et al. 2013).
Parkinsonian features are similar to those found in PD (e.g., dyskinesias,
rigidity, gait disorders, tremor) and occur in almost 70% of individuals with
DLB (Aarsland et al. 2001).
If dementia and parkinsonism coexist, the differential diagnosis is sorted out
by examining the relative course of the cognitive and motor symptoms. The
emergence of dementia after many years of motor symptoms supports a
diagnosis of PDD. In contrast, the early presence of dementia in a patient with
motor parkinsonism supports a diagnosis of DLB. Researchers use a 1-year
interval between onset of PD and onset of dementia to differentiate PDD from
DLB.
Many patients with Alzheimer’s dementia develop a clinical DLB picture,
reflected in the neuropathology, and many DLB patients have concurrent
Alzheimer’s dementia pathology. Daytime drowsiness and naps, staring spells,
and episodes of disorganized speech have high positive predictive value for the
diagnosis of DLB over Alzheimer’s dementia (Ferman et al. 2004). Both visual
hallucinations and delusions are more common in DLB (as well as in PDD) than
in Alzheimer’s dementia and have high positive predictive value. In fact, visual
hallucinations may be the most clinically useful feature to distinguish DLB from
AD (Tiraboschi et al. 2006). Neuroleptic sensitivity is much more common and
severe in DLB than in AD. Autonomic dysfunction, such as urinary
incontinence, tends to be an early sign in DLB, whereas it often occurs in late
stages of AD.
Although the progression of DLB tends to be similar overall to that of
Alzheimer’s dementia, the course of DLB is more variable. Because many
patients have a more fulminant course, some experts believe that DLB has a
worse prognosis than Alzheimer’s dementia (McKeith et al. 2004). DLB is
associated with considerable suffering for patients and families, in part because
of common, difficult-to-treat, and persistent NPSs (hallucinations, delusions,
and affective symptoms). Patients also tend to become affected early with
balance, sleep, and motor disorders and to become confined in their mobility.
Disease-Modifying Therapies
Alzheimer’s Disease
As articulated in the principles of care of the American Association for Geriatric
Psychiatry (AAGP) (Lyketsos et al. 2006), estrogen, anti-inflammatory agents
(e.g., prednisone, NSAIDs), and Ginkgo biloba are not effective treatments for
Alzheimer’s dementia. One large randomized controlled trial (RCT)
demonstrated that high doses of the antioxidant vitamin E delays progression of
Alzheimer’s dementia, lengthening the time before onset of the next phase by 2
years (Sano et al. 1997). Given safety concerns about dosing, the AAGP
recommended considering vitamin E for Alzheimer’s dementia but avoiding
doses above 400 IU/day. In a recent study evaluating the use of high-dose (2,000
IU/day) vitamin E in patients with mild to moderate AD, Dysken et al. (2014)
found that vitamin E may slow functional decline and decrease caregiver burden
for some patients. Under investigation are the usefulness of other antioxidants
(vitamins C and D), folate to reduce homocystinemia, and dietary modifications.
One of the most effective therapies for AD is the aggressive management of
associated vascular risk factors such as blood pressure (particularly keeping
systolic blood pressure below 160 mm Hg), high cholesterol, diabetes, obesity,
and sedentary lifestyle (Mielke et al. 2007). Although statins have shown
promise as treatments for cognitive decline and dementia in observational
studies, several large RCTs (e.g., Trompet et al. 2010) did not show any
significant effect on cognition.
A better understanding of the etiopathogenesis of Alzheimer’s dementia has
led to the development of therapies targeting amyloid precursor protein
metabolism, Aβ1–42 deposition or clearance, and ways by which amyloid injures
neurons. For example, inhibition of the enzymes β-secretase and β-secretase
targets the metabolism of amyloid precursor protein. Two types of
immunotherapies under investigation include injecting amyloid to create host
immunity (active immunization) or injecting intravenous immunoglobulin
antibodies to clear amyloid from the brain (passive immunization). Examples of
the latter include bapineuzumab and solanezumab, monoclonal antibodies that
bind to soluble Aβ and promote Aβ removal from the brain through the
bloodstream. Lastly, there are antitau therapies targeting tau
hyperphosphorylation and the inhibition of tau aggregation. Mangialasche et al.
(2010) published a summary of FDA clinical trials of disease-modifying agents.
For the most part, clinical trials have not been successful at producing safe
disease-modifying therapies for AD. Given the complexity of AD, the central
hypothesis of “one protein, one drug, one disease” needs to be modified
(Mangialasche et al. 2010). There are complex interactions at every level of the
human body, from genes to organs, as well as interactions between the person
and the environment. Few people have “pure” AD; amyloid plaques and Lewy
bodies may interact in yet unknown ways. It is difficult to account for these
nonlinear and rather unpredictable interactions in clinical trials. Many clinical
trials aim to find a selective compound that acts on a specific disease target (e.g.,
Aβ) to produce a desired clinical effect. However, some RCTs demonstrate that
even when therapies completely remove amyloid plaques, patients may still
develop end-stage dementia, suggesting that clearing amyloid plaques alone
cannot repair already damaged neurons or stop clinical progression of AD
(Holmes et al. 2008). Researchers neither completely understand the functions
of Aβ nor know the upper and lower safe limits of Aβ (Carrillo et al. 2013).
Many drugs bind to more than one target, increasing the risk of unforeseen and
unwanted complications. On the other hand, many enzymes, such as β-secretase,
have many substrates. It is challenging to design a drug to target β-secretase
because of its many substrates, as well as because of its wide substrate-binding
domain and the need for drugs to be able to cross the blood-brain barrier to
modulate the enzyme (Mangialasche et al. 2010).
Mangialasche et al. (2010) highlight other barriers to producing successful
disease-modifying therapies, including the following:
1. RCT protocols can be costly and time-consuming for the patient and the
caregiver, thus increasing withdrawal rates.
2. There is a lack of validated biomarkers with established cutoffs.
3. Studies that have unsuccessful pre-clinical and clinical results are not always
published, leading to repetition of errors.
4. Some drugs with positive results in preclinical and early clinical testing failed
large phase 3 RCTs.
5. Designing selective compounds without intolerable and potentially toxic side
effects is difficult.
6. Some drugs are hindered by the inability to reach a therapeutic dosage, or
treatment duration may have been too short to result in an effect.
7. There are genetic differences among patients (e.g., APOE*E4 carriers or
cytochrome P450 variability).
8. Reliable evaluation of patients requires adequate training and monitoring of
RCT raters.
9. Even if a drug targets mild to moderate stages of AD, the disease could have
already advanced too far for detection of a disease-modifying effect.
Indeed, even by the time MCI develops, the pathological process may have
advanced too far for treatments to be preventive, and it may be necessary to
target the disease earlier, at the stage of subjective memory impairment (Cooper
et al. 2013).
Given that disease-modifying therapies have largely been unsuccessful, that
targeting patients even in early stages of dementia may prove too late, and that
physicians are increasingly able to diagnose AD in very early stages (i.e., in a
pre-symptomatic phase) using biomarkers, trials have started targeting
prevention of AD. Carrillo et al. (2013) describe various ongoing trials in their
early phases. The following are some examples:
1. The Anti-Amyloid Treatment for Asymptomatic AD Trial targets cognitively
normal adults ages 65–80 years with PET evidence of Aβ. Because the
subjects have evidence of Aβ, they are more likely to demonstrate cognitive
decline over time during the trial, making them a good target population for
prevention. Subjects will be randomly assigned to receive solanezumab or
placebo. The primary outcome measure will be the rate of cognitive decline.
2. The Centre for Studies on Prevention of AD at the Douglas Institute in
Montreal, Quebec, is testing various preventive interventions in people older
than 60 years with a family history of AD. Interventions include medications
such as intranasal insulin and naproxen, and lifestyle modifications such as
aerobic exercise and dietary changes. The goals are to find optimal biomarker
end points and to identify interventions that move biomarkers or mitigate
their progression.
3. The Australian Imaging Biomarkers and Lifestyle Flagship Study of Aging is
investigating the interaction between vascular and AD pathologies and
exploring the possibility of delaying AD by reducing vascular risk factors
through physical activity. The target population is individuals older than 60
years with subjective memory complaints or a diagnosis of MCI and at least
one cardiovascular risk factor.
4. The Multidomain Alzheimer Preventive Trial in France is testing the
protective effects of a multifactorial intervention consisting of nutritional
advice, cognitive and physical activity, and omega-3 treatment. The targeted
population includes frail (because of evidence linking frailty with cognitive
decline) or prefrail individuals without dementia, ages 70 years and older,
with subjective memory complaints and a limitation in at least one IADL.
Source. Data from Stahl SM: Essential Psychopharmacology: The Prescriber’s Guide, Revised and
Updated Edition. New York, Cambridge University Press, 2006.
Several RCTs and withdrawal studies suggest that in patients with moderate
or severe AD, cholinesterase inhibitors are associated with clinically relevant
cognitive and functional benefits (Schwarz et al. 2012). The available evidence
also supports the use of memantine in patients with severe AD (National
Institute for Health and Care Excellence 2011; Schwarz et al. 2012). There is no
consensus, however, on when to switch from cholinesterase inhibitors to
memantine in individuals with severe AD (Schwarz et al. 2012).
Most experts recommend and data support initiating and titrating one of these
medications to the highest approved and tolerated dose, and assessing response
over 6–12 months (Rabins et al. 2006). Cholinesterase inhibitors lead to notable,
albeit temporary, symptomatic improvements in 10%–15% of cases.
Symptomatic improvements often last for 6–9 months. In clinical practice, the
judgment as to the response to treatment relies greatly on the expectations that
patients, their families, and their prescribing physicians have with this class of
medications (Massoud et al. 2011). Mild improvement or stabilization should be
considered an appropriate and realistic goal (Massoud et al. 2011). There is no
consensus, however, on how to estimate clinical efficacy (Schwarz et al. 2012).
There is debate, however, about whether patients should continue taking a
cholinesterase inhibitor for a longer period of time. In primary care settings in
the United States, most patients who start a prescription do not continue taking it
for more than a few months. Nevertheless, many experts recommend continuing
therapy once it begins because patients may develop a rapid cognitive and
functional decline when a cholinesterase inhibitor is discontinued (Schwarz et
al. 2012). It is unclear whether clinical deterioration following discontinuation
of a cholinesterase inhibitor is due to a loss of therapeutic effect or withdrawal
from the medication (Schwarz et al. 2012).
Other experts point out that some patients do well after a discontinuation trial
and that many benefit from switching agents. Massoud et al. (2011) reviewed
eight open-label or retrospective switching studies involving patients in the mild
to moderate stages of AD. In general, more than 50% of patients switched for
unsatisfactory response showed stabilization or improvement in global
evaluations, cognitive measures, and functional measures. Additionally, more
than 50% of individuals switched for intolerance tolerated the second agent. The
authors concluded that because cholinesterase inhibitors have individual
pharmacological properties, switching is a valid clinical choice for patients with
AD who either do not tolerate or have a lack of response to the initial agent.
They considered significant deterioration within the first 6 months of treatment
to represent a “lack of efficacy” and deterioration beyond the first 6 months of
treatment to represent “loss of efficacy” (measured by a decline of at least 2
points on the MMSE, as well as a documented deterioration in functional
autonomy, global impression, or behavior). The authors suggested that either a
lack or a loss of efficacy was a reasonable justification for switching agents. On
the other hand, they did not recommend switching for loss of response with the
initial cholinesterase inhibitors beyond 1 year of use, because this is most often
due to the natural course of AD (most evident during the transition from mild to
moderate or from moderate to severe stages). In these latter cases, the authors
recommended adding memantine to the cholinesterase inhibitor. In practice,
however, less than one-third of patients actually switch cholinesterase inhibitors.
One of the authors of this chapter (CGL) has followed a patient who benefited
from sequential use of four of the cholinesterase agents (huperzine, donepezil,
rivastigmine, and galantamine). Although clearly that was an extreme case,
which is probably quite rare, it points out that the issue of long-term therapy is
not a settled matter.
Although cholinesterase inhibitors and memantine are licensed for
monotherapy, combining the two types of drugs could theoretically potentiate
their benefits (Schwarz et al. 2012). Even though it is common practice to
combine cholinesterase inhibitors with memantine, many experts do not endorse
this practice because of a lack of clinical evidence to support additional benefit
(National Institute for Health and Care Excellence 2011; Schwarz et al. 2012).
Several randomized trials and observational studies demonstrated no additional
(or inconclusive) superiority of combination donepezil and memantine over
donepezil monotherapy (National Institute for Health and Care Excellence
2011).
Although clinical trials have suggested that cholinesterase inhibitors may be
of value in treating vascular dementia, none has been approved by the FDA for
that purpose. The results of one study suggest that compared with placebo,
donepezil is associated with increased mortality in vascular dementia. The
National Institute for Health and Care Excellence (2012) recommends that
cholinesterase inhibitors and memantine should not be prescribed clinically for
the treatment of cognitive decline in individuals with vascular dementia.
Both oral and transdermal preparations of rivastigmine have been approved
for the treatment of mild to moderate dementia in PDD. In a large, parallel-
group RCT of rivastigmine in PDD (Emre et al. 2004), patients who received
rivastigmine demonstrated an average 1-point advantage on the MMSE over the
24-week treatment period compared with those who received placebo. The
treatment group also demonstrated an average advantage of nearly 3 points on
the Alzheimer’s Disease Assessment Scale—Cognitive subscale, particularly in
areas of attention and executive function; 2 points on an ADLs scale; and 2
points on the total Neuropsychiatric Inventory score. In a Cochrane review of
cholinesterase inhibitors for PDD, DLB, and MCI associated with PD, Rolinski
et al. (2012) determined that current evidence supports the use of cholinesterase
inhibitors in patients with PDD, with a positive impact on global assessment,
cognitive function, NPSs, and ADLs. However, evidence from RCTs for
cholinesterase inhibitors other than rivastigmine is inconclusive (Ballard et al.
2011). Likewise, although memantine is well tolerated, it does not appear to
demonstrate positive effects on nonmotoric symptoms in PDD (Schwarz et al.
2012).
Cholinesterase inhibitors have been effective in treating cognitive impairment
in DLB, although this is considered an off-label use. Furthermore, they have
demonstrated therapeutic benefit in treating hallucinations and are considered
first-line therapy for treating psychosis in DLB. Studies on use of memantine for
DLB have been inconclusive (Schwarz et al. 2012). At best, memantine seems
to provide modest positive global effects in DLB. However, benefits from
memantine treatment are rapidly lost following discontinuation.
Although no cholinesterase inhibitors have demonstrated value in treating
cognitive deficits in patients with FTLD, a medication trial is reasonable when
the underlying cause of dementia is unclear.
Care management
Communication Use a calm, reassuring voice.
Avoid negative words and tone.
Use a light touch to reassure, calm, or redirect.
Allow patient sufficient time to respond to a question.
Help patient find words to express himself or herself.
Offer simple choices (no more than two at a time).
Simplifying the Use labeling or other visual cues.
environment Remove unnecessary objects to reduce confusion with
tasks.
Eliminate noise and distractions while you are
communicating or when patient is engaging in an activity.
Use simple visual reminders (e.g., arrows pointing to
bathroom).
Caregiver Understand that patient behaviors are not intentional.
education and Learn how to relax the rules (e.g., baths do not have to
support happen daily; there is no right or wrong in performing
activities or tasks as long as the patient and caregiver are
safe).
Go along with patient’s view of what is true and avoid
arguing or trying to reason or convince.
Task structuring Break each task into very simple steps.
Provide one- to two-step simple verbal commands.
Use verbal or tactile prompts for each step.
Provide structured daily routines that are predictable.
Activities Engage patient with meaningful activities that tap into
preserved capabilities and previous interests.
Introduce activities involving repetitive motion (washing
windows, folding towels, and putting coins in container).
Guide and cue the patient to initiate, sequence, organize,
and complete tasks.
Note. Domains and strategies listed are potential approaches used in randomized clinical trials but are not
exhaustive. One strategy may be effective for one patient but not another. Only consider the above
strategies following a thorough assessment and diagnostic workup.
Source. Adapted from Gitlin LN, Kales HC, Lyketsos CG: “Nonpharmacologic Management of Behavioral
Symptoms in Dementia.” Journal of the American Medical Association 308(19):2020–2029, 2012. Used
with permission.
Several different classes of medications have been studied. For some of them,
safety concerns exist and efficacy remains uncertain. The use of both
conventional and atypical antipsychotics is controversial because their efficacy
is modest and they have been associated with side effects (including rapid
cognitive decline), a higher risk of cerebrovascular or cardiovascular events, and
mortality in patients with dementia (Schneider et al. 2005). The FDA has issued
black box warnings regarding the use of antipsychotics in treating patients with
dementia-related psychosis (U.S. Food and Drug Administration 2008). Both
conventional and atypical antipsychotics carry this increased mortality risk in
dementia, whereas other psychotropic medications, such as antidepressants and
anticonvulsants, do not.
Because caregivers are the lifeline of the patient and are greatly affected by
dementia, they should be involved intimately in the development and
implementation of any dementia care program. As with addressing unmet needs
of individuals with dementia, a needs assessment can serve as a useful
foundation to gather data on unmet caregiver needs. Table 8–13 lists key
intervention areas involving caregivers (Lyketsos et al. 2006; Rabins et al. 2006;
Selwood et al. 2007). Mittelman et al. (2006) highlight the importance of the
delivery of interventions to caregivers. Their work suggests that caregiver
interventions can have effect sizes as large as or larger than medications in
delaying out-of-home placement for patients with dementia. Central to good
dementia care is making sure that caregivers are educated about dementia, have
an understanding of the diagnosis, are able to access resources, use respite
appropriately, and have an expert available around the clock to help them in
times of crisis.
Key Points
• Dementia is an epidemic clinical syndrome consisting of global cognitive
decline and memory deficits, with at least one other area of cognition
affected.
• Dementia can be accurately diagnosed and differentiated from cognitive
impairment not dementia and mild cognitive impairment.
• Amnestic mild cognitive impairment is likely the prodrome to Alzheimer’s
dementia, the most common form of dementia.
• The evaluation and differential diagnosis of dementia and of mild
neurocognitive disorders involve an initial focus on defining the
phenomenology of the syndrome and its associated features, followed by a
workup for a putative clause.
• The four pillars of dementia treatment are disease treatment, symptom
treatments, supportive care for the patient, and supportive care for the
caregiver. All of these areas must be addressed in contemporary dementia
care. A needs assessment is an efficient tool to help identify unmet care
needs.
• The concept of treatable and nontreatable dementias is no longer relevant; all
dementias are treatable, albeit not necessarily curable. One of the most
effective therapies for Alzheimer’s disease is aggressively managing
associated vascular risk factors.
• The amyloid cascade hypothesis of Alzheimer’s disease is rapidly evolving,
impacting clinical trials. Trials have started targeting prevention of
Alzheimer’s disease with antiamyloid therapies.
• Neuropsychiatric symptoms are nearly universal across dementia stages and
etiologies. No pharmacotherapy has U.S. Food and Drug Administration
approval for dementia-related neuropsychiatric symptoms.
Nonpharmacological interventions can be as effective as currently available
medications and should be considered first-line therapy except in emergency
situations.
• Disease-modifying therapies in Alzheimer’s disease have largely been
unsuccessful, and there are no disease-modifying treatments available for
dementia with Lewy bodies, Parkinson’s disease dementia, frontotemporal
lobar degeneration, or Creutzfeldt-Jakob disease. Cholinesterase inhibitors
provide only modest and temporary stabilization of the changes to cognition
and ADLs associated with the disease. They do not reverse or stop the
degenerative process.
References
Aarsland D, Ballard C, McKeith I, et al: Comparison of extrapyramidal signs in
dementia with Lewy bodies and Parkinson’s disease. J Neuropsychiatry
Clin Neurosci 13(3):374–379, 2001 11514644
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
7(3):270–279, 2011 21514249
American Psychiatric Association: Diagnostic and Statistical Manual of Mental
Disorders, 5th Edition. Washington, DC, American Psychiatric Association,
2013
Appleby BS, Lyketsos CG: Rapidly progressive dementias and the treatment of
human prion diseases. Expert Opin Pharmacother 12(1):1–12, 2011
21091283
Baker LD, Frank LL, Foster-Schubert K, et al: Effects of aerobic exercise on
mild cognitive impairment: a controlled trial. Arch Neurol 67(1):71–79,
2010 20065132
Ballard C, Corbett A: Management of neuropsychiatric symptoms in people
with dementia. CNS Drugs 24(9):729–739, 2010 20806986
Ballard C, Kahn Z, Corbett A: Treatment of dementia with Lewy bodies and
Parkinson’s disease dementia. Drugs Aging 28(10):769–777, 2011
21970305
Ballard C, Aarsland D, Francis P, et al: Neuropsychiatric symptoms in patients
with dementias associated with cortical Lewy bodies: pathophysiology,
clinical features, and pharmacological management. Drugs Aging
30(8):603–611, 2013 23681401
Banerjee S, Hellier J, Dewey M, et al: Sertraline or mirtazapine for depression in
dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-
controlled trial. Lancet 378(9789):403–411, 2011 21764118
Beach TG, Wilson JR, Sue LI, et al: Circle of Willis atherosclerosis: association
with Alzheimer’s disease, neuritic plaques and neurofibrillary tangles. Acta
Neuropathol 113(1):13–21, 2007 17021755
Bech-Azeddine R, Høgh P, Juhler M, et al: Idiopathic normal-pressure
hydrocephalus: clinical comorbidity correlated with cerebral biopsy
findings and outcome of cerebrospinal fluid shunting. J Neurol Neurosurg
Psychiatry 78(2):157–161, 2007 17012342
Bennett DA, Wilson RS, Boyle PA, et al: Relation of neuropathology to
cognition in persons without cognitive impairment. Ann Neurol 72(4):599–
609, 2012 23109154
Bentahir M, Nyabi O, Verhamme J: Presenilin clinical mutations can affect
gamma-secretase activity by different mechanisms. J Neurochem
96(3):732–742, 2006 16405513
Bergvall N, Brinck P, Eek D, et al: Relative importance of patient disease
indicators on informal care and caregiver burden in Alzheimer’s disease. Int
Psychogeriatr 23(1):73–85, 2011 20619068
Berman K, Brodaty H, Withall A, et al: Pharmacologic treatment of apathy in
dementia. Am J Geriatr Psychiatry 20(2):104–122, 2012 21841459
Bertoux M, Delavest M, de Souza LC, et al: Social Cognition and Emotional
Assessment differentiates frontotemporal dementia from depression. J
Neurol Neurosurg Psychiatry 83(4):411–416, 2012 22291219
Black BS, Johnston D, Handel S, et al: Manual for the Johns Hopkins Dementia
Care Needs Assessment (JHDCNA). Baltimore, MD, self-published, 2008
Black BS, Johnston D, Rabins PV, et al: Unmet needs of community-residing
persons with dementia and their informal caregivers: findings from the
maximizing independence at home study. J Am Geriatr Soc 61(12):2087–
2095, 2013 24479141
Blennow K, de Leon MJ, Zetterberg H: Alzheimer’s disease. Lancet
368(9533):387–403, 2006 16876668
Bowler JV, Hachinski V: Vascular cognitive impairment: a new approach to
vascular dementia. Baillieres Clin Neurol 4(2):357–376, 1995 7496625
Braak H, Ghebremedhin E, Rüb U, et al: Stages in the development of
Parkinson’s disease-related pathology. Cell Tissue Res 318(1):121–134,
2004 15338272
Braak H, Rüb U, Jansen Steur EN, et al: Cognitive status correlates with
neuropathologic stage in Parkinson disease. Neurology 64(8):1404–1410,
2005 15851731
Breitner JC, Wyse BW, Anthony JC, et al: APOE-epsilon4 count predicts age
when prevalence of AD increases, then declines: the Cache County Study.
Neurology 53(2):321–331, 1999 10430421
Cairns NJ, Bigio EH, Mackenzie IR, et al: Neuropathologic diagnostic and
nosologic criteria for frontotemporal lobar degeneration: consensus of the
Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol
114(1):5–22, 2007 17579875
Callahan CM, Boustani MA, Unverzagt FW, et al: Effectiveness of collaborative
care for older adults with Alzheimer disease in primary care: a randomized
controlled trial. JAMA 295(18):2148–2157, 2006 16684985
Camargos EF, Louzada LL, Quintas JL, et al: Trazodone improves sleep
parameters in Alzheimer disease patients: a randomized, double-blind, and
placebo-controlled study. Am J Geriatr Psychiatry (Jan):4, 2014 24495406
Canevelli M, Adali N, Voisin T, et al: Behavioral and psychological
subsyndromes in Alzheimer’s disease using the Neuropsychiatric Inventory.
Int J Geriatr Psychiatry 28(8):795–803, 2013 23147419
Carrillo MC, Brashear HR, Logovinsky V, et al: Can we prevent Alzheimer’s
disease? Secondary “prevention” trials in Alzheimer’s disease. Alzheimers
Dement 9(2): 123–131, 2013 23411394
Centers for Disease Control and Prevention: CDC’s Diagnostic Criteria for
Creutzfeldt-Jakob Disease (CJD), 2010. Atlanta, GA, CDC, August 26,
2010. Adapted from: World Health Organization: Global Surveillance,
Diagnosis, and Therapy of Human Transmissible Spongiform
Encephalopathies: Report of WHO Consultation. Geneva, Switzerland,
February 9–11, 1998; and Zerr I, Kallenberg K, Summers DM, et al:
Updated clinical diagnostic criteria for sporadic Creutzfeldt-Jakob disease.
Brain 132(Pt 10):2659–2668, 2009. Available at:
http://www.cdc.gov/ncidod/dvrd/cjd/diagnostic_criteria.html. Accessed
December 30, 2013.
Chow TW, Hynan LS, Lipton AM: MMSE scores decline at a greater rate in
frontotemporal degeneration than in AD. Dement Geriatr Cogn Disord
22(3):194–199, 2006 16899996
Claassen DO, Parisi JE, Giannini C, et al: Frontotemporal dementia mimicking
dementia with Lewy bodies. Cogn Behav Neurol 21(3):157–163, 2008
18797258
Collee JG, Bradley R, Liberski PP: Variant CJD (vCJD) and bovine spongiform
encephalopathy (BSE): 10 and 20 years on: part 2. Folia Neuropathol
44(2):102–110, 2006 16823692
Cools R, D’Esposito M: Inverted-U-shaped dopamine actions on human
working memory and cognitive control. Biol Psychiatry 69(12):e113–e125,
2011 21531388
Cooper C, Li R, Lyketsos C, Livingston G: Treatment for mild cognitive
impairment: systematic review. Br J Psychiatry 203(3):255–264, 2013
24085737
Cortelli P, Perani D, Montagna P, et al: Pre-symptomatic diagnosis in fatal
familial insomnia: serial neurophysiological and 18FDG-PET studies. Brain
129(Pt 3):668–675, 2006 16399807
de Medeiros K, Robert P, Gauthier S, et al: The Neuropsychiatric Inventory-
Clinician rating scale (NPI-C): reliability and validity of a revised
assessment of neuropsychiatric symptoms in dementia. Int Psychogeriatr
22(6):984–994, 2010 20594384
de Vasconcelos Cunha UG, Lopes Rocha F, Avila de Melo R, et al: A placebo-
controlled double-blind randomized study of venlafaxine in the treatment of
depression in dementia. Dement Geriatr Cogn Disord 24(1):36–41, 2007
17495474
Devanand DP, Mintzer J, Schultz SK, et al: Relapse risk after discontinuation of
risperidone in Alzheimer’s disease. N Engl J Med 367(16):1497–1507,
2012 23075176
Dowling NM, Tomaszewski Farias S, Reed BR, et al: Neuropathological
associates of multiple cognitive functions in two community-based cohorts
of older adults. J Int Neuropsychol Soc 17(4):602–614, 2011 21092373
Dubois B, Slachevsky A, Litvan I, et al: The FAB: a frontal assessment battery
at bedside. Neurology 55(11):1621–1626, 2000 11113214
Dysken MW, Sano M, Asthana S: Effect of vitamin E and memantine on
functional decline in Alzheimer disease: the TEAM-AD VA cooperative
randomized trial. JAMA 311(1):33–44, 2014 24381967
Emre M: Dementia associated with Parkinson’s disease. Lancet Neurol
2(4):229–237, 2003 12849211
Emre M, Aarsland D, Albanese A, et al: Rivastigmine for dementia associated
with Parkinson’s disease. N Engl J Med 351(24): 2509–2518, 2004
15590953
Engler H, Forsberg A, Almkvist O, et al: Two-year follow-up of amyloid
deposition in patients with Alzheimer’s disease. Brain 129(Pt 11):2856–
2866, 2006 16854944
Ferman TJ, Smith GE, Boeve BF, et al: DLB fluctuations: specific features that
reliably differentiate DLB from AD and normal aging. Neurology
62(2):181–187, 2004 14745051
Fernandez HH, Wu CK, Ott BR: Pharmacotherapy of dementia with Lewy
bodies. Expert Opin Pharmacother 4(11):2027–2037, 2003 14596656
Finney GR: Normal pressure hydrocephalus. Int Rev Neurobiol 84:263–281,
2009 19501723
Fisher A: Assessment of Motor and Process Skills: Development,
Standardization, and Administration Manual, Vol 1, 5th Edition. Fort
Collins, CO, Three Star Press, 2003
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical method
for grading the cognitive state of patients for the clinician. J Psychiatr Res
12(3):189–198, 1975 1202204
Galvin JE, Pollack J, Morris JC: Clinical phenotype of Parkinson disease
dementia. Neurology 67(9):1605–1611, 2006 17101891
Geda YE, Schneider LS, Gitlin LN, et al: Neuropsychiatric symptoms in
Alzheimer’s disease: past progress and anticipation of the future.
Alzheimers Dement 9(5):602–608, 2013 23562430
Geschwind MD, Shu H, Haman A, et al: Rapidly progressive dementia. Ann
Neurol 64(1):97–108, 2008 18668637
Gitlin LN, Kales HC, Lyketsos CG: Nonpharmacologic management of
behavioral symptoms in dementia. JAMA 308(19): 2020–2029, 2012
23168825
Graff-Radford NR: Normal pressure hydrocephalus. Neurol Clin 25(3):809–832,
2007 17659191
Graham JE, Rockwood K, Beattie BL, et al: Prevalence and severity of
cognitive impairment with and without dementia in an elderly population.
Lancet 349(9068): 1793–1796, 1997 9269213
Hachinski V: Vascular dementia: a radical redefinition. Dementia 5(3–4):130–
132, 1994 8087166
Hachinski V: The 2005 Thomas Willis Lecture: stroke and vascular cognitive
impairment: a transdisciplinary, translational and transactional approach.
Stroke 38(4):1396, 2007 17347469
Hamilton R, Patel S, Lee EB, et al: Lack of shunt response in suspected
idiopathic normal pressure hydrocephalus with Alzheimer disease
pathology. Ann Neurol 68(4):535–540, 2010 20687117
Hayden KM, Zandi PP, Lyketsos CG, et al: Vascular risk factors for incident
Alzheimer disease and vascular dementia: the Cache County Study.
Alzheimer Dis Assoc Disord 20(2):93–100, 2006 16772744
Holman RC, Belay ED, Christensen KY, et al: Human prion diseases in the
United States. PLoS ONE 5(1):e8521, 2010 20049325
Holmes C, Boche D, Wilkinson D, et al: Long-term effects of Abeta42
immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-
controlled phase I trial. Lancet 372(9634):216–223, 2008 18640458
Hughes TA, Ross HF, Musa S, et al: A 10-year study of the incidence of and
factors predicting dementia in Parkinson’s disease. Neurology 54(8):1596–
1602, 2000 10762499
Isoniemi H, Tenovuo O, Portin R, et al: Outcome of traumatic brain injury after
three decades—relationship to ApoE genotype. J Neurotrauma
23(11):1600–1608, 2006 17115907
Jellinger KA, Attems J: Prevalence and impact of cerebrovascular pathology in
Alzheimer’s disease and parkinsonism. Acta Neurol Scand 114(1):38–46,
2006 16774626
Johns MW: A new method for measuring daytime sleepiness: the Epworth
Sleepiness Scale. Sleep 14(6):540–545, 1991 1798888
Jones BN, Teng EL, Folstein MF, et al: A new bedside test of cognition for
patients with HIV infection. Ann Intern Med 119(10):1001–1004, 1993
8214976
Kales HC, Gitlin LN, Lyketsos CG: Management of neuropsychiatric symptoms
of dementia in clinical settings: recommendations from a multidisciplinary
expert panel. J Am Geriatr Soc 62(4):762–769, 2014 24635665
Kandiah N, Narasimhalu K, Lau PN, et al: Cognitive decline in early
Parkinson’s disease. Mov Disord 24(4):605–608, 2009 19191342
Kertesz A, Blair M, McMonagle P, et al: The diagnosis and course of
frontotemporal dementia. Alzheimer Dis Assoc Disord 21(2):155–163,
2007 17545742
Klassen BT, Ahlskog JE: Normal pressure hydrocephalus: how often does the
diagnosis hold water? Neurology 77(12):1119–1125, 2011 21849644
Knopman DS, DeKosky ST, Cummings JL, et al: Practice parameter: diagnosis
of dementia (an evidence-based review). Neurology 56(9):1143–1153, 2001
11342678
Lambert JC, Ibrahim-Verbaas CA, Harold D, et al: Meta-analysis of 74,046
individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat
Genet 45(12): 1452–1458, 2013 24162737
Leinonen V, Koivisto AM, Alafuzoff I, et al: Cortical brain biopsy in long-term
prognostication of 468 patients with possible normal pressure
hydrocephalus. Neurodegener Dis 10(1–4):166–169, 2012 22343771
Levy K, Lanctôt KL, Farber SB, et al: Does pharmacological treatment of
neuropsychiatric symptoms in Alzheimer’s disease relieve caregiver
burden? Drugs Aging 29(3):167–179, 2012 22350526
Lippa CF, Duda JE, Grossman M, et al: DLB and PDD boundary issues:
diagnosis, treatment, molecular pathology, and biomarkers. Neurology
68(11):812–819, 2007 17353469
Livingston G, Johnston K, Katona C, et al: Systematic review of psychological
approaches to the management of neuropsychiatric symptoms of dementia.
Am J Psychiatry 162(11):1996–2021, 2005 16263837
Longstreth WT Jr, Arnold AM, Beauchamp NJ Jr, et al: Incidence,
manifestations, and predictors of worsening white matter on serial cranial
magnetic resonance imaging in the elderly: the Cardiovascular Health
Study. Stroke 36(1):56–61, 2005 15569873
Lyketsos C: The prion dementias. Md Med J 48(1):18–22, 1999 10048280
Lyketsos C: Lessons from neuropsychiatry. J Neuropsychiatry Clin Neurosci
18(4): 445–449, 2006 17135372
Lyketsos CG, DelCampo L, Steinberg M, et al: Treating depression in
Alzheimer disease: efficacy and safety of sertraline therapy, and the benefits
of depression reduction: the DIADS. Arch Gen Psychiatry 60(7):737–746,
2003 12860778
Lyketsos CG, Rosenblatt A, Rabins P: Forgotten frontal lobe syndrome or
“executive dysfunction syndrome.” Psychosomatics 45(3):247–255, 2004
15123852
Lyketsos CG, Colenda CC, Beck C, et al: Position statement of the American
Association for Geriatric Psychiatry regarding principles of care for patients
with dementia resulting from Alzheimer disease. Am J Geriatr Psychiatry
14(7):561–572, 2006 16816009
Lyketsos CG, Samus QM, Baker A, et al: Effect of dementia and treatment of
dementia on time to discharge from assisted living facilities: the Maryland
Assisted Living Study. J Am Geriatr Soc 55(7):1031–1037, 2007 17608875
Lyketsos CG, Carrillo MC, Ryan JM, et al: Neuropsychiatric symptoms in
Alzheimer’s disease. Alzheimers Dement 7(5): 532–539, 2011 21889116
Mace NL, Rabins PV: The 36-Hour Day: A Family Guide to Caring for People
Who Have Alzheimer Disease, Related Dementias, and Memory Loss, 5th
Edition. Baltimore, MD, The Johns Hopkins University Press, 2011
Mangialasche F, Solomon A, Winblad B, et al: Alzheimer’s disease: clinical
trials and drug development. Lancet Neurol 9(7): 702–716, 2010 20610346
Martin M, Clare L, Altgassen AM, et al: Cognition-based interventions for
healthy older people and people with mild cognitive impairment. Cochrane
Database Syst Rev 1(1):CD006220, 2011 21249675
Massoud F: The role of functional assessment as an outcome measure in
antidementia treatment. Can J Neurol Sci 34 (suppl 1): S47–S51, 2007
17469682
Massoud F, Desmarais JE, Gauthier S: Switching cholinesterase inhibitors in
older adults with dementia. Int Psychogeriatr 23(3):372–378, 2011
21044399
McKeith I, Galasko D, Kosaka K, et al: Consensus guidelines for the clinical
and pathologic diagnosis of dementia with Lewy bodies (DLB): report of
the Consortium on DLB International Workshop. Neurology 47(5):1113–
1124, 1996 8909416
McKeith I, Mintzer J, Aarsland D, et al: Dementia with Lewy bodies. Lancet
Neurol 3(1):19–28, 2004 14693108
McKeith I, Dickson DW, Lowe J, et al: Diagnosis and management of dementia
with Lewy bodies: third report of the DLB Consortium. Neurology
65(12):1863–1872, 2005 16237129
Medway C, Morgan K: Review: the genetics of Alzheimer’s disease; putting
flesh on the bones. Neuropathol Appl Neurobiol 40(2):97–105, 2014
24443964
Meier U, Lemcke J, Neumann U: Predictors of outcome in patients with normal-
pressure hydrocephalus. Acta Neurochir Suppl (Wien) 96:352–357, 2006
16671484
Metzler-Baddeley C: A review of cognitive impairments in dementia with Lewy
bodies relative to Alzheimer’s disease and Parkinson’s disease with
dementia. Cortex 43(5):583–600, 2007 17715794
Michalczyk K, Ziman M: Current concepts in human prion protein (Prp)
misfolding, Prnp gene polymorphisms and their contribution to Creutzfeldt-
Jakob disease (CJD). Histol Histopathol 22(10):1149–1159, 2007 17616941
Mielke MM, Rosenberg PB, Tschanz J, et al: Vascular factors predict rate of
progression in Alzheimer disease. Neurology 69(19):1850–1858, 2007
17984453
Mittelman MS, Haley WE, Clay OJ, Roth DL: Improving caregiver well-being
delays nursing home placement of patients with Alzheimer disease.
Neurology 67(9): 1592–1599, 2006 17101889
Moghekar A, Rao S, Li M, et al: Large quantities of Abeta peptide are
constitutively released during amyloid precursor protein metabolism in vivo
and in vitro. J Biol Chem 286(18):15989–15997, 2011 21454701
Molinari V, Chiriboga D, Branch LG, et al: Provision of psychopharmacological
services in nursing homes. J Gerontol B Psychol Sci Soc Sci 65B(1):57–60,
2010 19846475
Moniz Cook ED, Swift K, James I, et al: Functional analysis-based interventions
for challenging behaviour in dementia. Cochrane Database Syst Rev
2:CD006929, 2012 22336826
Morelli L, Perry G, Tagliavini F: The contribution of the amyloid hypothesis to
the understanding of Alzheimer’s disease: a critical overview. Int J
Alzheimers Dis 2012:709613, 2012 22970405
Morrison CE, Borod JC, Brin MF, et al: Effects of levodopa on cognitive
functioning in moderate-to-severe Parkinson’s disease (MSPD). J Neural
Transm 111(10–11): 1333–1341, 2004 15480842
Mucke L, Selkoe DJ: Neurotoxicity of amyloidβ-protein: synaptic and network
dysfunction. Cold Spring Harb Perspect Med 2(7):a006338, 2012 22762015
Nagaraja D, Jayashree S: Randomized study of the dopamine receptor agonist
piribedil in the treatment of mild cognitive impairment. Am J Psychiatry
158(9):1517–1519, 2001 11532743
Nasreddine ZS, Phillips NA, Bédirian V, et al: The Montreal Cognitive
Assessment, MoCA: a brief screening tool for mild cognitive impairment. J
Am Geriatr Soc 53(4):695–699, 2005 15817019
National Institute for Health and Care Excellence: Donepezil, galantamine,
rivastigmine and memantine for the treatment of Alzheimer’s disease.
Review of NICE technology appraisal guidance 117, 2011. Available at:
http://guidance.nice.org.uk/TA217/Guidance/pdf/English. Accessed
February 2014.
National Institute for Health and Care Excellence: Dementia: Supporting people
with dementia and their carers in health and social care. Clinical Guideline
42, 2012. Available at: http://publications.nice.org.uk/dementia-
cg42/guidance#diagnosis-and-assessment-of-dementia. Accessed December
2013.
Neary D, Snowden J, Mann D: Frontotemporal dementia. Lancet Neurol
4(11):771–780, 2005 16239184
Negash S, Bennett DA, Wilson RS, et al: Cognition and neuropathology in
aging: multidimensional perspectives from the Rush Religious Orders Study
and Rush Memory and Aging Project. Curr Alzheimer Res 8(4):336–340,
2011 21222592
Newhouse P, Kellar K, Aisen P, et al: Nicotine treatment of mild cognitive
impairment: a 6-month double-blind pilot clinical trial. Neurology
78(2):91–101, 2012 22232050
Nowrangi MA, Rao V, Lyketsos CG: Epidemiology, assessment, and treatment
of dementia. Psychiatr Clin North Am 34(2): 275–294, vii, 2011 21536159
Okura T, Plassman BL, Steffens DC, et al: Prevalence of neuropsychiatric
symptoms and their association with functional limitations in older adults in
the United States: the aging, demographics, and memory study. J Am
Geriatr Soc 58(2):330–337, 2010 20374406
Palmer K, Berger AK, Monastero R, et al: Predictors of progression from mild
cognitive impairment to Alzheimer disease. Neurology 68(19):1596–1602,
2007 17485646
Peters ME, Rosenberg PB, Steinberg M: Neuropsychiatric symptoms as risk
factors for progression from CIND to dementia: the Cache County Study.
Am J Geriatr Psychiatry 21(11):1116–1124, 2013 23567370
Petersen RC, Thomas RG, Grundman M, et al: Vitamin E and donepezil for the
treatment of mild cognitive impairment. N Engl J Med 352(23):2379–2388,
2005 15829527
Petersen RC, Roberts RO, Knopman DS, et al: Mild cognitive impairment: ten
years later. Arch Neurol 66(12):1447–1455, 2009 20008648
Pimplikar SW: Reassessing the amyloid cascade hypothesis of Alzheimer’s
disease. Int J Biochem Cell Biol 41(6):1261–1268, 2009 19124085
Porsteinsson AP, Drye LT, Pollock BG, et al: Effect of citalopram on agitation in
Alzheimer disease: the CitAD randomized clinical trial. JAMA 311(7):682–
691, 2014 24549548
Prince M, Bryce R, Albanese E, et al: The global prevalence of dementia: a
systematic review and metaanalysis. Alzheimers Dement 9(1):63–75, 2013
23305823
Puoti G, Bizzi A, Forloni G, et al: Sporadic human prion diseases: molecular
insights and diagnosis. Lancet Neurol 11(7):618–628, 2012 22710755
Rabins PV, Lyketsos CG: Antipsychotic drugs in dementia: what should be
made of the risks? JAMA 294(15):1963–1965, 2005 16234504
Rabins PV, Lyketsos CG, Steele CD: Practical Dementia Care, 2nd Edition. New
York, Oxford University Press, 2006
Rabins PV, Schwartz S, Black BS, et al: Predictors of progression to severe
Alzheimer’s disease in an incidence sample. Alzheimers Dement 9(2):204–
207, 2013 23123228
Ranginwala NA, Hynan LS, Weiner MF, et al: Clinical criteria for the diagnosis
of Alzheimer disease: still good after all these years. Am J Geriatr
Psychiatry 16(5):384–388, 2008 18448850
Rogaeva E, Meng Y, Lee JH, et al: The neuronal sortilin-related receptor
SORL1 is genetically associated with Alzheimer disease. Nat Genet
39(2):168–177, 2007 17220890
Rolinski M, Fox C, Maidment I, et al: Cholinesterase inhibitors for dementia
with Lewy bodies, Parkinson’s disease dementia and cognitive impairment
in Parkinson’s disease. Cochrane Database Syst Rev 3:CD006504, 2012
22419314
Rosenberg PB, Johnston D, Lyketsos CG: A clinical approach to mild cognitive
impairment. Am J Psychiatry 163(11):1884–1890, 2006 17074938
Rosenberg PB, Drye LT, Martin BK, et al: Sertraline for the treatment of
depression in Alzheimer disease. Am J Geriatr Psychiatry 18(2):136–145,
2010 20087081
Rosenberg PB, Lanctôt KL, Drye LT, et al: Safety and efficacy of
methylphenidate for apathy in Alzheimer’s disease: a randomized, placebo-
controlled trial. J Clin Psychiatry 74(8):810–816, 2013 24021498
Roth HL: Dementia and sleep. Neurol Clin 30(4):1213–1248, 2012 23099135
Royall DR, Lauterbach EC, Kaufer D, et al: The cognitive correlates of
functional status: a review from the Committee on Research of the
American Neuropsychiatric Association. J Neuropsychiatry Clin Neurosci
19(3):249–265, 2007 17827410
Russ TC, Morling JR: Cholinesterase inhibitors for mild cognitive impairment.
Cochrane Database Syst Rev 9:CD009132, 2012 22972133
Sabbagh MN, Shah F, Reid RT, et al: Pathologic and nicotinic receptor binding
differences between mild cognitive impairment, Alzheimer disease, and
normal aging. Arch Neurol 63(12):1771–1776, 2006 17172618
Sabbagh MN, Adler CH, Lahti TJ, et al: Parkinson disease with dementia:
comparing patients with and without Alzheimer pathology. Alzheimer Dis
Assoc Disord 23(3):295–297, 2009 19812474
Samus QM, Johnston D, Black BS, et al: A multidimensional home-based care
coordination intervention for elders with memory disorders: the
Maximizing Independence at Home (MIND) pilot randomized trial. Am J
Geriatr Psychiatry 22(4):398–414, 2014 24502822
Sano M, Ernesto C, Thomas RG, et al: A controlled trial of selegiline, alpha-
tocopherol, or both as treatment for Alzheimer’s disease. The Alzheimer’s
Disease Cooperative Study. N Engl J Med 336(17):1216–1222, 1997
9110909
Scarmeas N, Brandt J, Blacker D, et al: Disruptive behavior as a predictor in
Alzheimer disease. Arch Neurol 64(12):1755–1761, 2007 18071039
Schneider JA, Wilson RS, Bienias JL, et al: Cerebral infarctions and the
likelihood of dementia from Alzheimer disease pathology. Neurology
62(7):1148–1155, 2004 15079015
Schneider LS, Dagerman KS, Insel P: Risk of death with atypical antipsychotic
drug treatment for dementia: meta-analysis of randomized placebo-
controlled trials. JAMA 294(15):1934–1943, 2005 16234500
Schwarz S, Froelich L, Burns A: Pharmacological treatment of dementia. Curr
Opin Psychiatry 25(6):542–550, 2012 22992546
Seitz DP, Gill SS, Herrmann N, et al: Pharmacological treatments for
neuropsychiatric symptoms of dementia in long-term care: a systematic
review. Int Psychogeriatr 25(2):185–203, 2013 23083438
Selkoe DJ: The molecular pathology of Alzheimer’s disease. Neuron 6(4):487–
498, 1991 1673054
Seltman RE, Matthews BR: Frontotemporal lobar degeneration: epidemiology,
pathology, diagnosis and management. CNS Drugs 26(10):841–870, 2012
22950490
Selwood A, Johnston K, Katona C, et al: Systematic review of the effect of
psychological interventions on family caregivers of people with dementia. J
Affect Disord 101(1–3):75–89, 2007 17173977
Sepehry AA, Lee PE, Hsiung GY, et al: Effect of selective serotonin reuptake
inhibitors in Alzheimer’s disease with comorbid depression: a meta-analysis
of depression and cognitive outcomes. Drugs Aging 29(10):793–806, 2012
23079957
Shimada H, Hirano S, Shinotoh H, et al: Mapping of brain acetylcholinesterase
alterations in Lewy body disease by PET. Neurology 73(4):273–278, 2009
19474411
Shulman JM, De Jager PL: Evidence for a common pathway linking
neurodegenerative diseases. Nat Genet 41(12):1261–1262, 2009 19935760
Simard M, van Reekum R: The acetylcholinesterase inhibitors for treatment of
cognitive and behavioral symptoms in dementia with Lewy bodies. J
Neuropsychiatry Clin Neurosci 16(4):409–425, 2004 15616167
Sink KM, Holden KF, Yaffe K: Pharmacological treatment of neuropsychiatric
symptoms of dementia: a review of the evidence. JAMA 293(5):596–608,
2005 15687315
Snowdon DA, Greiner LH, Mortimer JA, et al: Brain infarction and the clinical
expression of Alzheimer disease. The Nun Study. JAMA 277(10):813–817,
1997 9052711
Steinberg M, Shao H, Zandi P, et al: Point and 5-year period prevalence of
neuropsychiatric symptoms in dementia: the Cache County Study. Int J
Geriatr Psychiatry 23(2):170–177, 2008 17607801
Sultzer DL, Davis SM, Tariot PN, et al: Clinical symptom responses to atypical
antipsychotic medications in Alzheimer’s disease: phase 1 outcomes from
the CATIE-AD effectiveness trial. Am J Psychiatry 165(7):844–854, 2008
18519523
Teng EL, Chui HC: The Modified Mini-Mental State (3MS) examination. J Clin
Psychiatry 48(8):314–318, 1987 3611032
Theis W, Bleiler L, Alzheimer’s Association: 2013 Alzheimer’s disease facts
and figures. Alzheimers Dement 9(2):208–245, 2013 23507120
Tiraboschi P, Salmon DP, Hansen LA, et al: What best differentiates Lewy body
from Alzheimer’s disease in early stage dementia? Brain 129(Pt 3):729–
735, 2006 16401618
Treiber KA, Lyketsos CG, Corcoran C, et al: Vascular factors and risk for
neuropsychiatric symptoms in Alzheimer’s disease: the Cache County
Study. Int Psychogeriatr 20(3):538–553, 2008 18289451
Tremont G, Davis J, Papandonatos GD, et al: A telephone intervention for
dementia caregivers: background, design, and baseline characteristics.
Contemp Clin Trials 36(2):338–347, 2013 23916916
Trompet S, van Vliet P, de Craen AJ, et al: Pravastatin and cognitive function in
the elderly: results of the PROSPER study. J Neurol 257(1):85–90, 2010
19653027
Tschanz JT, Welsh-Bohmer KA, Lyketsos CG, et al: Conversion to dementia
from mild cognitive disorder: the Cache County Study. Neurology
67(2):229–234, 2006 16864813
Tschanz JT, Corcoran CD, Schwartz S, et al: Progression of cognitive,
functional, and neuropsychiatric symptom domains in a population cohort
with Alzheimer dementia: the Cache County Dementia Progression study.
Am J Geriatr Psychiatry 19(6):532–542, 2011 21606896
Tschanz JT, Norton MC, Zandi PP, et al: The Cache County Study on Memory
in Aging: factors affecting risk of Alzheimer’s disease and its progression
after onset. Int Rev Psychiatry 25(6):673–685, 2013 24423221
U.S. Food and Drug Administration: FDA Drug Safety and Availability:
Information for Healthcare Professionals: Conventional Antipsychotics,
2008. Available at: http://www.fda.gov/Drugs/DrugSafety/ucm124830.htm.
Accessed March 1, 2013.
U.S. Food and Drug Administration: FDA Drug Safety and Availability:
Revised Recommendations for Celexa (Citalopram Hydrobromide) Related
to a Potential Risk of Abnormal Heart Rhythms With High Doses, 2012.
Available at: http://www.fda.gov/Drugs/Drug-Safety/ucm297391.htm.
Accessed March 1, 2013.
van der Burg M, Bouwen A, Stessens J, et al: Scoring clock tests for dementia
screening: a comparison of two scoring methods. Int J Geriatr Psychiatry
19(7):685–689, 2004 15254925
Vanneste J, Augustijn P, Dirven C, et al: Shunting normal-pressure
hydrocephalus: do the benefits outweigh the risks? A multicenter study and
literature review. Neurology 42(1):54–59, 1992 1734324
Villemagne VL, Fodero-Tavoletti MT, Pike KE, et al: The ART of loss: Abeta
imaging in the evaluation of Alzheimer’s disease and other dementias. Mol
Neurobiol 38(1):1–15, 2008 18690556
Walsh DM, Selkoe DJ: A beta oligomers: a decade of discovery. J Neurochem
101(5): 1172–1184, 2007 17286590
Weintraub D, Burn DJ: Parkinson’s disease: the quintessential neuropsychiatric
disorder. Mov Disord 26(6):1022–1031, 2011 21626547
Weintraub D, Katz IR: Pharmacologic interventions for psychosis and agitation
in neurodegenerative diseases: evidence about efficacy and safety. Psychiatr
Clin North Am 28:941–983, ix–x, 2005 16325736
Weintraub D, Rosenberg PB, Drye LT, et al: Sertraline for the treatment of
depression in Alzheimer disease: week-24 outcomes. Am J Geriatr
Psychiatry 18(4):332–340, 2010 20220589
White L: Brain lesions at autopsy in older Japanese-American men as related to
cognitive impairment and dementia in the final years of life: a summary
report from the Honolulu-Asia Aging Study. J Alzheimers Dis 18(3):713–
725, 2009 19661625
Wilson RK, Williams MA: Normal pressure hydrocephalus. Clin Geriatr Med
22:935–951, viii, 2006 17000344
Wolfson C, Wolfson DB, Asgharian M, et al: A reevaluation of the duration of
survival after the onset of dementia. N Engl J Med 344(15):1111–1116,
2001 11297701
Wong DF, Rosenberg PB, Zhou Y: In vivo imaging of amyloid deposition in
Alzheimer disease using the radioligand 18F-AV-45 (florbetapir [corrected]
F 18). J Nucl Med 51(6):913–920, 2010 20501908
Woodruff BK: Evaluation of rapidly progressive dementia. Semin Neurol
27(4):363–375, 2007 17701874
Yang G, Wang Y, Tian J, et al: Huperzine A for Alzheimer’s disease: a
systematic review and meta-analysis of randomized clinical trials. PLoS
One 8(9):e74916, 2013 24086396
Zadikoff C, Fox SH, Tang-Wai DF, et al: A comparison of the mini mental state
exam to the Montreal Cognitive Assessment in identifying cognitive deficits
in Parkinson’s disease. Mov Disord 23(2):297–299, 2008 18044697
Suggested Readings
Blennow K, de Leon MJ, Zetterberg H: Alzheimer’s disease. Lancet 368:387–
403, 2006
Livingston G, Johnston K, Katona C, et al. for the Old Age Task Force of the
World Federation of Biological Psychiatry: Systematic review of
psychological approaches to the management of neuropsychiatric symptoms
of dementia. Am J Psychiatry 162:1996–2021, 2005
Lyketsos CG: Lessons from neuropsychiatry. J Neuropsychiatry Clin Neurosci
18:445–449, 2006
Mace NL and Rabins PV: The 36-Hour Day: A Family Guide to Caring for
People Who Have Alzheimer Disease, Related Dementias, and Memory
Loss, 5th Edition. Baltimore, MD, The Johns Hopkins University Press,
2011
Metzler-Baddeley C: A review of cognitive impairments in dementia with Lewy
bodies relative to Alzheimer’s disease and Parkinson’s disease with
dementia. Cortex 43:583–600, 2007
Neary D, Snowden J, Mann D: Frontotemporal dementia. Lancet Neurol 4:771–
780, 2005
Prince M, Bryce R, Albanese E, et al: The global prevalence of dementia: a
systematic review and metaanalysis. Alzheimers Dement 9(1):63–75.e2,
2013
Rabins PV, Lyketsos CG, Steele CD: Practical Dementia Care, 2nd Edition. New
York, Oxford University Press, 2006
Rosenberg PB, Johnston D, Lyketsos CG: A clinical approach to mild cognitive
impairment. Am J Psychiatry 163:1884–1890, 2006
Sink KM, Holden KF, Yaffe K: Pharmacological treatment of neuropsychiatric
symptoms of dementia: a review of the evidence. JAMA 293:596–608,
2005
Steinberg M, Shao H, Zandi P, et al: Point and 5-year period prevalence of
neuropsychiatric symptoms in dementia: the Cache County Study. Int J
Geriatr Psychiatry 23:170–177, 2008
CHAPTER 9
Depressive Disorders
Dan G. Blazer, M.D., Ph.D.
David C. Steffens, M.D., M.H.S.
Epidemiology
An overview of the epidemiology of psychiatric disorders in late life is
presented in Chapter 1, “Demography and Epidemiology of Psychiatric
Disorders in Late Life”; in this chapter, we discuss information relevant to
mood disorders. Using a community survey, investigators at Duke
University Medical Center attempted to untangle the different subtypes of
depression in late life (Blazer et al. 1987b). More than 1,300 older adults in
urban and rural communities who were age 60 or older were screened for
depressive symptomatology. Of the 27% reporting depressive symptoms,
19% had mild dysphoria only. Persons with symptomatic depression—that
is, subjects with more severe depressive symptoms—made up 4% of the
population. These individuals were primarily experiencing stressors, such
as physical illness and stressful life events. Only 2% had dysthymia, and
0.8% were experiencing a current major depressive episode. No cases of
current manic episode were identified.
In another study, the frequency of major depression in the community
was approximately 3.7% among individuals ages 75–84 and 2.1% among
those ages 75 years and older (Center for Multicultural Mental Health
Research 2012); 1.2% had a mixed depression and anxiety syndrome. These
data suggest that the DSM-5 depression categories do not apply to most
depressed older adults in the community. Surveys have confirmed the lower
frequency of major depression in the community (Kessler et al. 2005).
In hospital and long-term-care settings, the frequency of major
depression among older adults is much higher than in community settings.
Up to 21% of hospitalized elders meet criteria for a major depressive
episode, and an additional 20%–25% have minor (subsyndromal)
depression (Koenig et al. 1988). Rates of major depression among elderly
nursing home patients are even higher, exceeding 25% in some studies
(Parmelee et al. 1989).
How does one reconcile these seemingly disparate results? “Depression
in late life” remains a generic term that captures many constructs, some of
which are well defined and others of which are ill defined. The burden of
depression in elderly patients, as indicated by the just-described frequency
of significant depressive symptoms in community populations, is
unquestioned. Many older persons with atypical presentations of depression
do not meet criteria for major depression yet have clinically significant
depressive symptoms (Hybels et al. 2001).
The Epidemiologic Catchment Area surveys identified a bipolar disorder
in 9.7% of nursing home patients, which suggests that nursing homes may
have become a dumping ground for such patients (Weissman et al. 1991). In
clinical settings, a bipolar disorder has been identified in about 10%–25%
of geriatric patients with a mood disorder and 3%–10% of all older
psychiatric patients (Wylie et al. 1999; Young and Klerman 1992). About
5% of all individuals admitted as geropsychiatry inpatients present with
mania (Yassa et al. 1988).
Clinical Course
Episodes of depression across the life cycle, especially episodes of more
severe major depression, almost always remit or at least partially remit.
Nevertheless, depression is a chronic and recurrent illness. Data from a
community study from the Netherlands illustrate this chronicity. Among
subjects with clinically significant depressive symptoms, 23% improved,
44% experienced an unfavorable but fluctuating course, and 33%
experienced a severe and chronic course. In a second group of subjects with
less severe depression, 25% experienced a chronic course. Overall, 35% of
those subjects diagnosed with major depression and 52% of the subjects
diagnosed with DSM-IV dysthymic disorder experienced a chronic course
(Beekman et al. 2002).
Studies that have focused on older adults in clinical settings have found
similar chronicity (Alexopoulos et al. 1996; Baldwin and Jolley 1986;
Murphy 1983; Post 1962). In an early study, Post (1962) followed a clinical
sample for 6 years and found that 31% recovered and remained well over
the follow-up, 28% experienced at least one relapse but later recovered, and
23% only partially recovered, whereas 17% remained depressed throughout
the period of follow-up. Murphy (1983) followed a group of elderly
depressed subjects (many of whom were medically ill) over 1 year. Of those
subjects, 35% experienced a good outcome, 48% experienced a fluctuating
course or remained continuously ill, and 14% died. In an Australian study
of a group of elderly patients who were followed for 25 years after
experiencing severe depression earlier in life, only 12% fully remitted and
experienced no recurrences over the period of follow-up (Brodaty et al.
2001). The prognosis from clinical studies of depressed older adults with
late-life depression, however, is similar to that found among younger adults
if the older adult is not plagued with comorbid medical illness, functional
impairment, or cognitive impairment (Keller et al. 1982a, 1982b).
Comorbid depression is associated with a less favorable prognosis. For
example, when major depressive disorder is comorbid with persistent
depressive disorder (dysthymia), the prognosis is poor. Factors predicting
partial remission were similar to those predicting no remission, and poor
social support and functional limitations increased the risk for poor
outcome in these subjects (Hybels et al. 2005).
Cognitive impairment is often associated with depressive symptoms.
When the depression improves, the cognitive impairment often improves as
well. Nevertheless, comorbid depression and cognitive impairment are a
risk factor for the later emergence of Alzheimer’s disease (Alexopoulos et
al. 1993). Therefore, early depressive symptoms associated with mild
cognitive impairment may represent a preclinical sign and should be
considered a risk for impending Alzheimer’s disease or vascular dementia
(Li et al. 2001). Depression can further complicate Alzheimer’s disease
over time by increasing disability and physical aggression, thereby
contributing to depression among caregivers (González-Salvador et al.
1999). Depressive symptoms resolve spontaneously without requiring
intensive therapy (such as medication therapy) at a greater frequency
among patients with Alzheimer’s disease than among older adults
experiencing depression and vascular dementia, in whom depressive
symptoms tend to be persistent and refractory to drug treatment (Li et al.
2001). Considerable study in recent years has focused on structural brain
changes and the outcome of depression in elderly individuals. For example,
Ribeiz et al. (2013) found that both a lower baseline score on the Mini-
Mental State Examination (MMSE; Folstein et al. 1975) and lower baseline
orbitofrontal cortex volume predicted a poorer outcome.
Depression and medical problems are frequently comorbid, and the
causal pathway may be bidirectional (Blazer and Hybels 2005). Depression,
for example, is a frequent and important contributing cause of weight loss
in late life (Morley and Kraenzle 1994). Frailty, leading to profound weight
loss, can in turn contribute to clinically important depressive symptoms
(Fried 1994). Many chronic medical illnesses are associated with
depression, including cardiovascular disease, diabetes, osteoporosis, and hip
fracture, to name a few (Blazer et al. 2002b; Lenze et al. 2007; Lyles 2001;
Williams et al. 2002). The mechanisms that explain the close association
between depression and physical illness in older adults are for the most part
not well documented. Clues, however, have emerged. For example, platelet
activation is increased in older depressed patients, especially those with a
specific polymorphism in the serotonin transporter–linked promoter region.
This polymorphism leads to higher levels of platelet aggregation and β-
thromboglobulin, both pathophysiological changes that increase the risk for
myocardial infarction (Whyte et al. 2001).
Perhaps the best-established association between depression and
physical problems is that between depression and functional impairment
(Blazer et al. 1991; Bruce 2001). For example, in one study, older adults
who were depressed were 67% more likely than those who were not
depressed to experience impairment in activities of daily living and 73%
more likely to experience mobility restrictions 6 years following initial
evaluation (Penninx et al. 1999b). Disability, in turn, can increase the risk
for depressive symptoms (Kennedy et al. 1990; Roberts et al. 1997).
Explanations for this bidirectional association include the following: the
propensity for physical disability to lead to a higher frequency of negative
life events, which in turn increases the risk for depression; the loss of
independence and the concomitant need to depend on others; restricted
social and leisure activities secondary to physical disability; and the
isolation and reduced quality of social support often inherent with physical
disability (Blazer 1983). Functional decline, however, is not inevitable
when the older adult becomes depressed. For example, the instrumental
support provided to older adults, such as help in tasks necessary for daily
living, can be protective against the worsening of performance on
instrumental abilities, and therefore buffer against the onset of depression
(Hays et al. 2001).
Despite the frequency and clinical importance of late-life depression,
long-term psychiatric follow-up investigations involving survivors of severe
episodes of this disorder have been relatively scarce. The typical course of
major depression throughout the life cycle is remission and relapse. In
patients who have a history of recurrent episodes, new episodes tend to be
associated with similar symptoms and to last about as long as prior
episodes. Classic studies of depression suggest that the duration of major
depression throughout the life cycle is approximately 9 months if untreated
(Dunner 1985). As individuals age, however, they may experience episodes
more frequently, and these episodes can merge into a chronic condition.
Most clinicians and clinical investigators report that more than 70% of
elderly patients with major depression who are treated with antidepressant
medication (at an adequate dose for a sufficient time) recover from the
index episode of depression if the depression is uncomplicated by comorbid
factors. Reynolds et al. (1992) reported that treatment of physically healthy
depressed elders with combined interpersonal psychotherapy and
nortriptyline was associated with response rates nearing 80%. In a long-
term outcome study of treatment-resistant depression in older adults, 47%
of patients were clinically improved 15 months after treatment with an
antidepressant or electroconvulsive therapy (ECT); at 4-year follow-up, the
percentage had increased to 71% (Stoudemire et al. 1993). These optimistic
results are tempered by the fact that physical illness and impaired cognition
may complicate both the course of depression and the response to treatment
(Baldwin and Jolley 1986; Koenig et al. 1989a; Murphy et al. 1988). Once
an older patient has experienced one or more moderate to severe episodes
of major depressive disorder, he or she may need to continue antidepressant
therapy permanently to minimize the risk of relapse (Reynolds et al. 2006).
Persons with a persistent depressive disorder (dysthymia) experience a
more chronic clinical course than do persons with major depressive
disorder. By DSM-5 definition, an individual’s depressive symptoms must
last at least 2 years for a diagnosis of dysthymia to be made. An
undetermined percentage (as high as 4%–8%) of community-dwelling (and
possibly institutionalized) elders experience moderately severe depressive
symptoms for more than 2 years, although they report intermittent periods,
lasting longer than a few days, of relative freedom from depressive
symptoms. The severity of their symptoms is not great enough to meet the
criteria for major depressive disorder, and the intermittent symptom-free
periods disqualify them from the diagnosis of persistent depressive disorder
(dysthymia). Nevertheless, these individuals experience chronic depression.
Other older adults experience chronic depression secondary to medical or
even psychiatric disease (e.g., alcoholism, obsessive-compulsive disorder).
Each of these disorders contributes to residual depression in ambulatory
elderly individuals.
Factors associated with improved outcome in late-life depression include
a history of recovery from previous episodes, a family history of
depression, female gender, extroverted personality, current or recent
employment, absence of substance abuse, no history of major psychiatric
disorder, less severe depressive symptomatology, and absence of major life
events and serious medical illness (Baldwin and Jolley 1986; Cole et al.
1999; Post 1972). The results of a number of studies suggest a relationship
between social support during an index episode and outcome in
psychological distress and depression. Intuition suggests that adequate
support should enhance recovery from a severe or moderately severe
psychiatric disorder such as major depression. In a study involving 493
community respondents, Holahan and Moos (1981) found that decreases in
social support of family and within work environments were related to
increases in psychological maladjustment over a 1-year follow-up period.
Coping behavior may also affect the prognosis of late-life depression.
One of the coping behaviors most commonly used by recent generations of
older adults is religious involvement. In a study involving 100 middle-aged
or elderly adults, one-third of men and nearly two-thirds of women used
religious cognitions or behaviors to help them cope with a stressful period
(Koenig et al. 1988). A number of investigators have reported inverse
associations between religious coping and depressive symptoms in older
adults with or without medical illness (Braam et al. 1997b; Idler 1987;
Koenig 2007a; Koenig et al. 1992; Pressman et al. 1990). A study involving
850 hospitalized medically ill older adults found that those using religion to
cope were less likely to be depressed and more likely to experience
improvement in depressive symptoms over time (Koenig et al. 1992).
Religious involvement also appears to be a predictor of faster recovery from
depression in both community-dwelling and clinical samples of older adults
(Braam et al. 1997a; Koenig 2007b; Koenig et al. 1998).
Personality pathology is another measurable phenomenon that is known
to affect the outcome of major depression (Weissman et al. 1978).
Unfortunately, there are no published reports of personality as a predictor of
major depression outcome in elderly patients. In addition, studies with
mixed-age samples have generally been confounded by the interaction of
depressive symptomatology and personality variables at baseline
assessment—that is, a depressed affect may influence the underlying
personality. Given the stability of personality in late life, longitudinal
studies of relationships between personality and both onset and outcome of
major depression would be most helpful.
The outcome of bipolar disorder in elderly individuals remains virtually
unknown. In a long-term follow-up study involving 500 patients in Iowa,
Winokur (1975) found that bipolar disorder tended to occur in clusters over
time and speculated that early-onset bipolar illness may “burn itself out” in
time. Shulman and Post (1980) studied elderly patients with bipolar
disorder and found that only 8% had their first episode of mania before age
40. In a review of records of a small number of untreated patients with
severe and prolonged bipolar disorder, Cutler and Post (1982) found a
tendency toward more rapid recurrences late in the illness, with decreasing
periods of remission. In other words, if bipolar disorder reemerges in the
later years, the episodes of mania—or mania mixed with depression—may
once again cluster, just as the disorder typically clusters at earlier periods of
life. Most clinicians who have worked with patients with bipolar disorder in
late life recognize the tendency of these disorders to recur frequently for a
time, only to remit for an extended period.
Ambelas (1987) emphasized a relationship between life events and onset
of mania, noting that stressful events were more likely to precede early-
onset mania than late-onset mania. Likewise, Shulman (1989) stressed that
increased cerebral vulnerability due to organic insults (stroke, head trauma,
other brain insults) played a stronger role than life events in precipitating
late-onset mania (a factor that may also play a role in treatment resistance).
Young and Klerman (1992) emphasized the low rates of familial affective
disorder and the increased frequency of certain diseases and drug use
associated with late age at onset.
Controversy exists over whether age at onset of first manic episode
affects response to treatment. Glasser and Rabins (1984) described no
significant age-related differences in presentation or treatment response.
Young and Falk (1989) reported that late-onset mania was associated with
lower activity level, lower sexual drive, and less-disturbed thought
processes; however, they also found that older age was associated with
longer hospitalization, greater residual psychopathology, and poorer
response to pharmacotherapy. Eastham et al. (1998) suggested that elderly
patients with bipolar disorder often require lithium dosages that are 25%–
50% lower than those used in younger patients. Data on the use of valproic
acid in elderly patients with bipolar disorder are limited but encouraging.
There is almost no information on the use of carbamazepine or other drugs
in late-life bipolar disorder. ECT has been reported to be well tolerated and
effective in the treatment of these patients (Eastham et al. 1998).
All-cause mortality is a significant adverse outcome resulting from late-
life depression. In one review of multiple reports, 72% of the studies
demonstrated a positive association between depression and mortality in
elderly people (Schulz et al. 2002). In another review of 23 outcome studies
of depression in subjects ages 65 years and older, the pooled odds of dying
if subjects were depressed were 1.75 (Geerlings et al. 2002). A longer
follow-up predicted smaller effect size. Both severity and duration of
depressive symptoms predicted mortality in the elderly population in these
studies (Geerlings et al. 2002). For example, investigators from the
Epidemiologic Catchment Area study found a fourfold increase in the odds
of dying over a follow-up of 15 months if persons over age 55 years
experienced a mood disorder (Bruce and Leaf 1989). Also, community-
dwelling persons in the Netherlands who experienced both major and minor
depression were at increased risk for cardiac mortality (Penninx et al.
1999a).
The association between depression and mortality holds in many of these
studies, despite the addition of potentially confounding variables. In studies
from North Carolina and New York, however, investigators failed to find an
association (Blazer et al. 2001; Thomas et al. 1992).
One reason for the lack of association in some studies may be the
selection of specific control variables, especially chronic disease and
functional impairment. For example, in a study of the North Carolina
Established Populations for Epidemiologic Study of the Elderly cohort, the
unadjusted relative odds of mortality among depressed subjects at baseline
was 1.98 (Blazer et al. 2001). These odds moved toward unity when other
risk factors, such as chronic disease, were controlled, and when health
habits, cognitive impairment, functional impairment, and social support
were added to the model. Therefore, the specific control variables used in
mortality studies may determine the association between depression and
mortality.
The effect of depression on mortality may vary by sex. In elderly
Japanese American men but not women, depressive symptoms were a risk
for mortality in the physically healthy (Takeshita et al. 2002). In another
study, depressive symptoms were a significant risk factor for cardiovascular
but not cancer mortality in older women (Whooley and Browner 1998). In a
controversial report, investigators found that subthreshold depression, as
indicated by CES-D scores of 12–16, was not associated with mortality in
men but was negatively related to 3-year mortality in women (odds ratio
0.56) (Hybels et al. 2002). In other words, mild depressive symptoms were
protective in this highly controlled analysis of community-based data.
Murphy et al. (1988) examined all-cause mortality in a 4-year follow-up
study involving 120 depressed elderly psychiatric inpatients, comparing
them with 197 age- and gender-matched control subjects. Among the
depressed women, mortality was twice the expected rate; among the men, it
was three times the expected rate. Older men with physical health problems
and depression were significantly more likely to die than were similarly
aged, physically ill, nondepressed men. A study involving elderly veterans
hospitalized with medical illness found a significantly higher mortality rate
during hospitalization for 41 patients who were depressed than for 41
nondepressed patients matched for age, gender, and severity and type of
medical illness (Koenig et al. 1989b). Rovner et al. (1991) also found
higher death rates among elderly nursing home patients with depression.
Several other studies involving medically ill elderly patients like-wise
found greater mortality rates among those with depression than among
those without (Arfken et al. 1999; Black 1999; Covinsky et al. 1999).
These studies indicate higher rates of mortality for depressed elderly
patients (men in particular) with concurrent physical health problems; in
clinical samples, this relationship persisted after important covariates were
controlled. The association between late-life depression and mortality is
intuitively attractive, because older persons are thought to experience loss
of meaningful roles and emotional support through retirement, death of
friends or a spouse, decreased economic and material well-being, and
increased isolation and loneliness (Atchley 1989; Fassler and Gaviria
1978). When poor physical health compounds these age-related changes,
depression may be particularly prone to affect health outcomes.
Etiology1
Biological Origins
Genetics
The etiology of late-life affective disorders is undoubtedly multifactorial
(Table 9–2). Twin and family studies, along with studies focusing on
molecular genetics, provide strong evidence for a heritable contribution to
the etiology of major depression and bipolar disorder (Gatz et al. 1992).
Evidence that these genetic factors weigh heavily in the etiology of bipolar
disorders in late life is virtually nonexistent, although the biological nature
of this disorder would suggest some genetic contribution. Evidence from
studies of unipolar depression in late life suggests that the genetic
contribution is weaker in late-life depression than in depression at earlier
stages of the life cycle. In a study of elderly twins in Sweden, genetic
influences accounted for 16% of the variance in total depression scores on
the CES-D and 19% of the somatic symptoms. In contrast, genetic
influences minimally contributed to the variance of symptoms of depressed
mood and positive affect (Gatz et al. 1992).
Hypothesized genetic markers for late-life depression have usually not
stood the test of well-controlled studies, yet some studies present intriguing
possibilities. Many candidate genes, such as genes encoding enzymes for
serotonin synthesis, the norepinephrine transporter, and even the
neurotrophic factor, have been hypothesized and explored in animal studies.
As yet, however, these hypotheses await further testing in older adults with
depression (Smith et al. 2007). Some pharmacological studies with
depressed elders have shown associations between speed of response and
antidepressant side effects with the serotonin transporter promoter
polymorphism (Pollock 2000).
Despite many studies of the allele producing the ε4 type of
apolipoprotein E (APOE*E4), no association was found in a community
sample between APOE*E4 and depressive symptoms (Blazer et al. 2002a),
although in a clinical sample of 500 subjects, Yen et al. (2007) found that
APOE*E4 was associated with more severe depression, after adjusting for
vascular diseases and lipid profile. Investigators have also concentrated on
genes that may be associated with cerebrovascular lesions that are
associated with depression. In one study, subjects with late-onset major
depression exhibited a higher frequency of C677T mutation of the
methylene tetrahydrofolate reductase enzyme compared with control
subjects. This mutation may place older persons at risk for major depression
associated with cerebrovascular lesions (vascular depression) (Hickie et al.
2001). In another study, carriers of the C1166 allele of the type 1
angiotensin II receptor gene (AGTR1) had greater ratios of lesioned to
nonlesioned white matter tissue across multiple tracts (Taylor et al. 2013b).
Neurotransmitter Dysfunction
Decreased activity of serotonergic neurotransmission has been the focus of
much research on the pathophysiology of depression in younger adults.
Dysfunctions in the transmission of norepinephrine and dopamine have also
been implicated. Serotonin activity, specifically 5-HT2A receptor binding,
decreases dramatically in a variety of brain regions through midlife, yet
there is less decrease from midlife to late life. 5-HT2A receptors in normal
subjects decreased markedly from young adulthood to midlife (70% from
the levels at age 20 years through the fifth decade) and then leveled off as
age advanced (Sheline et al. 2002). Activity of these receptors, however,
may vary with age.
Endocrine Changes
Hypersecretion of corticotropin-releasing factor (CRF) has been associated
with depression for many years across the life cycle. CRF is thought to
mediate sleep and appetite disturbances, reduced libido, and psychomotor
changes (Arborelius et al. 1999) and is diminished with normal aging
(Gottfries 1990). Aging is associated with an increased responsiveness of
dehydroepiandrosterone sulfate to CRF (Luisi et al. 1998).
Serum testosterone levels decline with aging (Liverman and Blazer
2004) and have been found to be even lower in elderly men with dysthymia
than in men without depressive symptoms (Seidman et al. 2002). The
efficacy of testosterone treatment for major depression in men, however,
has not been established (Liverman and Blazer 2004). In women,
improvement of mood has resulted from hormone replacement (Sherwin
and Gelfand 1985).
Endocrine dysregulation over time has been associated with anatomical
changes related to late-life depressive symptoms, suggesting a vicious cycle
downward to chronic and moderately severe depressive symptoms.
Depressive symptoms have been hypothesized to cause atrophy of the
hippocampus (Sapolsky 1996, 2001; Sheline et al. 1996; Steffens et al.
2002). Stress that accumulates over the life cycle may lead to a sustained
increase in secretion of cortisol, leading to loss of preexisting hippocampal
neurons (Sapolsky 1996). This loss may be prevented in part by use of
antidepressant medications (Czéh et al. 2001).
Vascular Depression
For many years vascular risk factors have been known to be associated with
depressive symptoms (Post 1962). Because major depression is a frequent
outcome of stroke (Robinson and Price 1982) and hypertension (Rabkin et
al. 1983), investigators have proposed a vascular-based depression among
elderly individuals (Coffey et al. 1990; Krishnan et al. 1988; Kumar et al.
2002; Olin et al. 2002; Post 1962). In a study of 139 depressed older
individuals, 54% met neuroimaging criteria for subcortical ischemic
vascular depression. Age was most strongly associated with the increased
prevalence of subcortical changes; also associated were lassitude, a history
of hypertension, and poorer outcome (Krishnan et al. 2004; Taylor et al.
2013a). In a U.S. sample, vascular depression was more prevalent among
African American elderly persons than among Caucasians (Reinliebb et al.
2014). Vascular depression is associated with white matter hyperintensities
(Guttmann et al. 1998; Krishnan et al. 1997). These lesions probably
contribute to the disruption of neural circuits associated with depression
(Taylor et al. 2013a).
Clinical symptoms and signs associated with these vascular impairments
resemble impairments found in frontal lobe syndromes. Magnetic resonance
imaging (MRI) of depressed patients has revealed structural abnormalities
in areas related to limbic-cortical-striatal-pallidal-thalamic-cortical
pathways (George et al. 1994), including the frontal lobes (Krishnan et al.
1993), caudate (Krishnan et al. 1992), and putamen (Husain et al. 1991;
Sheline 2003). In MRI studies of mood disorders, structures that make up
this tract show volume loss or structural abnormalities (Sheline 2003).
Psychological Origins
Psychological factors, such as personality attributes, neuroticism, cognitive
distortions, and emotional control, may contribute to the onset of late-life
depression but are not specific to the origins of depression in older
individuals. Therefore, we briefly address some related studies.
In a study comparing older patients with and without personality
disorder, Morse and Lynch (2004) found that those with a personality
disorder were four times more likely to continue with or experience a
reemergence of depressive symptoms. Specific personality traits were not
correlated with clinical features of depression, such as age at onset and
number of previous episodes. Nevertheless, some of the traits were
associated with depressive symptoms such as hopelessness. Basic
personality attributes often underlie the origin and expression of depressive
symptoms in older adults.
Neuroticism—a construct rarely studied by psychiatrists in North
America despite being popular in Europe and Australia—is consistently
associated with late-life depressive symptoms in cross-sectional and
longitudinal studies of community samples (Henderson et al. 1993, 1997;
Lyness et al. 2002) and older adults in residential homes (Eisses et al.
2004). Cognitive distortions (Beck 1967) are among the most studied
psychological origins of depression across the life cycle. Depressed
individuals may overreact to life events or misinterpret these events and
exaggerate their adverse outcome. For example, in a study of the experience
and impact of adverse life events, older patients with major depression
reported more adverse life events in the recent past and a greater negative
impact of these events (particularly for interpersonal conflicts) than did
comparison groups of elderly patients with dysthymia and healthy control
subjects (Devanand et al. 2002). It is not clear whether the reported impact
reflects an increased vulnerability to events or a bias in reporting due to
current depressed mood. In another study from a community sample,
elderly persons with more frequent depressive symptoms used acceptance,
rumination, and catastrophizing (maladaptive cognitive distortions) to a
higher extent and positive reappraisal to a lower extent than did those with
fewer symptoms (Kraaij and de Wilde 2001). Presence of neuroticism was
associated with poor mood and cognitive outcomes among treated older
patients followed up to 3 years (Steffens et al. 2013).
Beekman and colleagues, in the Longitudinal Aging Study Amsterdam
(Beekman et al. 1995), found that major and minor depression, as well as
the persistence and emergence of depressive symptoms over 3 years, were
predicted by external locus of control (Beekman et al. 2001). Higher levels
of mastery—that is, a perception of being able to accomplish tasks and
having control over one’s life—have been shown to have a direct
association with fewer depressive symptoms in older individuals and to
buffer the adverse impact of disability on depression (Jang et al. 2002).
Self-efficacy may have a direct effect and also may work indirectly through
its effect on social support to prevent depressive symptoms, as indicated in
a sample of older adults followed for 1 year (Holahan and Holahan 1987).
In a study of patients with insomnia and depression, dysfunctional beliefs
about not being able to function and needing to avoid activities after a night
of disrupted sleep were related to mental and physical fatigue (Carney et al.
2014).
Social Origins
In addition to having biological and psychological origins, late-life
depression derives from social origins, including stressful life events,
bereavement, chronic stress or strain, low socioeconomic status, and
impaired social support. The relative contribution of these factors appears to
vary across the life cycle.
Some years ago, Murphy (1982) found a strong association between both
severe life events (e.g., bereavement, life-threatening illness of someone
else, major personal illness) and social difficulties (e.g., difficulties in
health of someone close to subject, housing issues, marital and family
relationships) with the onset of late-life depression. Elders lacking a
confidant were especially vulnerable to the effects of life stress. Social
support may therefore buffer the effect of a stressful event. The association,
however, may not be straightforward. Based on a meta-analysis of 25
studies of the relationship between negative life events and depression in
late life, Kraaij et al. (2002) reported that the total number of life events and
the total number of daily hassles were strongly associated with depressive
symptoms, as would be expected. In contrast, sudden unexpected events
were not related to depression.
Compared with younger adults, older adults are at greater risk for
depressive symptoms secondary to stressful life events. At least three
factors, however, modify this risk. First, ongoing problems may have a
smaller effect on the risk for depression in older individuals than in younger
individuals (Bruce 2002). For example, in one study, the onset of depressive
symptoms was not associated with baseline psychosocial stressors but was
associated with factors that changed through time (Kennedy et al. 1990).
Second, stressful events that are predictable or “on-time” events often cause
less depression in older adults than in younger adults. For example, death of
a spouse is a severe and at times catastrophic event leading to depression.
For young or middle-aged adults, this event is unexpected and the
adjustment is especially difficult. Older adults, in contrast, recognize that
death of a spouse is expected (by observing their peers) and have actually
rehearsed the event, such as by considering what they might do if a spouse
dies. Third, many events that can lead to depression, such as divorce and
difficulties with the law, are more frequent early in life than in late life. In
one study, significant difficulty with the law (something more serious than a
traffic violation) was reported during the preceding year by 9% of younger
individuals but less than 1% of older individuals (Hughes et al. 1988).
Bereavement is a common cause of depressive symptoms in late life
(Clayton 1990; de Beurs et al. 2001; Prigerson et al. 1994). In a study of
1,810 community-dwelling older adults, onset of clinically significant
depressive symptoms over a 3-year follow-up was predicted by death of a
partner or other relatives (Prigerson et al. 1994). Although some studies
have found bereavement to predict depressive symptoms, others have not
(Prince et al. 1998).
Yet another common cause of depression in later life is chronic strain.
For example, the prevalence of depressive symptoms in caregivers of
people with dementia is 43%–47% (Livingston et al. 1996; Waite et al.
2004). Therefore, providing support for caregivers is important to prevent
the onset and progression of depression in this vulnerable group of elders.
In cross-sectional analyses using data from a community study of older
Mexican Americans, financial strain was associated with level of depressive
symptoms (Black et al. 1998).
Lower socioeconomic status has been associated with depression across
the life cycle. Both the frequency of depressive symptoms and their
persistence over 2–4 years were associated with socioeconomic
disadvantage in a sample of community-dwelling adults ages 50 years and
older who originally met criteria for major depression (Mojtabai and Olfson
2004). In another study, although the level of education did not predict
emergence of depressive symptoms over 1 year, emergence of depression
over a 3-year period was predicted by lower level of education (Beekman et
al. 2001).
Social support is a multifactorial construct that includes perception,
structure of the social network, and tangible help and assistance (Turner and
Turner 1999). Perceived social support has proved to be the most robust
predictor of late-life depressive symptoms (Bruce 2002). Investigators from
Hong Kong, in a community study, found that depressive symptoms were
associated with impaired social support (including network size, network
composition, social contact frequency, satisfaction with social support, and
instrumental-emotional support) (Chi and Chou 2001). Findings from
another longitudinal study substantiated that poor social support predicted
depressive symptoms at follow-up after 3–6 years (Henderson et al. 1997).
The impact of social support on depression may vary by sex. In one study
of middle-aged and older patients, impaired social support was associated
with poorer outcome of major depression in older men but not older women
(George et al. 1989).
The clinician must not assume that older adults in general experience a
deficit in social support. Social support is perceived to be adequate by older
individuals, even among clinical samples (Blazer 1982). Old social
networks thin out, but new ones emerge for many people. Most older people
believe that they have enough contact with both family and friends and
assess the relationships that they have with their social networks as positive
(Cornoni-Huntley et al. 1990). Even so, when the social network is depleted
suddenly, either through loss of someone close to the older adult (e.g., a
spouse or child) or through a change in the quality of the relationship (e.g.,
a dispute within the family), impaired social support may emerge as a most
important contributor to late-life depression.
In a recent review of studies exploring social relations and depression in
late life, many of the factors already discussed were confirmed. By
considering factors across multiple domains, the investigators found that
social support, quality of social relations, and the presence of confidants
were the factors significantly associated with depression. In contrast the
quantitative aspects of social relations, such as frequency of contact, were
more inconsistent in association (Schwarzbach et al. 2014).
How does the clinician or investigator resolve the discrepancy between a
relatively lower frequency of major depression in the older adult
community with increased biological vulnerability, and perhaps only
slightly better social resources, compared with younger adults?
Psychological factors may modify the biological and social risks for
depression. Most older adults, for example, who experience a significant
physical illness do not become depressed. Older people may in general
possess psychological strengths that actually protect them from the onset of
clinically significant depressive symptoms. Two such potential strengths are
described below.
Socioemotional selectivity theory may explain differences across the life
cycle in the experience of events that lead to depression (Carstensen et al.
2000). The theory focuses on the perception by older individuals of time
left in life, rather than on past experiences. Younger adults have much to
learn and relatively long futures over which to learn. They are motivated by
pursuit of knowledge, even when this requires that they suppress emotional
well-being. In contrast, older individuals perceive that they have lived
longer than they should live and therefore deemphasize negative experience
and prioritize emotionally meaningful goals. In one study, negative
emotional experiences (e.g., the perception of stressors) declined from
young adulthood until around age 60 years (Carstensen et al. 2000). Periods
of highly positive emotional experience endured as meaningful among older
adults compared with younger adults.
Adults are also thought to acquire increased wisdom as they age.
Wisdom is a nebulous concept. Investigators with the Berlin Aging Group,
however, have operationalized wisdom and studied it in community
samples (Baltes and Staudinger 2000). Wisdom is an expert knowledge
system concerning the fundamental pragmatics of life, including knowledge
and judgment about the meaning and conduct of life and the orchestrating
of human development toward excellence while attending conjointly to
personal and collective well-being. Five criteria can be used to assess
wisdom: rich factual knowledge; rich procedural knowledge (e.g., the
ability to develop strategies for addressing problems); lifespan
contextualization (e.g., integrating life experiences); relativism of values
and life priorities (e.g., tolerance for differences in society); and recognition
and management of uncertainty (accepting that the future cannot be known
with certainty and that the ability to assess one’s sociocultural environment
is inherently constrained). Wisdom is thought to accumulate over the life
cycle if severe physical illness and cognitive impairment do not intervene.
Cumulative wisdom over time should protect older individuals from
spiraling down into depression when confronted with a complex of negative
experiences.
Treatment
Treatment of depression in late life is four-pronged, involving
psychotherapy, pharmacotherapy, neurostimulation, and family therapy.
These four approaches are discussed in this section.
Psychotherapy
Cognitive-behavioral therapy (CBT) is the only psychotherapy that was
designed specifically to treat depression (Beck 1967). Even the more
recently developed technique of interpersonal therapy is primarily a
cognitive-behavioral orientation to improving interpersonal relationships
(Klerman et al. 1984). The advantage of using CBT in treating the older
adult is that the therapy is directive and time limited, usually involving
between 10 and 25 sessions. CBT has been found to be effective for
depressed elderly patients (Gallagher and Thompson 1982; Steuer et al.
1984) and for patients with chronic medical illnesses such as type II
diabetes (Lustman et al. 1998), heart disease (Kohn et al. 2000), and
irritable bowel syndrome (Boyce et al. 2000). It may be particularly useful
for patients who show only a partial response to antidepressant drug therapy
(Scott et al. 2000).
The goal of behavioral and cognitive therapies is to change behavior and
modes of thinking. This change is accomplished through behavioral
interventions such as weekly activity schedules, mastery and pleasure logs,
and graded task assignments. Cognitive approaches to the restructuring of
negative cognitions or automatic thoughts include subjecting these
cognitions to empirical reality testing, examining distortions (e.g.,
overgeneralizations, catastrophizing, dichotomous thinking), and generating
new ways of viewing one’s life (Steuer et al. 1984). Depressed patients
typically regard themselves and their present and future in somewhat
idiosyncratic or negative ways. Such patients believe that they are
inadequate or defective and think that unpleasant experiences are caused by
a problem with themselves and that they are therefore worthless, helpless,
and hopeless. This cognitive triad leads the older adult to believe that he or
she has a never-ending depression and that nothing pleasant will ever
happen again. The cognitive model presupposes that these symptoms of
depression are consequences of negative thinking patterns.
Thompson et al. (1987) randomly assigned 91 elderly individuals with
major depression to cognitive therapy, behavioral therapy, or brief dynamic
therapy (the latter stresses the importance of the patient-therapist
relationship and emphasizes realistic collaborative aspects of the
therapeutic alliance). Patients in each group underwent 16–20 sessions of
therapy conducted by expert clinicians; 20 additional patients were assigned
to a waiting-list control group. By the end of 6 weeks, 52% of the patients
in therapy were in complete remission, and 18% showed significant
improvement. All therapies were equally efficacious and superior to waiting
for treatment.
Results of empirical studies suggest that compared with control subjects,
elders who engage in psychotherapy experience incremental improvement.
Not only does the percentage of elderly individuals who respond to these
treatments compare favorably with the percentage of younger subjects who
respond, the degree of improvement appears equal to that obtained with
medications, especially for individuals with milder forms of depression.
Drug therapy is not appropriate for some elders, and cognitive therapy,
behavioral therapy, and brief dynamic psychotherapy are viable alternatives.
In addition, evidence has emerged that suggests that the long-term benefit
of CBT may be greater than that of pharmacotherapy, especially if the
medications are discontinued during the first year of treatment (Reynolds et
al. 1999).
Older adults who have minor depression or adjustment disorders, or who
experience dysphoria because of losses of various types, often require less
intensive forms of psychotherapy. Active listening and simple support may
be sufficient to help distressed elders cope with their situation. Because
religion is an important factor in the lives of many older adults, referral to a
pastoral counselor may be particularly helpful and acceptable (Koenig et al.
2004).
Pharmacotherapy
The use of selective serotonin reuptake inhibitors (SSRIs) has been growing
in elderly patients (with or without medical illness). Citalopram (Nyth and
Gottfries 1990), escitalopram (Gorwood et al. 2007), fluoxetine
(Heiligenstein et al. 1995), paroxetine (Bump et al. 2001), and sertraline
(Cohn et al. 1990) have been shown to be effective in treating geriatric
depression. SSRIs have also proved effective in depressed older adults who
have had a stroke (Cole et al. 2001) or who have vascular disease in general
(Krishnan et al. 2001) or Alzheimer’s disease (Lyketsos et al. 2000). These
agents have become the drugs of first choice for treating mild to moderate
forms of depression. Important advantages of the use of these drugs in
treating elderly patients are the lack of anticholinergic, orthostatic, and
cardiac side effects; lack of sedation; and safety in overdose. Nevertheless,
for a significant number of older adults, SSRIs cause other unacceptable
effects, including excessive activation and disturbance of sleep, tremor,
headache, significant gastrointestinal side effects, hyponatremia, and weight
loss.
Other agents that affect both the serotonergic and noradrenergic systems
are often considered the best second-line therapy if the patient’s response to
an SSRI is not adequate. Duloxetine (Raskin et al. 2008) and venlafaxine
(Staab and Evans 2000) have been shown to be effective in treating geriatric
depression.
TCAs are the agents of choice for some patients with more severe forms
of major depression who can tolerate the side effects and do not respond to
the medications mentioned above. Medications that are effective yet
relatively free of side effects (especially cardiovascular effects) are
preferred. In recent years, nortriptyline and desipramine have become the
more popular medications for treating older adults with endogenous or
melancholic major depression. However, doxepin remains a favorite among
many practitioners. It is recommended that all elderly patients have an
electrocardiogram (ECG) before initiation of treatment and again after
therapeutic blood levels have been achieved. If the ECG shows a second-
degree (or higher) block, a bifascicular bundle branch block, a left bundle
branch block, or a QTc interval greater than 480 milliseconds, treatment
with TCAs should not be initiated or should be stopped in patients already
taking these medications (Caravati and Bossart 1991; Stoudemire and
Atkinson 1988).
Antidepressant doses administered to persons in late life should be case
specific but are generally lower than those given to persons in midlife. As
shown in Table 9–4, starting therapeutic daily doses of antidepressants are
as follows: citalopram, 10–40 mg; fluoxetine, 5–20 mg; paroxetine, 10–30
mg; sertraline, 12.5–50 mg; mirtazapine, 7.5–30 mg; and venlafaxine, 37.5–
200 mg (in divided doses). With regard to TCAs, 25 mg of desipramine
orally twice a day or 25–50 mg of nortriptyline orally at bedtime is
frequently adequate for relieving depressive symptoms. Plasma levels of
TCAs can be helpful in determining dosing: desipramine levels greater than
125 ng/mL and nortriptyline levels of 50–150 ng/mL have been found to be
therapeutic.
Trazodone and bupropion (Weihs et al. 2000) are alternatives in patients
who cannot tolerate TCAs, SSRIs, or mixed agents like venlafaxine.
Trazodone has advantages over TCAs in that it is virtually free of
anticholinergic effects, and it has advantages over the newer antidepressants
in that it has strong sedative effects. Nevertheless, the drug is not without
side effects, including excessive daytime sedation, priapism (occasionally),
and significant orthostatic hypotension. The therapeutic daily dose of
trazodone is 300 mg or more, an amount that many older patients cannot
tolerate because of sedation. Bupropion can be effective in treating
depression in elderly people but generally is used once other medications
have proved ineffective. Bupropion therapy should be initiated at 75 mg
twice daily, with an increase to 150 mg twice daily (not to exceed 150 mg
in a single dose). Agitation is the most common side effect that troubles
older individuals. Two newer agents, vilazodone and vortioxetine, have
limited data in elderly patients. For both medications, no dose adjustment
on the basis of age is recommended.
Monoamine oxidase inhibitors (MAOIs) are another alternative to TCAs,
SSRIs, and mixed serotonergic/noradrenergic antidepressants. An important
consideration is that if MAOIs are being considered because of intolerance
to side effects of other antidepressants, older adults usually do not tolerate
MAOIs any better. If treatment with an MAOI is to follow treatment with
an SSRI, a minimum of 1–2 weeks (or 2–4 weeks following fluoxetine)
must elapse after discontinuation of SSRI therapy before initiation of
MAOI therapy, to avoid a serotonergic syndrome. If a patient’s depression
is severe and ECT is contemplated, use of an MAOI also precludes
initiation of ECT until 10–14 days after the drug is discontinued. Such a
delay may seriously impede clinical management of the suicidal elder.
Neurostimulation
Electroconvulsive Therapy
ECT continues to be the most effective form of treatment for individuals
with more severe major depressive episodes (O’Connor et al. 2001). The
induction of a seizure via ECT appears to be effective in reversing a major
depression. ECT was first established as a treatment in 1938, but it is not
used as much as it was immediately after its development. Despite its
effectiveness, ECT is not the first-line treatment of choice for a patient with
major depression and should be prescribed only because other therapeutic
modalities have been ineffective. ECT has been shown to be effective in
selected individuals, primarily those who have major depression with
melancholia, and especially those who have major depression with
psychotic symptoms associated with agitation or withdrawal. Many older
adults with such syndromes either fail to respond to antidepressant
medications or experience toxicity (usually postural hypotension) when
taking antidepressants. The presence of self-destructive behavior, such as a
suicide attempt or refusal to eat, increases the necessity for intervening
effectively; in such situations, ECT may be the treatment of choice.
If ECT is selected as an intervention, the clinician must first discuss in
detail with the patient and the family the nature of the treatment and the
reasons for this recommendation. It is important to explain why ECT is
necessary; what procedures the patient will undergo during a course of
ECT; how many treatments can be expected; how long hospitalization will
continue; whether ECT can be performed on an outpatient basis; what the
risks and side effects of ECT are; and what results, both immediate and
long-term, can be expected. Even when an elderly patient is severely
depressed, careful and thoughtful discussion with the patient and family
will usually result in a willingness by the patient (often with encouragement
from the family) to undergo the course of ECT treatments. Once treatment
is begun, fears of ECT usually remit.
The medical workup before ECT includes acquisition of a complete
medical history, a physical examination, and consultation with a
cardiologist if any cardiac abnormalities are recognized. Knowledge of any
family history of psychiatric disorders, suicide, or treatment with ECT is
helpful in predicting a patient’s response to treatment. Laboratory
examination includes a complete blood count, a urinalysis, routine
chemistries, chest and spinal X rays (the latter to document previous
compression fractures), an ECG, and a computed tomography (CT) scan or
MRI (with CT or MRI available, an electroencephalogram and skull X ray
are not routinely required). The presence of some abnormalities seen in
magnetic resonance images does not militate against the use of ECT,
however. For example, a series of older adults with major depression were
found to have subcortical arteriosclerotic encephalopathy, as demonstrated
by MRI, but promptly improved after undergoing ECT (Coffey et al. 1987).
Before an older individual undergoes ECT, all medications should be
withdrawn, if possible. As noted in the pharmacotherapy section earlier in
this chapter, any MAOIs must be withdrawn 10–14 days before the
procedure to prevent any toxic interactions with the anesthetic used during
ECT. Reserpine and anticholinesterase drugs should also be withdrawn for
at least 1 week prior to ECT. Lithium carbonate, TCAs, antipsychotics, and
antianxiety agents (including sedative-hypnotics) are not absolutely
contraindicated in older patients who are to undergo ECT; however,
benzodiazepines increase the seizure threshold and should be avoided.
Generally, a short-acting barbiturate, such as chloral hydrate (500 mg orally
at bedtime), is the most appropriate sedative-hypnotic, although chloral
hydrate should not be given on the night preceding administration of ECT,
if possible. Use of low-dose haloperidol or thiothixene is probably the most
appropriate means of controlling severe agitation or psychotic symptoms
during the course of ECT treatment.
The basic techniques for ECT are well described. Thirty minutes before
treatment, an anticholinergic agent is administered intramuscularly to
prevent complications of cardiac arrhythmias and aspiration. Directly
before treatment, a short-acting anesthetic, such as thiopental or
methohexital, is administered until an eyelash response is no longer present.
Then a muscle relaxant, such as succinylcholine, is administered to prevent
severe muscle contractions. Investigators are increasingly using unilateral
electrode placement to the nondominant cerebral hemisphere, because
evidence has accumulated that less confusion occurs after unilateral
treatment than after bilateral treatment. Nevertheless, unilateral electrode
placement does not preclude development of memory difficulties. (Some
investigators question the efficacy of unilateral versus bilateral electrode
placement, but bilateral electrode placement has not been clearly
established as therapeutically superior to unilateral electrode placement.)
The electrical stimulus is applied, and the seizure is monitored either by
applying a tourniquet to one arm and observing the tonic and clonic
movements in the extremity peripheral to the tourniquet or by using direct
electroencephalographic monitoring. Direct electroencephalographic
monitoring is preferred, and a seizure lasting 25 seconds or more is required
for optimal results.
Seizure duration varies with age. In a study involving 228 patients
treated with ECT, Hinkle et al. (1986) found that of patients older than age
60 years, a greater percentage were likely to have a seizure of 30 seconds or
less. When ECT is repeated, use of caffeine may increase the likelihood of
inducing a seizure without the necessity of restimulation using higher
electrical parameters (which could lead to increased central nervous system
toxicity).
ECT treatments are generally administered three times per week, and
usually 6–12 treatments are necessary for adequate therapeutic response. A
clear improvement is often noted after one of the treatments, with the
patient reporting a remarkable improvement in mood and functioning. Two
or three treatments are generally given after the ECT administration that
leads to improvement.
The risks and side effects of ECT in elderly patients are similar to those
in the general population. Cardiovascular effects are of greatest concern and
include premature ventricular contractions, ventricular arrhythmias, and
transient systolic hypertension. Frequent monitoring during treatment
decreases the (already low) risk that one of these side effects will lead to
permanent problems. Confusion and amnesia often result after a treatment,
but the duration of this confusional episode is brief. Even with the use of
unilateral nondominant treatment, however, some patients have prolonged
memory difficulties. Headaches are a common symptom with ECT; they
usually respond to nonnarcotic analgesics. Status epilepticus and vertebral
compression fractures are some of the rare but more serious adverse effects.
Compression fractures are a particular risk in older women because of the
high incidence of osteoporosis in the postmenopausal population.
The overall success rate of ECT in patients who have not responded to
drug therapy is usually 80% or greater, and there is no evidence that
effectiveness is lower in older adults (Avery and Lubrano 1979). Wesner
and Winokur (1989) examined the influence of age on the natural history of
major depressive disorder and found that ECT reduced the rate of chronicity
when it was used in patients age 40 or older but, surprisingly, not in those
younger than age 40 years.
A review of records in Canada from 1992 to 2004 found that rates of
ECT use in elderly patients increased during the 1990s but then stabilized at
approximately 12.5 persons per year per 100,000 in the population
(Rapoport et al. 2006). Rosenbach et al. (1997) reported that the number of
beneficiaries receiving ECT increased from 12,000 in 1987 to 15,560 in
1992, an increase of more than 20% (after calculations were adjusted for
increased numbers of beneficiaries between 1987 and 1992). In a
prospective, multisite study, Tew et al. (1999) compared characteristics and
treatment outcomes of 133 adult (age 59 or younger), 63 young-old (ages
60–74 years), and 72 old-old (age 75 or older) patients treated with ECT for
major depression. They found that patients less than 60 years old had a
significantly lower rate of response to ECT (54%) than did young-old
patients (73%) or old-old patients (67%). The investigators concluded that
despite a higher level of physical illness and cognitive impairment, patients
age 75 or older who had severe major depression tolerated ECT in a manner
similar to the way in which younger patients tolerated the treatment, and the
old-old patients demonstrated a similar or even better response. There is
also evidence that ECT may be more effective and have fewer side effects
than antidepressants when used to treat depression in old-old patients
(Manly et al. 2000).
The relapse rate with no prophylactic intervention may exceed 50% in
the year after a course of ECT. This relapse rate can be decreased if
antidepressants or lithium carbonate is prescribed after the treatment.
Maintenance ECT may be necessary for some patients who exhibit a high
likelihood of recurrence despite use of prophylactic medication and/or who
experience high toxicity and therefore cannot tolerate prophylactic
medications. For such patients, weekly or monthly treatments (usually on
an outpatient basis) are prescribed, with careful monitoring of response and
side effects. Following an effective course of ECT, the combination of
continuation ECT and antidepressant drug therapy has been shown to have
greater efficacy than use of medications alone (Gagné et al. 2000).
Despite the effectiveness of ECT, few deny that treatment may lead to
memory difficulties. In a study by Frith et al. (1983), 70 severely depressed
patients were randomly assigned to eight real or sham ECT treatments and
were divided according to the degree of recovery from depression
afterward. Compared with nondepressed control subjects, the depressed
patients were impaired on a wide range of tests of memory and
concentration before treatment, but their performance on most tests
improved after treatment. Real ECT induced impairments in concentration,
short-term memory, and learning but significantly facilitated access to
remote memories. At 6-month follow-up, all differences between real and
sham ECT groups had disappeared.
Price and McAllister (1989) examined the efficacy of ECT in elderly
depressed patients with dementia. Overall, the patients achieved an 86%
response rate, with only 21% experiencing a significant worsening of
cognition; the cognition problems were transient in most cases. Of
particular importance is that 49% of the patients treated with ECT showed
improvement in memory function after treatment. Likewise, Stoudemire et
al. (1995) found that over time, ECT may lead to significant improvement
in memory of cognitively impaired older adults with depression. Although
data on the safety and efficacy of ECT in patients with concurrent medical
illness derive primarily from retrospective studies involving psychiatric
patients with stable disease, these data do support the use of ECT in patients
with cardiovascular, neurological, endocrine, or metabolic conditions, as
well as a variety of other conditions (Stoudemire et al. 1998).
Repetitive Transcranial Magnetic Stimulation
A recent advance in clinical neurostimulation is repetitive transcranial
magnetic stimulation (rTMS). As summarized in a review paper (Aleman
2013), rTMS involves generation of a time-varying magnetic field by a
current pulse through a simulator coil placed over the scalp, inducing
electric current intracranially, and thus neural activation. The intensity of
stimulation is set at a fixed percentage of the individual’s motor threshold,
measured as the output required to produce movement of thumb or fingers.
When generally agreed-upon guidelines for stimulation are followed, rTMS
appears to be a safe and well-tolerated treatment, with minimal risk of
inducing an epileptic seizure. When used to treat depression, the rTMS
stimulus is placed over the prefrontal cortex, in most cases over the
dorsolateral prefrontal cortex. Typical treatment includes a 20- to 40-minute
session, 5 days a week for 4–8 weeks. In adult populations with major
depression, results with rTMS are mixed, with most studies favoring active
rTMS treatment over a sham or control condition (Hovington et al. 2013).
Although studies of rTMS in geriatric depression are limited, one study
of MRI-defined vascular depression in elderly patients used active versus
sham rTMS (Jorge et al. 2008). The older depressed patients receiving
active stimulation experienced significantly greater improvement in
depression severity and had higher response rates compared with those
receiving sham rTMS. The investigators found that greater left and right
gray matter volumes were associated with response to rTMS.
Family Therapy
The final component of therapy for the depressed elderly patient is work
with the family. Not only may family dysfunction contribute to the
depressive symptoms experienced by the older individual, but family
support is critical to a successful outcome in the treatment of the depressed
elderly patient. A clinician must attend to 1) those members of the family
who will be available to the elderly person; 2) the frequency and quality of
interactions between the older adult and family members, as well as among
other family members; 3) the overall family atmosphere; 4) family values
regarding psychiatric disorders; 5) family support and tolerance of
symptoms (such as expressions of wishing not to live); and 6) stressors
encountered by the family other than the depression experienced by the
elder (Blazer 2002).
Most depressed elders do not resist interaction between the clinician and
family members. With the patient’s permission, the family should be
instructed regarding the nature of the depressive disorder and the potential
risks associated with depression in late life, especially suicide. Family
members can assist the clinician in observing changes in the patient’s
behavior, such as an increase in discomfort (either physical or emotional),
increased withdrawal and decreased verbalization, and preoccupation with
medications or weapons. The family can assist by removing possible
implements of suicide from places of easy access. The family can also take
responsibility for administering medications to an older adult who is
unreliable or whose potential for suicide is high.
Family members can benefit from simple instructions regarding how to
communicate with an elderly depressed patient. Methods of responding to
expressions of low self-esteem and pessimism, such as paraphrase and
expression of understanding without a sense of responsibility to intervene,
can be especially effective. Families can be taught, for example, to
acknowledge to the patient, “I hear what you are saying, and I understand.”
Behavioral techniques for dealing with demanding or overly dependent
elders can be taught to families as well. A depressed elder’s demand for
constant attention from a family member may necessitate “weaning” the
patient from continued contact.
When the symptoms of depression become so severe that hospitalization
is required, family members are valuable in facilitating hospitalization.
Without a proper alliance between clinician and family, a family may be
resistant to hospitalization and undermine the clinician’s attempts to treat
the older adult appropriately. It is usually necessary for the clinician to take
responsibility for saying that hospitalization is essential—that the situation
has reached the point at which the family has no choice. The clinician
informs the patient—in the presence of the family—of the necessity of
hospitalization, and the family in turn can support the clinician’s position.
In such a situation, the patient rarely resists hospitalization for long.
Conclusion
Depressive disorders are among the most frequent clinical problems
encountered by those who care for older adults. We perhaps know more
about these disorders in older adults, especially their successful treatment,
than any other late-life psychiatric disorders. In addition, we have improved
diagnostic capabilities and better treatments (especially treatments used in
combination) than we did 30 years ago. It is not an understatement to
suggest that where there is depression in later life, there is hope.
Key Points
• Late-life depression overall may not be as frequent as at other stages of
the life cycle, yet the frequency is much higher in physically and
cognitively impaired older adults than in community-based samples.
• The biopsychosocial model works well in placing the origins of late-life
depression in context. Most cases derive from a variety of causes.
• Older adults appear more vulnerable to biological causes of depression,
such as depression secondary to vascular lesions in the brain.
• Social causes of depression in older adults do not appear to be more
frequent but differ from social causes of depression in young or middle-
aged adults.
• Older adults who are cognitively intact may experience a buffering of
depression because of a lifetime of cumulative wisdom coupled with a
different view of events given their age.
• The diagnostic workup of the depressed older adult is centered on a
detailed history, ideally from the patient and family.
• For moderately severe depression, a combination of antidepressant
therapy and psychotherapy (e.g., interpersonal therapy) is optimal.
• Electroconvulsive therapy is indicated for more severe and treatment-
resistant depressive disorders in late life and is generally well tolerated.
References
Aleman A: Use of repetitive transcranial magnetic stimulation for treatment
in psychiatry. Clin Psychopharmacol Neurosci 11(2):53–59, 2013
24023548
Alexopoulos GS, Meyers BS, Young RC, et al: The course of geriatric
depression with “reversible dementia”: a controlled study. Am J
Psychiatry 150(11):1693–1699, 1993 8105707
Alexopoulos GS, Meyers BS, Young RC, et al: Recovery in geriatric
depression. Arch Gen Psychiatry 53(4):305–312, 1996 8634008
Ambelas A: Life events and mania. A special relationship? Br J Psychiatry
150:235–240, 1987 3651678
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Arborelius L, Owens MJ, Plotsky PM, et al: The role of corticotropin-
releasing factor in depression and anxiety disorders. J Endocrinol
160(1):1–12, 1999 9854171
Arfken CL, Lichtenberg PA, Tancer ME: Cognitive impairment and
depression predict mortality in medically ill older adults. J Gerontol A
Biol Sci Med Sci 54(3):M152–M156, 1999 10191844
Atchley RC: A continuity theory of normal aging. Gerontologist 29(2):183–
190, 1989 2519525
Avery D, Lubrano A: Depression treated with imipramine and ECT: the
DeCarolis study reconsidered. Am J Psychiatry 136(4B):559–562,
1979 426143
Baldwin RC, Jolley DJ: The prognosis of depression in old age. Br J
Psychiatry 149(5): 574–583, 1986 3814949
Baltes PB, Staudinger UM: Wisdom: a metaheuristic (pragmatic) to
orchestrate mind and virtue toward excellence. Am Psychol 55(1):122–
136, 2000 11392856
Beck AT: Depression: Clinical, Experimental, and Theoretical Aspects.
New York, Hoeber, 1967
Beekman AT, Deeg DJ, van Tilburg T, et al: Major and minor depression in
later life: a study of prevalence and risk factors. J Affect Disord 36(1–
2):65–75, 1995 8988267
Beekman AT, Deeg DJ, Geerlings SW, et al: Emergence and persistence of
late life depression: a 3-year follow-up of the Longitudinal Aging
Study Amsterdam. J Affect Disord 65(2):131–138, 2001 11356236
Beekman AT, Geerlings SW, Deeg DJ, et al: The natural history of late-life
depression: a 6-year prospective study in the community. Arch Gen
Psychiatry 59(7):605–611, 2002 12090813
Black SA: Increased health burden associated with comorbid depression in
older diabetic Mexican Americans. Results from the Hispanic
Established Population for the Epidemiologic Study of the Elderly
survey. Diabetes Care 22(1):56–64, 1999 10333904
Black SA, Goodwin JS, Markides KS: The association between chronic
diseases and depressive symptomatology in older Mexican Americans.
J Gerontol A Biol Sci Med Sci 53A(3):M188–M194, 1998 9597050
Blazer D: Social support and mortality in an elderly community population.
Am J Epidemiol 115(5):684–694, 1982 7081200
Blazer D: Impact of late-life depression on the social network. Am J
Psychiatry 140(2):162–166, 1983 6849426
Blazer D: Psychiatry and the oldest old. Am J Psychiatry 157(12):1915–
1924, 2000 11097951
Blazer D: Depression in Late Life, 3rd Edition. New York, Springer, 2002
Blazer D: Depression in late life: review and commentary. J Gerontol A
Biol Sci Med Sci 58(3):M249–M265, 2003 12634292
Blazer D, Bachar JR, Hughes DC: Major depression with melancholia: a
comparison of middle-aged and elderly adults. J Am Geriatr Soc
35(10):927–932, 1987a 3655175
Blazer D, Hughes DC, George LK: The epidemiology of depression in an
elderly community population. Gerontologist 27(3): 281–287, 1987b
3609795
Blazer D, Hughes DC, Fowler N: Anxiety as an outcome symptom of
depression in elderly and middle-aged adults. Int J Geriatr Psychiatry
4(5):273–278, 1989
Blazer D, Burchett B, Service C, et al: The association of age and
depression among the elderly: an epidemiologic exploration. J Gerontol
46(6):M210–M215, 1991 1834726
Blazer D, Hybels CF, Pieper CF: The association of depression and
mortality in elderly persons: a case for multiple, independent pathways.
J Gerontol A Biol Sci Med Sci 56(8):M505–M509, 2001 11487603
Blazer D, Burchett BB, Fillenbaum GG: APOE epsilon4 and low
cholesterol as risks for depression in a biracial elderly community
sample. Am J Geriatr Psychiatry 10(5):515–520, 2002a 12213685
Blazer DG, Moody-Ayers S, Craft-Morgan J, et al: Depression in diabetes
and obesity: racial/ethnic/gender issues in older adults. J Psychosom
Res 53(4):913–916, 2002b 12377303
Blazer DG 2nd, Hybels CF: Origins of depression in later life. Psychol Med
35(9): 1241–1252, 2005 16168147
Boyce P, Gilchrist J, Talley NJ, et al: Cognitive-behaviour therapy as a
treatment for irritable bowel syndrome: a pilot study. Aust NZ J
Psychiatry 34(2):300–309, 2000 10789535
Braam AW, Beekman AT, Deeg DJ, et al: Religiosity as a protective or
prognostic factor of depression in later life; results from a community
survey in the Netherlands. Acta Psychiatr Scand 96(3):199–205, 1997a
9296551
Braam AW, Beekman AT, van Tilburg TG, et al: Religious involvement and
depression in older Dutch citizens. Soc Psychiatry Psychiatr Epidemiol
32(5):284–291, 1997b 9257519
Breitbart W, Rosenfeld B, Pessin H, et al: Depression, hopelessness, and
desire for hastened death in terminally ill patients with cancer. JAMA
284(22):2907–2911, 2000 11147988
Bridges P: The drug treatment of depression in old age, in Affective
Disorders in the Elderly. Edited by Murphy E. Edinburgh, Scotland,
Churchill Livingstone, 1986, pp 91–149
Brodaty H, Luscombe G, Parker G, et al: Early and late onset depression in
old age: different aetiologies, same phenomenology. J Affect Disord
66(2–3):225–236, 2001 11578676
Brown KW, Levy AR, Rosberger Z, et al: Psychological distress and cancer
survival: a follow-up 10 years after diagnosis. Psychosom Med
65(4):636–643, 2003 12883115
Bruce ML: Depression and disability in late life: directions for future
research. Am J Geriatr Psychiatry 9(2):102–112, 2001 11316615
Bruce ML: Psychosocial risk factors for depressive disorders in late life.
Biol Psychiatry 52(3):175–184, 2002 12182924
Bruce ML, Leaf PJ: Psychiatric disorders and 15-month mortality in a
community sample of older adults. Am J Public Health 79(6):727–730,
1989 2786347
Bukberg J, Penman D, Holland JC: Depression in hospitalized cancer
patients. Psychosom Med 46(3):199–212, 1984 6739680
Bump GM, Mulsant BH, Pollock BG, et al: Paroxetine versus nortriptyline
in the continuation and maintenance treatment of depression in the
elderly. Depress Anxiety 13(1):38–44, 2001 11233459
Callahan CM, Nienaber NAS, Hendrie HC, et al: Depression of elderly
outpatients: primary care physicians’ attitudes and practice patterns. J
Gen Intern Med 7(1):26–31, 1992 1548545
Caravati EM, Bossart PJ: Demographic and electrocardiographic factors
associated with severe tricyclic antidepressant toxicity. J Toxicol Clin
Toxicol 29(1):31–43, 1991 2005664
Carney CE, Moss TG, Lachowski AM, et al: Understanding mental and
physical fatigue complaints in those with depression and insomnia.
Behav Sleep Med 12(4):272–289, 2014 24128300
Carstensen LL, Pasupathi M, Mayr U, et al: Emotional experience in
everyday life across the adult life span. J Pers Soc Psychol 79(4):644–
655, 2000 11045744
Center for Multicultural Mental Health Research: 2011 Data provided to the
IOM committee, Cambridge Health Alliance and Harvard Medical
School, Somerville, MA, Nov 2011, in The Mental Health and
Substance Use Workforce for Older Adults: In Whose Hands? Edited
by Eden J, Le M, Maslow K, et al. Washington, DC, National
Academies Press, 2012
Chi I, Chou KL: Social support and depression among elderly Chinese
people in Hong Kong. Int J Aging Hum Dev 52(3):231–252, 2001
11407488
Clayton PJ: Bereavement and depression. J Clin Psychiatry 51(suppl):34–
38, discussion 39–40, 1990 2195011
Coffey CE, Hinkle PE, Weiner RD, et al: Electroconvulsive therapy of
depression in patients with white matter hyperintensity. Biol Psychiatry
22(5):629–636, 1987 3580437
Coffey CE, Figiel GS, Djang WT, et al: Subcortical hyperintensity on
magnetic resonance imaging: a comparison of normal and depressed
elderly subjects. Am J Psychiatry 147(2):187–189, 1990 2301657
Cohn CK, Shrivastava R, Mendels J, et al: Double-blind, multicenter
comparison of sertraline and amitriptyline in elderly depressed patients.
J Clin Psychiatry 51(suppl B):28–33, 1990 2258379
Cole MG, Bellavance F, Mansour A: Prognosis of depression in elderly
community and primary care populations: a systematic review and
meta-analysis. Am J Psychiatry 156(8):1182–1189, 1999 10450258
Cole MG, Elie LM, McCusker J, et al: Feasibility and effectiveness of
treatments for post-stroke depression in elderly inpatients: systematic
review. J Geriatr Psychiatry Neurol 14(1):37–41, 2001 11281315
Cornoni-Huntley J, Blazer D, Lafferty M, et al (eds): Established
Populations for Epidemiologic Studies of the Elderly: Resource Data
Book, Vol 2. Bethesda, MD, National Institute on Aging, 1990
Covinsky KE, Kahana E, Chin MH, et al: Depressive symptoms and 3-year
mortality in older hospitalized medical patients. Ann Intern Med
130(7):563–569, 1999 10189325
Cutler NR, Post RM: Life course of illness in untreated manic-depressive
patients. Compr Psychiatry 23(2):101–115, 1982 7067425
Czéh B, Michaelis T, Watanabe T, et al: Stress-induced changes in cerebral
metabolites, hippocampal volume, and cell proliferation are prevented
by antidepressant treatment with tianeptine. Proc Natl Acad Sci USA
98(22):12796–12801, 2001 11675510
de Beurs E, Beekman A, Geerlings S, et al: On becoming depressed or
anxious in late life: similar vulnerability factors but different effects of
stressful life events. Br J Psychiatry 179(5):426–431, 2001 11689400
Devanand DP: Comorbid psychiatric disorders in late life depression. Biol
Psychiatry 52(3):236–242, 2002 12182929
Devanand DP, Turret N, Moody BJ, et al: Personality disorders in elderly
patients with dysthymic disorder. Am J Geriatr Psychiatry 8(3):188–
195, 2000 10910415
Devanand DP, Kim MK, Paykina N, et al: Adverse life events in elderly
patients with major depression or dysthymic disorder and in healthy-
control subjects. Am J Geriatr Psychiatry 10(3):265–274, 2002
11994213
Diniz BS, Butters MA, Albert SM, et al: Late-life depression and risk of
vascular dementia and Alzheimer’s disease: systematic review and
meta-analysis of community-based cohort studies. Br J Psychiatry
202(5):329–335, 2013 23637108
Dunner DL: Affective disorder: clinical features, in Psychiatry, Vol 1.
Edited by Michels R, Cavenar J. Philadelphia, JB Lippincott, 1985, pp
59–60
Eastham JH, Jeste DV, Young RC: Assessment and treatment of bipolar
disorder in the elderly. Drugs Aging 12(3):205–224, 1998 9534021
Eisses AM, Kluiter H, Jongenelis K, et al: Risk indicators of depression in
residential homes. Int J Geriatr Psychiatry 19(7):634–640, 2004
15254919
Fassler LB, Gaviria M: Depression in old age. J Am Geriatr Soc
26(10):471–475, 1978 701698
Fillenbaum G: Multidimensional Functional Assessment of Older Adults:
The Duke Older Americans Resources and Services Procedures.
Hillsdale, NJ, Erlbaum, 1988
Fiske A, Gatz M, Pedersen NL: Depressive symptoms and aging: the effects
of illness and non-health-related events. J Gerontol B Psychol Sci Soc
Sci 58(6):P320–P328, 2003 14614116
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Frasure-Smith N, Lespérance F, Talajic M: Depression following
myocardial infarction: impact on 6-month survival JAMA
270(15):1819–1825, 1993 8411525 [Erratum in: JAMA 271(14)]
Fried L: Frailty, in Principles of Geriatric Medicine and Gerontology, 3rd
Edition. Edited by Hazzard W, Bierman E, Blass J, et al. New York,
McGraw-Hill, 1994, pp 1149–1156
Frith CD, Stevens M, Johnstone EC, et al: Effects of ECT and depression
on various aspects of memory. Br J Psychiatry 142(6): 610–617, 1983
6882984
Gagné GGJr, Furman MJ, Carpenter LL, et al: Efficacy of continuation
ECT and antidepressant drugs compared to long-term antidepressants
alone in depressed patients. Am J Psychiatry 157(12):1960–1965, 2000
11097961
Gallagher DE, Thompson LW: Treatment of major depressive disorder in
older adult outpatients with brief psychotherapies. Psychotherapy
19(4):482–490, 1982
Gallo JJ, Rabins PV, Lyketsos CG, et al: Depression without sadness:
functional outcomes of nondysphoric depression in later life. J Am
Geriatr Soc 45(5):570–578, 1997 9158577
Gatz M, Pedersen NL, Plomin R, et al: Importance of shared genes and
shared environments for symptoms of depression in older adults. J
Abnorm Psychol 101(4): 701–708, 1992 1430610
Geerlings SW, Beekman AT, Deeg DJ, et al: Duration and severity of
depression predict mortality in older adults in the community. Psychol
Med 32(4):609–618, 2002 12102375
George LK, Blazer DG, Hughes DC, et al: Social support and the outcome
of major depression. Br J Psychiatry 154(4):478–485, 1989 2590779
George MS, Ketter TA, Post RM: Prefrontal cortex dysfunction in clinical
depression. Depression 2(2):59–72, 1994
Glasser M, Rabins P: Mania in the elderly. Age Ageing 13(4):210–213,
1984 6475649
Glassman AH, Shapiro PA: Depression and the course of coronary artery
disease. Am J Psychiatry 155(1):4–11, 1998 9433332
González-Salvador MT, Arango C, Lyketsos CG, et al: The stress and
psychological morbidity of the Alzheimer patient caregiver. Int J
Geriatr Psychiatry 14(9):701–710, 1999 10479740
Gorwood P, Weiller E, Lemming O, Katona C: Escitalopram prevents
relapse in older patients with major depressive disorder. Am J Geriatr
Psychiatry 15(7):581–593, 2007 17586783
Gottfries CG: Neurochemical aspects on aging and diseases with cognitive
impairment. J Neurosci Res 27(4):541–547, 1990 2079715
Guttmann CR, Jolesz FA, Kikinis R, et al: White matter changes with
normal aging. Neurology 50(4):972–978, 1998 9566381
Hays JC, Saunders WB, Flint EP, et al: Social support and depression as
risk factors for loss of physical function in late life. Aging Ment Health
1(3):209–220, 1997
Hays JC, Steffens DC, Flint EP, et al: Does social support buffer functional
decline in elderly patients with unipolar depression? Am J Psychiatry
158(11):1850–1855, 2001 11691691
Heiligenstein JH, Ware JE Jr, Beusterien KM, et al: Acute effects of
fluoxetine versus placebo on functional health and well-being in late-
life depression. Int Psychogeriatr 7(S1 suppl):125–137, 1995 8580388
Henderson AS, Jorm AF, MacKinnon A, et al: The prevalence of depressive
disorders and the distribution of depressive symptoms in later life: a
survey using Draft ICD-10 and DSM-III-R. Psychol Med 23(3):719–
729, 1993 8234578
Henderson AS, Korten AE, Jacomb PA, et al: The course of depression in
the elderly: a longitudinal community-based study in Australia.
Psychol Med 27(1):119–129, 1997 9122292
Hickie I, Scott E, Naismith S, et al: Late-onset depression: genetic, vascular
and clinical contributions. Psychol Med 31(8):1403–1412, 2001
11722155
Hinkle P, Coffey CE, Weiner R, et al: ECT seizure duration varies with age.
Paper presented at the American Geriatrics Society, Chicago, IL, 1986
Holahan CK, Holahan CJ: Self-efficacy, social support, and depression in
aging: a longitudinal analysis. J Gerontol 42(1):65–68, 1987 3794199
Holahan CJ, Moos RH: Social support and psychological distress: a
longitudinal analysis. J Abnorm Psychol 90(4):365–370, 1981 7264067
Hovington CL, McGirr A, Lepage M, et al: Repetitive transcranial magnetic
stimulation (rTMS) for treating major depression and schizophrenia: a
systematic review of recent meta-analyses. Ann Med 45(4):308–321,
2013 23687987
Hughes DC, Blazer DG, George LK: Age differences in life events: a
multivariate controlled analysis. Int J Aging Hum Dev 27(3):207–220,
1988 3243654
Husain MM, McDonald WM, Doraiswamy PM, et al: A magnetic
resonance imaging study of putamen nuclei in major depression.
Psychiatry Res 40(2):95–99, 1991 1763144
Hybels CF, Blazer DG, Pieper CF: Toward a threshold for subthreshold
depression: an analysis of correlates of depression by severity of
symptoms using data from an elderly community sample.
Gerontologist 41(3):357–365, 2001 11405433
Hybels CF, Pieper CF, Blazer DG: Sex differences in the relationship
between subthreshold depression and mortality in a community sample
of older adults. Am J Geriatr Psychiatry 10(3):283–291, 2002
11994215
Hybels CF, Blazer DG, Steffens DC: Predictors of partial remission in older
patients treated for major depression: the role of comorbid dysthymia.
Am J Geriatr Psychiatry 13(8):713–721, 2005 16085788
Idler EL: Religious involvement and the health of the elderly: some
hypotheses and an initial test. Soc Forces 66(1):226–238, 1987
Jang Y, Haley WE, Small BJ, et al: The role of mastery and social resources
in the associations between disability and depression in later life.
Gerontologist 42(6):807–813, 2002 12451162
Jorge RE, Moser DJ, Acion L, et al: Treatment of vascular depression using
repetitive transcranial magnetic stimulation. Arch Gen Psychiatry
65(3):268–276, 2008 18316673
Keller MB, Shapiro RW, Lavori PW, et al: Recovery in major depressive
disorder: analysis with the life table and regression models. Arch Gen
Psychiatry 39(8):905–910, 1982a 7103679
Keller MB, Shapiro RW, Lavori PW, et al: Relapse in major depressive
disorder: analysis with the life table. Arch Gen Psychiatry 39(8):911–
915, 1982b 7103680
Kennedy GJ, Kelman HR, Thomas C: The emergence of depressive
symptoms in late life: the importance of declining health and increasing
disability. J Community Health 15(2):93–104, 1990 2141337
Kessler RC, Chiu WT, Demler O, et al: Prevalence, severity, and
comorbidity of 12-month DSM-IV disorders in the National
Comorbidity Survey Replication Arch Gen Psychiatry 62(6):617–627,
2005 15939839 [Erratum in: Arch Gen Psychiatry 62(7)]
Klerman GL, Weissman MM, Rounsaville BJ, et al: Interpersonal
Psychotherapy of Depression. New York, Basic Books, 1984
Koenig HG: Religion and depression in older medical inpatients. Am J
Geriatr Psychiatry 15(4):282–291, 2007a 17384313
Koenig HG: Religion and remission of depression in medical inpatients
with heart failure/pulmonary disease. J Nerv Ment Dis 195(5):389–395,
2007b 17502804
Koenig HG, Blazer DG: Epidemiology of geriatric affective disorders. Clin
Geriatr Med 8(2):235–251, 1992 1600475
Koenig HG, Meador KG, Cohen HJ, et al: Depression in elderly
hospitalized patients with medical illness. Arch Intern Med
148(9):1929–1936, 1988 3415405
Koenig HG, Goli V, Shelp F, et al: Antidepressant use in elderly medical
inpatients: lessons from an attempted clinical trial. J Gen Intern Med
4(6):498–505, 1989a 2685207
Koenig HG, Shelp F, Goli V, et al: Survival and health care utilization in
elderly medical inpatients with major depression. J Am Geriatr Soc
37(7):599–606, 1989b 2738278
Koenig HG, Cohen HJ, Blazer DG, et al: Religious coping and depression
among elderly, hospitalized medically ill men. Am J Psychiatry
149(12):1693–1700, 1992 1443246
Koenig HG, George LK, Peterson BL: Religiosity and remission of
depression in medically ill older patients. Am J Psychiatry 155(4):536–
542, 1998 9546001
Koenig HG, George LK, Titus P: Religion, spirituality, and health in
medically ill hospitalized older patients. J Am Geriatr Soc 52(4):554–
562, 2004 15066070
Kohn CS, Petrucci RJ, Baessler C, et al: The effect of psychological
intervention on patients’ long-term adjustment to the ICD: a
prospective study. Pacing Clin Electrophysiol 23(4 Pt 1):450–456,
2000 10793433
Kraaij V, de Wilde EJ: Negative life events and depressive symptoms in the
elderly: a life span perspective. Aging Ment Health 5(1):84–91, 2001
11513018
Kraaij V, Arensman E, Spinhoven P: Negative life events and depression in
elderly persons: a meta-analysis. J Gerontol B Psychol Sci Soc Sci
57(1):P87–P94, 2002 11773227
Krishnan KR, Goli V, Ellinwood EH, et al: Leukoencephalopathy in
patients diagnosed as major depressive. Biol Psychiatry 23(5):519–522,
1988 3345325 [Erratum in: Biol Psychiatry 25(6)]
Krishnan KR, McDonald WM, Escalona PR, et al: Magnetic resonance
imaging of the caudate nuclei in depression. Preliminary observations.
Arch Gen Psychiatry 49(7):553–557, 1992 1627046
Krishnan KR, McDonald WM, Doraiswamy PM, et al: Neuroanatomical
substrates of depression in the elderly. Eur Arch Psychiatry Clin
Neurosci 243(1):41–46, 1993 8399409
Krishnan KR, Hays JC, Blazer DG: MRI-defined vascular depression. Am J
Psychiatry 154(4):497–501, 1997 9090336
Krishnan KR, Doraiswamy PM, Clary CM: Clinical and treatment response
characteristics of late-life depression associated with vascular disease:
a pooled analysis of two multicenter trials with sertraline. Prog
Neuropsychopharmacol Biol Psychiatry 25(2):347–361, 2001
11294481
Krishnan KR, Taylor WD, McQuoid DR, et al: Clinical characteristics of
magnetic resonance imaging-defined subcortical ischemic depression.
Biol Psychiatry 55(4): 390–397, 2004 14960292
Kumar A, Mintz J, Bilker W, et al: Autonomous neurobiological pathways
to late-life major depressive disorder: clinical and pathophysiological
implications. Neuropsychopharmacology 26(2):229–236, 2002
11790518
Lenze EJ, Munin MC, Skidmore ER, et al: Onset of depression in elderly
persons after hip fracture: implications for prevention and early
intervention of late-life depression. J Am Geriatr Soc 55(1):81–86,
2007 17233689
Li YS, Meyer JS, Thornby J: Longitudinal follow-up of depressive
symptoms among normal versus cognitively impaired elderly. Int J
Geriatr Psychiatry 16(7):718–727, 2001 11466752
Liverman C, Blazer D (eds): Testosterone and Aging: Clinical Research
Directions. Washington, DC, National Academies Press, 2004
Livingston G, Manela M, Katona C: Depression and other psychiatric
morbidity in carers of elderly people living at home. BMJ
312(7024):153–156, 1996 8563534
Luisi S, Tonetti A, Bernardi F, et al: Effect of acute corticotropin releasing
factor on pituitary-adrenocortical responsiveness in elderly women and
men. J Endocrinol Invest 21(7):449–453, 1998 9766260
Lustman PJ, Griffith LS, Freedland KE, et al: Cognitive behavior therapy
for depression in type 2 diabetes mellitus. A randomized, controlled
trial. Ann Intern Med 129(8):613–621, 1998 9786808
Lyketsos CG, Sheppard JM, Steele CD, et al: Randomized, placebo-
controlled, double-blind clinical trial of sertraline in the treatment of
depression complicating Alzheimer’s disease: initial results from the
Depression in Alzheimer’s Disease study. Am J Psychiatry
157(10):1686–1689, 2000 11007727
Lyles KW: Osteoporosis and depression: shedding more light upon a
complex relationship. J Am Geriatr Soc 49(6):827–828, 2001
11454124
Lyness JM, Caine ED, King DA, et al: Depressive disorders and symptoms
in older primary care patients: one-year outcomes. Am J Geriatr
Psychiatry 10(3):275–282, 2002 11994214
Magaziner J, Simonsick EM, Kashner TM, et al: Predictors of functional
recovery one year following hospital discharge for hip fracture: a
prospective study. J Gerontol 45(3):M101–M107, 1990 2335719
Manly DT, Oakley SP Jr, Bloch RM: Electroconvulsive therapy in old-old
patients. Am J Geriatr Psychiatry 8(3):232–236, 2000 10910422
Massey EW, Bullock R: Peroneal palsy in depression. J Clin Psychiatry
39(4):287–292, 1978 580277
Meyers BS, Kalayam B, Mei-Tal V: Late-onset delusional depression: a
distinct clinical entity? J Clin Psychiatry 45(8):347–349, 1984 6746580
Mischoulon D, Burger JK, Spillmann MK, et al: Anemia and macrocytosis
in the prediction of serum folate and vitamin B12 status, and treatment
outcome in major depression. J Psychosom Res 49(3):183–187, 2000
11110989
Mojtabai R, Olfson M: Major depression in community-dwelling middle-
aged and older adults: prevalence and 2- and 4-year follow-up
symptoms. Psychol Med 34(4):623–634, 2004 15099417
Morley JE, Kraenzle D: Causes of weight loss in a community nursing
home. J Am Geriatr Soc 42(6):583–585, 1994 8201141
Morse JQ, Lynch TR: A preliminary investigation of self-reported
personality disorders in late life: prevalence, predictors of depressive
severity, and clinical correlates. Aging Ment Health 8(4):307–315,
2004 15370047
Murphy E: Social origins of depression in old age. Br J Psychiatry
141(2):135–142, 1982 7116052
Murphy E: The prognosis of depression in old age. Br J Psychiatry
142(2):111–119, 1983 6839065
Murphy E, Smith R, Lindesay J, et al: Increased mortality rates in late-life
depression. Br J Psychiatry 152(3):347–353, 1988 3167369
Musselman DL, Evans DL, Nemeroff CB: The relationship of depression to
cardiovascular disease: epidemiology, biology, and treatment. Arch
Gen Psychiatry 55(7):580–592, 1998 9672048
Nyth AL, Gottfries CG: The clinical efficacy of citalopram in treatment of
emotional disturbances in dementia disorders: a Nordic multicentre
study. Br J Psychiatry 157(6):894–901, 1990 1705151
O’Connor MK, Knapp R, Husain M, et al: The influence of age on the
response of major depression to electroconvulsive therapy: a C.O.R.E.
report. Am J Geriatr Psychiatry 9(4):382–390, 2001 11739064
Olin JT, Schneider LS, Katz IR, et al: Provisional diagnostic criteria for
depression of Alzheimer disease. Am J Geriatr Psychiatry 10(2):125–
128, 2002 11925273
Parker G, Roy K, Hadzi-Pavlovic D, et al: The differential impact of age on
the phenomenology of melancholia. Psychol Med 31(7):1231–1236,
2001 11681549
Parmelee PA, Katz IR, Lawton MP: Depression among institutionalized
aged: assessment and prevalence estimation. J Gerontol 44(1):M22–
M29, 1989 2783434
Penninx BW, Geerlings SW, Deeg DJ, et al: Minor and major depression
and the risk of death in older persons. Arch Gen Psychiatry
56(10):889–895, 1999a 10530630
Penninx BW, Leveille S, Ferrucci L, et al: Exploring the effect of
depression on physical disability: longitudinal evidence from the
established populations for epidemiologic studies of the elderly. Am J
Public Health 89(9):1346–1352, 1999b 10474551
Pollock BG: Geriatric psychiatry: psychopharmacology: general principles,
in Kaplan and Sadock’s Comprehensive Textbook of Psychiatry. Edited
by Sadock BJ, Sadock VA. Baltimore, MD, Williams & Wilkins, 2000,
pp 3086–3090
Post F: The Significance of Affective Symptoms at Old Age. London,
Oxford University Press, 1962
Post F: The management and nature of depressive illnesses in late life: a
follow-through study. Br J Psychiatry 121(563):393–404, 1972
4342641
Post F: The functional psychoses, in Geriatric Psychiatry. Edited by Isaacs
A, Post F. New York, Wiley, 1978, pp 77–98
Pressman P, Lyons JS, Larson DB, et al: Religious belief, depression, and
ambulation status in elderly women with broken hips. Am J Psychiatry
147(6):758–760, 1990 2343920
Price TR, McAllister TW: Safety and efficacy of ECT in depressed patients
with dementia: a review of clinical experience. Convuls Ther 5(1):61–
74, 1989 11940996
Prigerson HG, Reynolds CF 3rd, Frank E, et al: Stressful life events, social
rhythms, and depressive symptoms among the elderly: an examination
of hypothesized causal linkages. Psychiatry Res 51(1):33–49, 1994
8197270
Prince MJ, Harwood RH, Thomas A, et al: A prospective population-based
cohort study of the effects of disablement and social milieu on the
onset and maintenance of late-life depression. The Gospel Oak Project
VII. Psychol Med 28(2):337–350, 1998 9572091
Rabkin JG, Charles E, Kass F: Hypertension and DSM-III depression in
psychiatric outpatients. Am J Psychiatry 140(8):1072–1074, 1983
6869595
Radloff LS: The CES-D Scale: a self-report depression scale for research in
the general population. Appl Psychol Meas 1(3):385–401, 1977
Rapoport MJ, Mamdani M, Herrmann N: Electroconvulsive therapy in
older adults: 13-year trends. Can J Psychiatry 51(9):616–619, 2006
17007229
Raskin J, Xu JY, Kajdasz DK: Time to response for duloxetine 60 mg once
daily versus placebo in elderly patients with major depressive disorder.
Int Psychogeriatr 20(2):309–327, 2008 17588276
Reinliebb ME, Persaud A, Singh D, et al: Vascular depression:
overrepresented among African Americans? Int J Geriatr Psychiatry
29(5):470–477, 2014 24123266
Reynolds CF3rd, Frank E, Perel JM, et al: Combined pharmacotherapy and
psychotherapy in the acute and continuation treatment of elderly
patients with recurrent major depression: a preliminary report. Am J
Psychiatry 149(12):1687–1692, 1992 1443245
Reynolds CF3rd, Frank E, Perel JM, et al: Nortriptyline and interpersonal
psychotherapy as maintenance therapies for recurrent major
depression: a randomized controlled trial in patients older than 59
years. JAMA 281(1):39–45, 1999 9892449
Reynolds CF3rd, Dew MA, Pollock BG, et al: Maintenance treatment of
major depression in old age. N Engl J Med 354(11): 1130–1138, 2006
16540613
Ribeiz SR, Duran F, Oliveira MC, et al: Structural brain changes as
biomarkers and outcome predictors in patients with late-life depression:
a cross-sectional and prospective study. PLoS One8(11):e80049, 2013
24244606
Roberts RE, Kaplan GA, Shema SJ, et al: Does growing old increase the
risk for depression? Am J Psychiatry 154(10):1384–1390, 1997
9326820
Robinson RG, Price TR: Post-stroke depressive disorders: a follow-up
study of 103 patients. Stroke 13(5):635–641, 1982 7123596
Rosenbach ML, Hermann RC, Dorwart RA: Use of electroconvulsive
therapy in the Medicare population between 1987 and 1992. Psychiatr
Serv 48(12):1537–1542, 1997 9406260
Rovner BW, German PS, Brant LJ, et al: Depression and mortality. JAMA
265(8):993–996, 1991 1992213 [Erratum in: JAMA 265(20)]
Sapolsky RM: Why stress is bad for your brain. Science 273(5276):749–
750, 1996 8701325
Sapolsky RM: Depression, antidepressants, and the shrinking hippocampus.
Proc Natl Acad Sci USA 98(22):12320–12322, 2001 11675480
Schleifer SJ, Macari-Hinson MM, Coyle DA, et al: The nature and course
of depression following myocardial infarction. Arch Intern Med
149(8):1785–1789, 1989 2788396
Schulz R, Drayer RA, Rollman BL: Depression as a risk factor for non-
suicide mortality in the elderly. Biol Psychiatry 52(3):205–225, 2002
12182927
Schwarzbach M, Luppa M, Forstmeier S, et al: Social relations and
depression in late life: a systematic review. Int J Geriatr Psychiatry
29(1):1–21, 2014 23720299
Scott J, Teasdale JD, Paykel ES, et al: Effects of cognitive therapy on
psychological symptoms and social functioning in residual depression.
Br J Psychiatry 177(5):440–446, 2000 11059998
Seidman SN, Araujo AB, Roose SP, et al: Low testosterone levels in elderly
men with dysthymic disorder. Am J Psychiatry 159(3):456–459, 2002
11870011
Sheline YI: Neuroimaging studies of mood disorder effects on the brain.
Biol Psychiatry 54(3):338–352, 2003 12893109
Sheline YI, Wang PW, Gado MH, et al: Hippocampal atrophy in recurrent
major depression. Proc Natl Acad Sci USA 93(9):3908–3913, 1996
8632988
Sheline YI, Mintun MA, Moerlein SM, Snyder AZ: Greater loss of 5-
HT(2A) receptors in midlife than in late life. Am J Psychiatry
159(3):430–435, 2002 11870007
Sherwin BB, Gelfand MM: Sex steroids and affect in the surgical
menopause: a double-blind, cross-over study.
Psychoneuroendocrinology 10(3):325–335, 1985 3903822
Shulman KI: Mania in old age, in Affective Disorders in the Elderly. Edited
by Murphy E. Edinburgh, Scotland, Churchill Livingstone, 1986, pp
203–216
Shulman KI: The influence of age and aging on manic disorder. Int J
Geriatr Psychiatry 4(2):63–65, 1989
Shulman K, Post F: Bipolar affective disorder in old age. Br J Psychiatry
136(1):26–32, 1980 7357218
Smith GS, Gunning-Dixon FM, Lotrich FE, et al: Translational research in
late-life mood disorders: implications for future intervention and
prevention research. Neuropsychopharmacology 32(9):1857–1875,
2007 17327888
Spar JE, Ford CV, Liston EH: Bipolar affective disorder in aged patients. J
Clin Psychiatry 40(12):504–507, 1979 500573
Spiegel D: Cancer and depression. Br J Psychiatry Suppl 169(30
suppl):109–116, 1996 8864156
Staab JP, Evans DL: Efficacy of venlafaxine in geriatric depression.
Depress Anxiety 12(S1 suppl 1):63–68, 2000 11098416
Steffens DC, Payne ME, Greenberg DL, et al: Hippocampal volume and
incident dementia in geriatric depression. Am J Geriatr Psychiatry
10(1):62–71, 2002 11790636
Steffens DC, McQuoid DR, Smoski MJ, et al: Clinical outcomes of older
depressed patients with and without comorbid neuroticism. Int
Psychogeriatr 25(12):1985–1990, 2013 23941723
Steuer JL, Mintz J, Hammen CL, et al: Cognitive-behavioral and
psychodynamic group psychotherapy in treatment of geriatric
depression. J Consult Clin Psychol 52(2):180–189, 1984 6715645
Stoudemire A, Atkinson P: Use of cyclic antidepressants in patients with
cardiac conduction disturbances. Gen Hosp Psychiatry 10(6):389–397,
1988 3060396
Stoudemire A, Hill CD, Morris R, et al: Long-term outcome of treatment-
resistant depression in older adults. Am J Psychiatry 150(10):1539–
1540, 1993 8379561
Stoudemire A, Hill CD, Morris R, et al: Improvement in depression-related
cognitive dysfunction following ECT. J Neuropsychiatry Clin Neurosci
7(1):31–34, 1995 7711488
Stoudemire A, Hill CD, Marquardt M, et al: Recovery and relapse in
geriatric depression after treatment with antidepressants and ECT in a
medical-psychiatric population. Gen Hosp Psychiatry 20(3):170–174,
1998 9650035
Sullivan MD, LaCroix AZ, Baum C, et al: Functional status in coronary
artery disease: a one-year prospective study of the role of anxiety and
depression. Am J Med 103(5):348–356, 1997 9375701
Takeshita J, Masaki K, Ahmed I, et al: Are depressive symptoms a risk
factor for mortality in elderly Japanese American men?: the Honolulu-
Asia Aging Study. Am J Psychiatry 159(7):1127–1132, 2002 12091190
Taylor WD, Aizenstein HJ, Alexopoulos GS: The vascular depression
hypothesis: mechanisms linking vascular disease with depression. Mol
Psychiatry 18(9): 963–974, 2013a 23439482
Taylor WD, Zhao Z, Ashley-Koch A, et al: Fiber tract-specific white matter
lesion severity: findings in late-life depression and by AGTR1 A1166C
genotype. Hum Brain Mapp 34(2):295–303, 2013b 22021115
Tew JDJr, Mulsant BH, Haskett RF, et al: Acute efficacy of ECT in the
treatment of major depression in the old-old. Am J Psychiatry
156(12):1865–1870, 1999 10588398
Thakur M, Hays J, Krishnan KR: Clinical, demographic and social
characteristics of psychotic depression. Psychiatry Res 86(2):99–106,
1999 10397412
Thomas C, Kelman HR, Kennedy GJ, et al: Depressive symptoms and
mortality in elderly persons. J Gerontol 47(2):S80–S87, 1992 1538079
Thompson LW, Gallagher D, Breckenridge JS: Comparative effectiveness
of psychotherapies for depressed elders. J Consult Clin Psychol
55(3):385–390, 1987 3597953
Turner R, Turner J: Social integration and support, in Handbook of
Sociology of Mental Health. Edited by Aneshensel C, Phelan J. New
York, Kluwer Academic, 1999, pp 301–319
Waite A, Bebbington P, Skelton-Robinson M, et al: Social factors and
depression in carers of people with dementia. Int J Geriatr Psychiatry
19(6):582–587, 2004 15211540
Weihs KL, Settle EC Jr, Batey SR, et al: Bupropion sustained release versus
paroxetine for the treatment of depression in the elderly. J Clin
Psychiatry 61(3):196–202, 2000 10817105
Weissman MM, Prusoff BA, Klerman GL: Personality and the prediction of
long-term outcome of depression. Am J Psychiatry 135(7):797–800,
1978 665790
Weissman MM, Bruce ML, Leaf PJ, et al: Affective disorders, in
Psychiatric Disorders in America: The Epidemiologic Catchment Area
Study. Edited by Robins LN, Regier DA. New York, Free Press, 1991,
pp 53–80
Wesner RB, Winokur G: The influence of age on the natural history of
unipolar depression when treated with electroconvulsive therapy. Eur
Arch Psychiatry Neurol Sci 238(3):149–154, 1989 2721532
Whooley MA, Browner WS; Study of Osteoporotic Fractures Research
Group: Association between depressive symptoms and mortality in
older women. Arch Intern Med 158(19):2129–2135, 1998 9801180
Whyte EM, Pollock BG, Wagner WR, et al: Influence of serotonin-
transporter-linked promoter region polymorphism on platelet activation
in geriatric depression. Am J Psychiatry 158(12):2074–2076, 2001
11729031
Williams SA, Kasl SV, Heiat A, et al: Depression and risk of heart failure
among the elderly: a prospective community-based study. Psychosom
Med 64(1):6–12, 2002 11818580
Winokur G: The Iowa 500: heterogeneity and course in manic-depressive
illness (bipolar). Compr Psychiatry 16(2):125–131, 1975 1120412
Wylie ME, Mulsant BH, Pollock BG, et al: Age at onset in geriatric bipolar
disorder: effects on clinical presentation and treatment outcomes in an
inpatient sample. Am J Geriatr Psychiatry 7(1):77–83, 1999 9919324
Yassa R, Nair V, Nastase C, et al: Prevalence of bipolar disorder in a
psychogeriatric population. J Affect Disord 14(3):197–201, 1988
2968383
Yen YC, Rebok GW, Gallo JJ, et al: ApoE4 allele is associated with late-life
depression: a population-based study. Am J Geriatr Psychiatry
15(10):858–868, 2007 17911363
Yesavage JA, Brink TL, Rose TL, et al: Development and validation of a
geriatric depression screening scale: a preliminary report. J Psychiatr
Res 17(1):37–49, 1982–1983 7183759
Young RC, Falk JR: Age, manic psychopathology, and treatment response.
Int J Geriatr Psychiatry 4(2):73–78, 1989
Young RC, Klerman GL: Mania in late life: focus on age at onset. Am J
Psychiatry 149(7):867–876, 1992 1609864
Zesiewicz TA, Gold M, Chari G, et al: Current issues in depression in
Parkinson’s disease. Am J Geriatr Psychiatry 7(2):110–118, 1999
10322237
Suggested Readings
Alexopoulos G, Meyers B, Young RC, et al: The course of geriatric
depression with “reversible dementia”: a controlled study. Am J
Psychiatry 150:1693–1699, 1993
Aziz R, Steffens DC: What are the causes of late-life depression? Psychiatr
Clin North Am 36:497–516, 2013
Blazer DG: Psychiatry and the oldest old. Am J Psychiatry 157:1915–1924,
2000
Blazer DG: Depression in Late Life, 3rd Edition. New York, Springer, 2002
Blazer DG: Depression in late life: review and commentary. J Gerontol A
Biol Sci Med Sci 58:249–265, 2003
Blazer DG 2nd, Hybels CF: Origins of depression in later life. Psychol Med
35:1241–1252, 2005
Koenig HG, Meador KG, Cohen HJ, et al: Depression in elderly
hospitalized patients with medical illness. Arch Intern Med 148:1929–
1936, 1988
Krishnan KR, Goli V, Ellinwood EH, et al: Leukoencephalopathy in
patients diagnosed as major depressive. Biol Psychiatry 23:519–522,
1988 [Erratum in: Biol Psychiatry 25(6):822, 1989]
Reynolds CF 3rd, Dew MA, Pollock BG, et al: Maintenance treatment of
major depression in old age. N Engl J Med 354:1130–1138, 2006
Steffens DC, Payne ME, Greenberg DL, et al: Hippocampal volume and
incident dementia in geriatric depression. Am J Geriatr Psychiatry
10:62–71, 2002
Taylor WS, Steffens DC, MacFall JR, et al: White matter hyperintensity
progression and late-life depression outcomes. Arch Gen Psychiatry
60:1090–1096, 2003
1The discussion on etiology is abstracted, with permission, from Blazer D,
Hybels C: “Origins of Depression in Later Life.” Psychological Medicine
35:1241–1252, 2005.
CHAPTER 10
Diagnosis
Bipolar disorder is a cycling illness that affects an individual’s ability to
regulate moods, which may be manic, hypomanic, or depressed. Under the
DSM-5 rubric of bipolar and related disorders are five main entities: bipolar
I disorder, bipolar II disorder, cyclothymic disorder, substance/medication-
induced bipolar and related disorder, bipolar and related disorder due to
another medical condition, and other specified or unspecified bipolar and
related disorder (American Psychiatric Association 2013).
For a diagnosis of bipolar I disorder, the patient must have experienced
at least one manic episode—that is, an alteration in mood that is euphoric,
expansive, or irritable and is associated with increased energy. These
changes must last for at least 1 week and be accompanied by three of the
seven associated symptoms listed in DSM-5 (e.g., decreased need for sleep,
racing thoughts, pressured speech, increased behaviors that may have high
likelihood for bad outcome). It is not necessary for a patient to have
experienced a depressive episode to be diagnosed with a bipolar disorder,
although the vast majority of patients with bipolar disorders have
experienced depression and many report it to be the most commonly
experienced mood problem.
For a diagnosis of bipolar II disorder, the patient must have experienced
one or more major depressive episodes and at least one hypomanic episode.
A hypomanic episode is defined as at least 4 days of altered mood
(expansive, euphoric, irritable) occurring with at least three of the seven
associated symptoms listed in DSM-5.
Bipolar I and bipolar II are the most recognized and most common
bipolar conditions. They are defined by symptom presentation and intensity.
The following two diagnoses are defined by the causes of the altered mood:
1) substance/medication-induced bipolar and related disorder refers to an
episode of altered mood that meets criteria for a manic episode but occurs
during or soon after substance intoxication or withdrawal, or after exposure
to a medication, and 2) bipolar and related disorder due to another medical
condition refers to an episode of altered mood that meets criteria for a
manic episode but is caused by a known medical condition.
Cyclothymic disorder is characterized by the presence, over a 2-year
period, of numerous periods of hypomanic symptoms (that do not meet
criteria for a hypomanic episode) and depressive symptoms (that do not
meet criteria for a major depressive episode). During this period, the person
has not been without these hypomanic or depressive symptoms for more
than 2 months at a time. It is unclear whether cyclothymia is truly a disorder
disease process, a temperament variation, or a premorbid state for bipolar II
disorder (Baldessarini et al. 2011), given that there is a 15%–50% risk that a
person with cyclothymic disorder will later meet criteria for bipolar I or
bipolar II disorder (American Psychiatric Association 2013).
Finally, the last two categories of bipolar disorders refer to episodes of
altered mood that interfere with functioning but do not completely meet
criteria for any disorder in the bipolar and related disorders diagnostic class.
Other specified bipolar and related disorders may include hypomanic
episodes that last only 2–3 days, or hypomanic episodes without a history of
major depressive episodes, or cyclothymia that has lasted less than 24
months. The diagnosis of unspecified bipolar and related disorder applies to
presentations in which symptoms characteristic of a bipolar disorder cause
clinical distress or impairment but do not meet the full criteria for any of the
specified disorders. For the most part, little is known about bipolar II and
related disorders in late life; therefore, all information presented in this
chapter focuses on bipolar I disorder unless otherwise noted.
Prevalence
The exact prevalence of bipolar disorder in late life is uncertain. Based on
five large-scale studies that used very different sampling methods, the
prevalence of bipolar disorder in the community has been generally
reported to range from 0.08% to 0.5%. The Epidemiologic Catchment Area
(ECA) study conducted in the early 1990s sampled 18,263 community-
dwelling Americans at five sites to determine the prevalence of mental
illnesses for those ages 15 years and older (Weissman et al. 1988). The
ECA study found that bipolar disorder for adults ages 65 years and older
had a 1-year prevalence range of 0.0%–0.5%, with a cross-site mean of
0.1%. This was markedly lower than the prevalence among young (ages
18–44 years; 1.4%) and middle-age (ages 45–64 years; 0.4%) adults.
Similarly, a large health maintenance organization administrative database
review containing almost 300,000 unique individuals found a prevalence of
0.25% for bipolar disorder in persons ages 65 years and older and a
prevalence rate of 0.46% in adults ages 40–64 years (Unützer et al. 1998).
Hirschfeld et al. (2003) sent the Mood Disorder Questionnaire (a validated
screening instrument for bipolar I and II disorders) to 127,000 people. For
the 85,258 responders, the overall screen rate for bipolar disorder was 3.4%,
but for adults ages 65 years and older, the screen rate was 0.5%. Klap et al.
(2003) conducted a telephone survey of 9,585 households and found a
prevalence rate of 0.08% for adults ages 65 years and older compared with
1.17% for adults ages 30–64 years. Finally, the National Comorbidity
Survey Replication studied the incidence of psychiatric disorders in
community-dwelling adults (Chou et al. 2011). Of the 2,575 participants
ages 55 years and older, the 3-month incidence rate was 0.54% for bipolar I
disorder and 0.34% for bipolar II disorder. Interestingly, each of these
surveys suggested that the prevalence of bipolar disorder declines with age
or in aging cohorts.
Winokur (1975) was the first to propose that manic patients may “burn
out” after a finite number of episodes. In a retrospective study, Angst et al.
(1973) described a finite number of episodes in patients with mania,
suggesting that the illness may be self-limiting. However, in a later
prospective study, Angst and Preisig (1995) followed 209 patients with
bipolar disorder over a period of 40 years until a median age of 68. They
found that manic episodes did not decrease with age and that many patients
continued to have episodes into their 60s. It should be noted that the decline
in prevalence rates of bipolar disorder with age is consistent with findings
for other mental illnesses, such as depression and schizophrenia.
Because most of the large-scale prevalence surveys have excluded
patients who were institutionalized in hospitals, nursing homes, or other
residential treatment centers, critics have suggested that the prevalence data
may underrepresent the true prevalence because older patients with mental
illness are more likely to require institutionalized care. Two surveys of
mental illness in nursing home patients found a prevalence of bipolar
disorder of 3%–10% (Koenig and Blazer 1992; Tariot et al. 1993). Speer
(1992) reported that bipolar disorder was present in 17.4% of residential
psychiatric programs for older adults.
Chart reviews of inpatient admissions revealed prevalence rates of
bipolar disorder among older adults in psychiatric inpatient treatment
settings to be 4.7%–18.5% (for a review of published surveys, see Depp and
Jeste 2004 and Dols et al. 2014a). The mean prevalence was 6.0%–8.7%,
although Depp et al. (2004) argued that this was likely to be an
underestimation, because many of the inpatient surveys did not include
bipolar depressed subjects or reported prevalence only for those whose
bipolar disorder began after age 60 years. In summary, the prevalence of
bipolar disorder may depend in part on the diagnostic criteria or
measurement used and on the population sampled. In general, community
surveys show that bipolar disorder appears to decrease with age, a change
consistent with the declines seen in other mental disorders. Nevertheless,
the proportion of older patients utilizing services (either inpatient or
outpatient) appears to be the same as in younger populations. Furthermore,
there may be increasing utilization of institutional care with age.
Gender
Epidemiological studies in the United States have indicated that bipolar
disorder is approximately equally common in men and women (American
Psychiatric Association 2000). Depp and Jeste (2004) pooled 17 studies
reporting various samples of late-life bipolar disorder and found that the
weighted mean of elderly women with bipolar disorder was 69% (range
45%–89%). However, they noted that this percentage was similar to the
gender ratio among older adults in the general population.
Comorbidity
Psychiatric Comorbidity
Psychiatric comorbidity is frequently seen in patients with bipolar disorder
and has been a major point of discussion in the literature on bipolar
disorder, yet there is very little information about its presence in late-life
bipolar disorder. Only a scattering of studies have looked at substance abuse
comorbidity, whereas individual reports (mostly regional or individual
hospital–based data) are available for personality disorders, posttraumatic
stress disorder (PTSD), and anxiety disorders. There are no published
reports on comorbid eating disorders or attention disorders, which have
been found to be significant in surveys of mixed-age subjects with bipolar
disorder.
Sajatovic et al. (2006) conducted a review of the national Veterans
Health Administration database to examine the prevalence of dementia,
PTSD, and anxiety disorders in older patients with bipolar disorder. They
identified 4,668 subjects with bipolar disorder (mean age 70 years); of
these, 4.5% had comorbid dementia, 5.4% had PTSD, and 9.4% had an
anxiety disorder. In the Netherlands, Dols et al. (2014b) looked at a cohort
of elderly patients with bipolar disorder and found that comorbid anxiety
disorders were relatively rare (1-month panic disorder, 2%; 1-month social
anxiety, 3%; 6-month general anxiety disorder, 5%). Lifetime alcohol
dependence was 24.8% and abuse 13.9% among those with bipolar
disorder.
The best evidence for the prevalence of comorbid substance abuse and
bipolar disorders comes from the National Comorbidity Study, in which
61% of individuals with bipolar disorder also had a substance use disorder
(Kessler et al. 1997). Unfortunately, this survey excluded adults older than
age 55 years from the data set. Cassidy et al. (2001) reviewed rates of
substance abuse in 392 patients who were hospitalized at a state psychiatric
facility for bipolar disorder. Nearly 60% had some history of lifetime
substance abuse (consistent with the finding of the National Comorbidity
Study), but in the 51 patients older than age 60, only 29% had a history of
lifetime substance abuse. Supporting this finding of lower-than-expected
substance abuse disorders in older patients with bipolar disorder are two
small inpatient retrospective studies (Ponce et al. 1999; Sajatovic et al.
1996), a review of elderly bipolar utilizers of mental health system
outpatient services (Depp et al. 2005), and a large review of the Veterans
Health Administration database (Sajatovic et al. 2006). The reason for this
unexpected finding is unclear.
Only one study has reviewed the presence of personality disorders
together with co-occurring late-life mental disorders. Molinari and
Marmion (1995) reviewed 76 geriatric outpatients and inpatients and found
that 63% of the patients had one of the personality disorders. Of the 27
subjects with bipolar disorder, 70% were found to have a personality
disorder. The authors suggest that the unusually high rate of personality
disorders may be due in part to the difficult and chronic patterns of affective
disorders.
Medical Comorbidity
Because of the high association that secondary mania has with late-life
bipolar disorder, there have been more studies assessing the presence of
comorbid medical problems—the most common being neurological
illnesses—than studies assessing psychiatric comorbidities. Depp and Jeste
(2004) reviewed eight studies that reported the presence of illness and noted
that despite a wide variety in reporting strategies, the sample-weighted
prevalence for neurological illnesses was 23.1%. Shulman et al. (1992)
compared 50 geriatric patients hospitalized for mania with 50 age-matched
patients hospitalized for unipolar depression. They found that the rates of
neurological illness in manic patients were significantly higher (36% vs.
8%), suggesting that neurological disease is a risk factor for the
development of mania in late life.
Other comorbid medical disorders are also common in bipolar disorder
and especially in late-life bipolar disorder. Lala and Sajatovic (2012)
reported that a mean of three to four medical conditions appeared to be the
norm in elderly patients with bipolar disorder. Regenold et al. (2002)
reviewed the inpatient charts of 243 older (ages 50–74 years) psychiatric
inpatients. They found that type II diabetes was present in 26% of those
with bipolar disorder, which is a much higher rate than in patients with
unipolar depression, patients with schizophrenia, and inpatients with bipolar
disorder in other studies (9.9% in mixed-age inpatient sample; Cassidy et al.
1999). Dols et al. (2014b) interviewed 101 elderly patients with bipolar
disorder (mean age 68.9±7.8 years) and found an average of 1.7 medical
comorbid conditions, predominately hypertension (27.8%), arthrosis
(29.1%), allergies (25.6%), and peripheral atherosclerotic disease (18.8%).
Only 21.8% had no somatic illnesses. Metabolic syndrome was found in
28.7% of patients.
Dementia
Dementia has become an increasing concern for older adults in general and
possibly a special concern for older adults with bipolar disorder. Studies of
four inpatient samples found that the rate of comorbid dementia was highly
variable, ranging from 3% to 25% (Broadhead and Jacoby 1990;
Himmelhoch et al. 1980; Ponce et al. 1999; Stone 1989). Sajatovic et al.
(2006) reported dementia in 4.5% of veterans treated for bipolar disorder
through the Veterans Health Administration system. Comorbidity does not
necessarily imply an association, which has been a particular concern for
late-life bipolar disorder. Tsai et al. (2003) reported that 30.7% of the early-
onset patients in their inpatient sample had Mini-Mental State Examination
(MMSE) scores below 24. Furthermore, Dhingra and Rabins (1991)
reviewed 25 elderly patients who had been hospitalized 5–7 years
previously for manic episodes and found that 32% experienced a significant
decline in their MMSE scores. Kocsis et al. (1993) followed 38 elderly
patients with bipolar disorder treated with lithium and found that rates of
cognitive and functional impairment were much higher than in the general
population.
Mortality
Individuals with mental illness at all ages have higher mortality rates, from
both natural and unnatural causes, than the general population (Laursen et
al. 2007). This is especially true for patients with bipolar disorder. Ramsey
et al. (2013) reported on the 26-year follow-up data of participants from the
Epidemiologic Catchment Area Study. They found that odds of mortality
for patients with lifetime manic spectrum episodes (either full manic,
hypomanic, or subthreshold manic symptoms) was much higher (odds ratio
1.4) than for those with no lifetime manic spectrum episodes. Dhingra and
Rabins (1991) found that mortality rates among elderly patients with
bipolar disorder who had been hospitalized 5–7 years previously were
higher than expected compared with population norms. Shulman et al.
(1992) found that the mortality rate over a 10- to 15-year follow-up for
elderly hospitalized patients with bipolar disorder was significantly higher
than that of elderly hospitalized unipolar depressed patients (50% vs. 20%),
suggesting that mania appears to have a poorer prognosis and to be a more
severe form of affective illness than unipolar depression.
In summary, psychiatric comorbidity is frequent in patients with bipolar
disorder, but the prevalence of the comorbidities in older patients with
bipolar disorder is relatively unknown. Substance abuse, the most common
and best-reported comorbid condition in the literature, appears less
commonly in older than younger patients with bipolar disorder. Lifetime
history also appears less commonly. Dementia and poorer cognitive
performance on neurological testing (see “Neurocognitive Testing”
subsection later in this chapter) are possibly increased, or apparent earlier,
in older adults with bipolar disorder. Medical comorbidity is also higher in
older adults with bipolar disorder (Beyer et al. 2005), with special concern
about neurological illness and diabetes. All of these problems may
contribute to higher mortality rates for patients with bipolar disorder than
for individuals without psychiatric illness and for unipolar depressed
patients.
Course
After reviewing studies that retrospectively looked at course of bipolar
disorder prior to hospitalization, Goodwin and Jamison (1990) reported that
depression was the initial episode more often in older adults than in
younger patients. Various investigators have described a latency period of
10–20 years between first depressed episode and onset of mania
(Broadhead and Jacoby 1990; Shulman and Post 1980; Shulman et al. 1992;
Snowdon 1991; Stone 1989). Kessing (2006), in a study of Denmark’s
health care utilization, found that first psychiatric hospitalizations for older
adults with bipolar disorder were much more likely to be for depressive
episodes than for manic episodes. Furthermore, older adults with bipolar
disorder appear to experience more mixed symptoms than the classic manic
presentation (Post 1968; Spar et al. 1979).
Previously, it was believed that elderly patients with bipolar disorder
may differ in symptom presentation from younger patients. However, a
review from the Systematic Treatment Enhancement Program for Bipolar
Disorder (STEP-BD) found no statistically significant association between
age and symptom presentation of acute depression or mood elevation or
psychosis (Al Jurdi et al. 2012).
As noted previously, Angst and Preisig (1995) followed 209
psychiatrically hospitalized patients with bipolar disorder over a 40-year
period until a median age of 68 years. Of these patients, only 16% had fully
recovered (defined as no episodes in the previous 5 years and a Global
Assessment of Functioning [GAF] Scale score above 60), whereas 26% had
no episodes but had GAF scores below 60. The largest group (36%) had
experienced episodes within the previous 5 years, and another 16%
exhibited a chronic course. Seven percent had committed suicide.
Interestingly, despite the high risk of suicide reported with aging and
among individuals with bipolar disorder, the risk of suicide in late-life
bipolar disorder appears lower than expected. Tsai et al. (2002) studied
suicide rates in Taiwan among patients with bipolar disorder and found that
the highest risk was during the first 7–12 years after the onset of the illness
and for individuals under age 35 years. Depp and Jeste (2004) suggested
that older patients with early-onset bipolar disorder may constitute a
“survivor cohort.” Young and Falk (1989) found that older patients with
mania who need hospitalization tend to have a slower resolution of
symptoms and a longer duration of hospitalization than do younger adult
patients.
A study comparing health-related quality of life and functioning in
elderly adults with bipolar disorder (n=54) and a group without psychiatric
illness (n=38) found that even patients in remission from bipolar disorder
had quality-of-life scores lower than those of the normal comparison group.
Overall, bipolar disorder was associated with substantial disability,
comparable to schizophrenia, and incomplete improvement in functioning
even among those classified as remitters (Depp et al. 2006).
Similar to younger patients with bipolar disorder, older adults with the
disorder have a high use rate of mental health services. Sajatovic et al.
(1996) found that older patients with bipolar disorder were hospitalized at
the same rate as older patients with schizophrenia. Bartels et al. (2000)
found that older individuals with bipolar disorder used outpatient services
four times as often as a similarly aged group of individuals with unipolar
depression. Brennan et al. (2002) found that older veterans with comorbid
bipolar disorder and substance use disorders had an increased probability of
mental health care service use and readmission. However, Depp et al.
(2005) compared 2,903 elderly individuals with bipolar disorder who
received outpatient services with younger bipolar groups and found that the
elderly individuals were less likely to use inpatient, outpatient, and
emergency room psychiatric care but were more likely to use case
management and conservator services. Overall, the elderly bipolar group
had less substance use but more cognitive disorders and a lower global
functioning than the younger bipolar groups.
Age at Onset
The mean age at onset for bipolar disorder is in the late teens to early 20s
(Weissman et al. 1996). Although most studies have found bipolar disorder
to be unimodal in distribution, a few studies have noted two peaks for the
onset of mania, with the first occurring in the mid-20s and a second, smaller
peak occurring closer to middle age, in the late 40s (Angst 1978; Goodwin
and Jamison 1984; Petterson 1977). Others have suggested a trimodal
distribution pattern, with distinct groups represented by onset in late teens,
mid-20s, and early 40s (Bellivier et al. 2003).
Despite being phenotypically similar to the other age-at-onset subtypes,
the late-onset subtype has historically been viewed as an “organic” variant
of bipolar disorder. This may be due in part to the relative small number of
new-onset bipolar disorders in older adults (especially in late life) compared
with younger adults, as well as the known physical changes associated with
age, such as the suggestions that women may be at risk for first episodes
around the time of menopause (Angst 1978; Petterson 1977; Sibisi 1990;
Zis et al. 1979) and that men may have increased risk in their 70s (Spicer et
al. 1973) or 80s (Sibisi 1990), possibly due to neurological causes. Shulman
and Post (1980) reviewed the course of 67 elderly patients with bipolar
disorder whose first manic episode occurred around age 60; the authors
noted a pattern of an initial depressive episode in mid-life, followed by a
long latency period (mean of 15 years) before the onset of mania. They
hypothesized that this group may have underlying cerebral changes due to
aging that convert them to mania.
Many researchers have emphasized the importance of dividing bipolar
disorder into early- and late-onset subtypes, noting heuristic evidence of
differences in familial incidence of mood disorders, neurological disease,
and mortality. Patients with early-onset illness may have an increased
prevalence of close family members with affective disorders compared with
patients who had later-onset illness (Baron et al. 1981; Hopkinson 1964;
James 1977; Mendlewicz et al. 1972; Post 1968; Snowdon 1991; Stenstedt
1952; Taylor and Abrams 1973). Despite arguments that this would suggest
a higher genetic loading for early-onset patients and/or a higher incidence
of secondary bipolar disorder for late-onset patients, this issue is not clear
because other studies have not substantiated this finding (Broadhead and
Jacoby 1990; Carlson et al. 1977; Depp et al. 2004; Glasser and Rabins
1984; Hays et al. 1998; Tohen et al. 1994). Notably, even if the number of
affectively ill relatives is higher in the early-onset group, the percentage of
affectively ill relatives in the late-onset group is still considerable and
corresponds to the 4%–22% generally cited for the relatives of manic
patients (James 1977; Mendlewicz et al. 1972; Post 1968; Stenstedt 1952;
Taylor and Abrams 1973).
The existence of a relationship between late-onset illness and
neurological abnormalities is a much more consistent finding. Although the
definition of neurological illness varied in eight studies that examined this
issue, six of the studies showed significantly higher rates in the late-onset
patients (Almeida and Fenner 2002; Huang et al. 2012; Snowdon 1991;
Subramaniam et al. 2007; Tohen et al. 1994; Wylie et al. 1999), whereas the
other two studies showed trends toward increased levels of neurological
illness in late-onset patients (Broadhead and Jacoby 1990; Hays et al.
1998). Late-onset subjects were also more likely to have a higher risk for
mortality.
The importance of identifying a late-onset bipolar disorder as a distinct
syndrome has come under question. Besides the information listed above,
few meaningful differences have been found between early-onset and late-
onset groups, except for possibly less overall psychopathology and
improved treatment response (Carlson et al. 1977, 2000; Chu et al. 2010;
Depp et al. 2004; Leboyer et al. 2005). Furthermore, DSM-5 (American
Psychiatric Association 2013) does not make such a distinction.
Interestingly, some reports have suggested that age at onset may be a more
important differential for individuals with an early onset (before age 13;
Perlis et al. 2009) than for those with late onset.
Neurocognitive Testing
Cognitive dysfunctions are increasingly recognized as a core feature of
bipolar disorder. The affected abilities are most often noted in assessments
of executive function, verbal memory, and processing speed. These
dysfunctions are not solely related to residual mood effects, drug effects, or
other confounding factors but rather appear to be related to illness severity.
They are also apparent in first-degree relatives. These findings have
suggested that bipolar disorder may be associated with various
neurobiological factors that may reflect a neurodegenerative process (e.g.,
glutamatergic excitotoxicity, neuroinflammation, oxidative stress,
mitochondrial dysfunction). Adults with bipolar disorder exhibit a range of
cognitive deficits that persist across illness stages and during symptom
remission, the severity of which may rival those described in schizophrenia
(Balanzá-Martínez et al. 2005). Studies of older adults with bipolar disorder
have found patterns similar to that of younger adults with bipolar disorder:
deficits in executive functioning, working memory, verbal memory,
attention, construction, and processing speed (Aprahamian et al. 2014;
Delaloye et al. 2011; Gildengers et al. 2010; Gunning-Dixon et al. 2008;
Lewandowski et al. 2014; Samamé et al. 2013; Young et al. 2006). Depp et
al. (2007) suggested that the neurocognitive deficits in geriatric patients
with bipolar disorder were related to quality of life rather than to severity or
duration of psychiatric symptoms, suggesting that bipolar disorder often
involves disabling and enduring cognitive impairments. In a study by
Köhler et al. (2013), the presence of psychotic symptoms was associated
with poorer cognitive functioning and more rapid cognitive decline over a
6-year follow-up.
The course of cognitive functioning over the lifespan in adults with
bipolar disorder has been less clear. Some research has suggested that
compared with mentally healthy individuals of similar age and education,
older adults with bipolar disorder not only had worse performance but also
appeared to have a faster decline (Dhingra and Rabins 1991; Gildengers et
al. 2004, 2009). These findings would seem to support the hypothesis that
bipolar disorder may be related to a neurodegenerative (progressive)
process. However, other research has not found accelerated decline in older
adults with bipolar disorder (Delaloye et al. 2011; Depp et al. 2007). In the
largest longitudinal study, Gildengers et al. (2013) found that both older
adults with bipolar disorder and age-matched comparators exhibited a
decline in neuropsychological functioning over a 2-year follow-up period;
however, the bipolar group displayed worse cognitive function in all
domains at baseline and follow-up. The research group did not find an
accelerated decline in the bipolar group over time, suggesting that cognitive
impairment and associated functional disability of older adults with bipolar
disorder appear to be due to long-standing neuroprogressive processes
compounded by normal cognitive aging rather than a neurodegenerative
process.
Neuroimaging
Stroke
Compared with unipolar depression, mania following a stroke is relatively
uncommon. However, a controlled study of patients with secondary mania
showed that when manic symptoms are present, the right hemisphere of the
brain (both cortical and subcortical areas) is more frequently the site of the
lesion (Starkstein et al. 1987, 1991). Further research suggests that
development of mania after right-sided lesions occurs more often when the
basal region of the right temporal lobe is involved (Starkstein et al. 1990).
Huang et al. (2012) found that silent cerebral infarctions were frequently
observed in older adults with bipolar disorder but were often neglected or
unobserved, regardless of the patients’ age at onset.
Volumetric Neuroimaging
Neuroimaging in mixed-age subjects with bipolar disorder is still a rising
field, but as the literature has evolved, an emerging hypothesis for
understanding bipolar disorder has developed, suggesting that affective
instability may be the result of changes or altered processes in certain areas
of the brain (Phillips 2006). There appears to be increased activity among
the subcortical and limbic regions that make an initial assessment of
“emotional stimulus” (amygdala, anterior insula), resulting in increased
activity in regions associated with mood generation and decision processing
of the emotional material (ventromedial and ventrolateral prefrontal
cortices, ventral anterior cingulate gyrus). Finally, there is reduced activity
in regions that regulate these responses and attentional processes
(dorsomedial prefrontal cortices), causing the mood lability. Neuroimaging
results specific to late-life bipolar disorders have been relatively limited, but
the published studies have supported significant volumetric abnormalities in
certain areas that are consistent with the overall theory presented above.
Volumetric studies of total brain volumes for patients with bipolar
disorder have shown only limited changes. Young et al. (1999) compared 30
geriatric manic patients with control subjects but did not find any difference
in the ventricular-brain ratios. They did note that subjects with bipolar
disorder had greater cortical sulcal widening, which correlated with age at
onset and age at first manic episode. Tanaka et al. (1982) suggested that
atrophy may occur at an earlier age in subjects with bipolar disorder, despite
the fact that cortical atrophy in their elderly subjects with bipolar disorder
was similar to that in normal (healthy) control subjects. Beyer et al. (2004a)
also did not find any differences in total brain volume or lateral ventricular
volumes between elderly subjects with bipolar disorder and controls;
however, they did find a volume decrease in those with late-onset (after age
45 years) versus those with early-onset bipolar disease.
Other volumetric findings include smaller caudate (Beyer et al. 2004a)
and larger hippocampal (Beyer et al. 2004b) volumes in patients with late-
life bipolar disorder compared with controls. The latter finding appeared to
be consistent with use of lithium, suggesting a role for neuroprotection.
Magnetic Resonance Imaging Hyperintensities
Possibly related to strokes, “hyperintense” signals viewed on T2-weighted
magnetic resonance imaging (MRI) have been one of the earliest and most
consistent neuroimaging findings in the study of bipolar disorder.
Hyperintensities represent areas of neuronal cell death, although the
mechanism by which this occurs is unclear. Pathological examination has
found that hyperintensities could characterize areas of arteriosclerotic
disease, demyelination, loss of axons, arteriolar hyalinization, rarefaction,
infarctions, and necrosis (Bradley et al. 1984; Braffman et al. 1988;
Chimowitz et al. 1992; Fazekas et al. 1993; Fujikawa et al. 1997; George et
al. 1986). Fujikawa et al. (1997) coined the term silent cerebral ischemia to
refer to these hyperintensities. Electroencephalographic studies in patients
with dementia suggest that white matter hyperintensities may result in a
functional brain disconnection. Therefore, some researchers have suggested
that for patients with mood disorders, hyperintensities may “disconnect”
different pathways in the mood regulation circuits.
Dupont et al. (1987) were the first to report the presence of
hyperintensities in patients with bipolar disorder, and most (Altshuler et al.
1995; Aylward et al. 1994; Botteron et al. 1995; Dupont et al. 1990, 1995;
Figiel et al. 1991; Krabbendam et al. 2000; McDonald et al. 1991, 1999;
Swayze et al. 1990) but not all (Brown et al. 1992; Strakowski et al. 1993)
studies have supported this finding. Because of differences in study groups
and methodology, there is a wide range in frequency of T2 hyperintensities
found in subjects with bipolar disorder (5%–62%) and normal control
subjects (0%–42%). However, in all but one study, the odds ratio was in the
same direction and the overall effect size was highly significant (Bearden et
al. 2001). Two meta-analyses reported similar findings with common odds
ratios of 3.3 (Altshuler et al. 1995) and 3.29 (Videbech 1997), strongly
supporting the relationship between hyperintensities and bipolar disorder.
One group of researchers who did not find a relationship studied patients
with bipolar disorder at the time of their first hospitalization, although they
did find a trend toward the presence of hyperintensities (1.7 times higher
rate) (Strakowski et al. 1993). Interestingly, this finding is not inconsistent
with data from Aylward et al. (1994) and Hickie et al. (1995), who noted
that the presence of hyperintensities was significant only in an older
subgroup of patients with bipolar disorder (older than age 39 years), or
Dupont et al. (1995), who noted a correlation of hyperintensities only in
patients with mania onset after adolescence. Thus, increased age is related
to the presence of hyperintensities, a finding that is not unexpected because
hyperintensities are also seen in normal aging populations as well as in
subjects with unipolar depression.
The presence of hyperintensities not only is more common among
patients with bipolar disorder than among control subjects (Dupont et al.
1995) but remains significantly more common when medical risk factors
(e.g., hypertension, vascular disease) are controlled for (Altshuler et al.
1995; Hickie et al. 1995; McDonald et al. 1999). Specific studies of bipolar
disorder in late life have consistently found a higher presence of
hyperintensities in individuals with bipolar disorder than in control subjects.
In a study of patients older than age 50 years with mania, McDonald et al.
(1991) found no significant difference with respect to the presence of
hyperintensities between the patients with bipolar disorder and the control
sample, yet the mean number of large hyperintensities (>2.5 mm) was
significantly higher in the elderly patients with bipolar disorder. McDonald
et al. (1999), in a mixed bipolar sample that included elderly individuals,
found increased hyperintensities in the subependymal region, subcortical
gray nuclei, and deep white matter, whereas de Asis et al. (2006) found
increased hyperintensities in the frontal deep white matter in a group of
older individuals with bipolar disorder when compared with control
subjects. This increased number was especially prominent on the right side
in the late-onset group.
The presence of hyperintensities may be especially important in late-life
bipolar disorder because of their impact on treatment response and severity
of illness. Data from studies on bipolar disorder are very limited, but MRI
hyperintensities have been found to be associated with longer hospital stays
(Dupont et al. 1990) and more frequent rehospitalizations (McDonald et al.
1999). The role of hyperintensities in contributing to cognitive decline is
not clear. Some researchers (Bearden et al. 2001) have noted that
hyperintensities appear to be associated with cognitive impairment with
increasing age and chronicity of disorder. However, Rej et al. (2014)
dispute this, noting that in their small sample, cognitive dysfunction was not
associated with white matter hyperintensity burden or gray matter volume,
which suggests that other potential neuroprogressive pathways, such as
inflammation, mitochondrial dysfunction, serum anticholinergic burden,
and altered neurogenesis, may play a role.
Differential Diagnosis
There are five potential presentations of patients with late-life bipolar
disorder: 1) those who had early onset of bipolar disease and have now
reached old age; 2) those who previously experienced only episodes of
depression but have now switched to a manic episode; 3) those who have
never had an affective illness but develop mania because of a specific
medical or neurological event (e.g., head trauma, cerebrovascular accident,
hyperthyroidism); 4) those who have never been recognized as having
bipolar symptoms or who have been misdiagnosed with another disorder;
and 5) those who have never had an affective illness but develop mania for
unknown reasons. It is unknown how common each presentation may be,
although in the author’s personal experience, the most frequently
encountered presentation is that of a patient who developed bipolar disorder
earlier in life and is now seeking treatment. However, based on findings by
Hirschfeld et al. (2003), it is not uncommon for the diagnosis of bipolar
illness to have been missed previously.
Because the onset of bipolar disorder in late life is relatively uncommon,
every patient who presents with a new onset of mania should undergo a
good medical evaluation, with special emphasis on the neurological
examination. Because older adults may be receiving a higher number of
medications, these should be reviewed for possible temporal association. A
laboratory workup consisting of a thyroid panel and basic tests should also
be completed. Finally, consideration should be given to neuroimaging,
especially if the presentation is associated with psychosis.
Treatment
Treatment of bipolar disorder is often challenging for many reasons. In
addition to the challenge of medication management in older adults, elderly
patients with bipolar disorder often have incomplete response, recurrent
episodes, potentially severe psychopathology, and higher mortality rates.
Furthermore, the disease itself tends to cause problems with poor insight
and poor compliance with treatment. Finally, limited data are available on
use of medications and psychotherapies for older patients with bipolar
disorder. Many of the treatment practices are based on results from clinical
trials in younger populations that have been adapted for geriatric use.
Lithium
Lithium has traditionally been identified as the gold standard for treatment
of bipolar disorder and has been widely prescribed to older patients with the
disorder (Shulman et al. 2003; Umapathy et al. 2000). Because of the need
to better understand treatment in this vulnerable population, the National
Institute for Mental Health (NIMH) commissioned a study to evaluate the
efficacy and tolerability of lithium and valproate in late-life mania. Young
et al. (2012) conducted a blinded trial of 224 patients with mania over age
60 who were administered either lithium or valproate. They found that both
lithium and valproate were well tolerated and efficacious, but lithium was
associated with greater reduction in mania scores.
The recommended lithium level for acute mania in geriatric patients is
unclear. In the study by Young et al. (2012), the mean dose of lithium was
780±315 mg/day, with a mean serum concentration of 0.76±0.35 mEq/L.
However, some case series have suggested that elderly patients may
respond to lower lithium levels (0.5–0.8 mEq/L) than those recommended
for younger adults (0.6–1.2 mEq/L) (Chen et al. 1999; Prien et al. 1972;
Roose et al. 1979), whereas other studies have not found a difference
(DeBattista and Schatzberg 2006; Young et al. 1992).
Special care must be taken in dosing geriatric patients with lithium. With
aging, the renal clearance of lithium decreases and the elimination half-life
increases (Foster 1992; Shulman et al. 1987; Sproule et al. 2000).
Furthermore, medications commonly prescribed to the elderly, such as
thiazide diuretics, nonsteroidal anti-inflammatory agents, and angiotensin-
converting enzyme inhibitors, can increase lithium concentrations. Other
medications, such as theophylline, can decrease lithium concentrations.
Finally, because lithium use can contribute to hypothyroidism and a decline
in renal clearance, lithium should be used with caution in patients with
kidney problems or thyroid disorders.
Lithium toxicity in elderly individuals is not uncommon (Foster 1992).
Commonly reported adverse effects of lithium in the elderly include
cognitive impairment, ataxia, urinary frequency, weight gain, edema,
tremor, and worsening of psoriasis and arthritis. Because of adverse effects
(including neurotoxicity) that can occur even at therapeutic levels,
appropriate lithium serum levels in the elderly are largely determined by
medical status, frailty, and conservative dosing (Sajatovic et al. 2005b;
Young et al. 2004).
Anticonvulsants
Physicians have increasingly been prescribing anticonvulsants for the
treatment of bipolar disorder. Since 1993, four anticonvulsants have been
approved by the U.S. Food and Drug Administration (FDA) to treat bipolar
disorder: valproate, carbamazepine, oxcarbazepine, and lamotrigine. For
information regarding the anticonvulsants, the literature is limited to case
reports and subanalyses of data from the inclusion of elderly patients in the
larger Phase III trials.
Valproate
Since 2000 there has been a marked increase in the prescription of valproate
for bipolar disorder, especially for elderly patients. In 1999, Oshima and
Higuchi proposed that lithium be the first choice in treatment guidelines for
geriatric bipolar disorder. However, Shulman et al. (2003) noted that
prescriptions of valproate for elderly patients with bipolar disorder
increased while prescriptions of lithium decreased, so that valproate is now
the most prescribed medication treatment for elderly persons with bipolar
disorder.
In the NIMH study on the tolerability and efficacy of lithium and
valproate in late-life mania (Young et al. 2012), valproate performed well
and was tolerated as well as lithium was. The mean daily dose was
1,200±550 mg/day, and the mean serum concentration was 74±21 μg/mL.
In general, the recommended blood level concentration for valproate in
adults is 50–120 μg/mL (Bowden et al. 2002), although Chen et al. (1999)
found that manic elderly patients who had a blood level concentration from
65–90 μg/mL improved more than patients with lower concentrations.
As patients age, the elimination half-life of valproate may be prolonged
and the free fraction of plasma valproate increases. The clinical significance
of this is unknown, although it should be noted that usual laboratory tests
measure the total valproate level. Thus, the reported level may
underrepresent the actual dose available to the brain in geriatric patients
(Sajatovic et al. 2005b; Young et al. 2004). Common medications taken
concurrently may also influence the level of valproate: aspirin can increase
the valproate free fraction, and phenytoin and carbamazepine may decrease
the valproate level. In turn, valproate may affect other medications’ effects.
It can inhibit the metabolism of lamotrigine so that the dose of lamotrigine
may need to be lowered to minimize side effects. Valproate also may
increase the unbound fraction of warfarin; coagulation parameters should
therefore be monitored in patients undergoing anticoagulation therapy
(Panjehshahin et al. 1991).
The most common side effects associated with valproate are nausea,
somnolence, and weight gain. Less common side effects that may be
particularly important to geriatric patients are hair thinning,
thrombocytopenia, hepatotoxicity, and pancreatitis (the latter two are less
likely to occur with age) (Bowden et al. 2002). Notably, valproate is
available in sprinkle and liquid formulations for patients who have
difficulty swallowing. In addition, Regenold and Prasad (2001) have
reported on the intravenous use of valproate in three geriatric patients.
Carbamazepine
Carbamazepine was approved for the treatment of bipolar mania in 1996,
and the extended-release formulation was approved in 2005. Some
researchers have suggested that carbamazepine may be the preferred mood-
stabilizing agent, rather than lithium, for patients with secondary mania
(Evans et al. 1995; Sajatovic 2002); however, very little information is
available on the use of carbamazepine (in either preparation) for elderly
patients with bipolar disorder. Okuma et al. (1990) noted that seven elderly
patients with mania were included in a larger sample of 50 treated with
carbamazepine in a double-blind study that showed good efficacy.
Before initiating carbamazepine, the physician should check liver
enzymes, electrolytes, and complete blood cell count. Because
carbamazepine can also affect the heart’s rhythm, an electrocardiogram
should be considered. In elderly patients, carbamazepine may be started at
100 mg either once or twice daily and gradually increased every 3–5 days to
400–800 mg/day (McDonald 2000). Target serum levels are between 6 and
12 μg/L.
Carbamazepine is metabolized in the liver by cytochrome P450 (CYP)
enzyme 3A4/5. Because carbamazepine can induce its own metabolism,
dose increases may need to be adjusted in the first 1–2 months.
Furthermore, carbamazepine clearance is decreased in an age-dependent
manner, presumably due to a reduction in CYP 3A4/5 metabolism,
suggesting that elderly patients may require lower doses compared with
younger patients to achieve similar blood levels (Battino et al. 2003).
Notably, carbamazepine also may alter the pharmacokinetics of other
medications, including oral hormones, calcium channel blockers,
cimetidine, terfenadine, and erythromycin (Sajatovic 2002).
Possible adverse effects associated with carbamazepine include sedation,
ataxia, nystagmus/blurred vision, leukopenia, hyponatremia (secondary to
the syndrome of inappropriate antidiuretic hormone secretion [SIADH]),
and agranulocytosis. The FDA (U.S. Food and Drug Administration 2007)
has recommended that patients of Asian ancestry have a genetic blood test
to identify an inherited variant of the human leukocyte antigen allele HLA-
B*1502 (found almost exclusively in people of Asian ancestry) before
starting therapy. Patients testing positive should not be treated with
carbamazepine.
Lamotrigine
Lamotrigine was approved by the FDA in 2003 for the maintenance phase
of bipolar disorder. Sajatovic et al. (2005a) conducted a retrospective
analysis of two placebo-controlled, double-blind clinical trials for
maintenance therapy in bipolar disorder, focusing on 98 subjects who were
age 55 years or older. They found that, similar to the parent study,
lamotrigine significantly delayed the time to intervention for any mood
episode, whereas lithium and placebo did not. In a subanalysis of the type
of mood episode that was more likely to recur, the authors found that
lamotrigine was significantly more effective than lithium and placebo at
increasing time to intervention for depressive recurrences, but lithium
performed much better in increasing time to intervention for manic
episodes. Overall, the authors found that lamotrigine was well tolerated
(compared with lithium) by the older patients with bipolar disorder, and no
increased incidence of rash was noted (Sajatovic et al. 2007). In a follow-up
analysis evaluating clinical correlates of treatment response, Gildengers et
al. (2012) noted that lamotrigine worked best in depressed patients with
high cardiometabolic risk factors and a low level of manic symptoms with
their depression. A small case series (Robillard and Conn 2002) of five
female geriatric bipolar patients with depressive episodes suggested good
efficacy when lamotrigine was used as an augmenting agent as well.
Lamotrigine is metabolized in the liver and eliminated through the
hepatic glucuronide conjugation. Some minor decreases in hepatic
glucuronidation occur with aging, although their impact on lamotrigine
dosing in the elderly is not thought to be significant (Hussein and Posner
1997; Posner et al. 1991). Overall, lamotrigine is well tolerated, although
serious skin rashes (Stevens-Johnson syndrome) have been reported. It has
been suggested that lamotrigine may have fewer negative effects on
cognition than other anticonvulsant medications, which may be important
for some geriatric patients (Aldenkamp et al. 2003).
Antidepressants
Antidepressants are frequently prescribed for the treatment of bipolar
depression in elderly patients (Beyer et al. 2008), although the use of
antidepressants in bipolar disorder is a point of continued concern among
psychiatrists (Ghaemi 2012). Three issues highlight the controversy: 1) the
literature is ambiguous as to the efficacy of antidepressants in bipolar
depression, 2) antidepressants have the potential to induce a manic episode,
and 3) antidepressants may also induce a rapid-cycling course. Thase and
Denko (2008) reviewed the general literature, focusing especially on two
large clinical trials from the Stanley Foundation and the National Institute
of Mental Health (STEP-BD) that have attempted to clarify the benefits and
risks of antidepressant use in bipolar depression. The results of both trials
did not show that antidepressant augmentation of mood stabilizers
distinguished itself as more effective than placebo or the use of a second
mood stabilizer; however, possible benefit was noted for certain subgroups.
On the basis of the data, the authors were not able to recommend the use of
antidepressants or to conclude that antidepressants should be avoided
(Thase 2007).
Given these limitations, the American Psychiatric Association (2002)
has maintained its recommendations that primary treatment of bipolar
depression should be with a mood stabilizer and that antidepressant
augmentation of the mood stabilizer may be considered if there is limited or
no response.
There are no specific studies of the use of antidepressants in geriatric
populations. In a retrospective study of elderly inpatients who had
antidepressant-induced mania, Young et al. (2003) found that tricyclic
antidepressants were more likely than other antidepressants to induce
manias in late life, suggesting that the use of selective serotonin reuptake
inhibitors may be preferable in elderly patients.
Antipsychotic Agents
Atypical antipsychotic agents are increasingly being used for the treatment
of various phases of bipolar disorder. Olanzapine, risperidone, quetiapine
and quetiapine XR (extended release), ziprasidone, aripiprazole, and
asenapine are currently approved by the FDA for the treatment of acute
mania; olanzapine-fluoxetine combination, quetiapine and quetiapine XR,
and lurasidone are approved for the treatment of acute bipolar depression;
and olanzapine, aripiprazole, quetiapine and quetiapine XR, risperidone,
and ziprasidone are approved for maintenance phase treatment. However,
limited data are available about their efficacy in the geriatric population.
Beyer et al. (2001) reported on a pooled subanalysis of three double-
blind, placebo-controlled acute bipolar mania clinical trials with olanzapine,
focusing on subjects older than age 50 years. In comparison with placebo,
olanzapine was found to be efficacious for the treatment of acute mania
without any significant change in the side-effect profile. Information on
quetiapine, risperidone, clozapine, ziprasidone, aripiprazole, and asenapine
is much more limited. Case reports and open-label studies in geriatric
bipolar patient treatment are published for quetiapine (Madhusoodanan et
al. 2000), risperidone (Madhusoodanan et al. 1995, 1999), clozapine (Frye
et al. 1996; Shulman et al. 1997), and asenapine (Baruch et al. 2013). No
published reports are currently available for ziprasidone or aripiprazole.
In general, a lower-dose strategy in the elderly has been recommended
for most atypical antipsychotics (Alexopoulos et al. 2004), although this
may be less of a concern in the acute state. A major concern regarding
atypical antipsychotic use is the potential risk of metabolic abnormalities
such as obesity, diabetes, and dyslipidemia. This risk may be less of a
concern for elderly patients because they have less propensity for weight
gain and other metabolic effects associated with atypical antipsychotics
(Meyer 2002). In 2006, a black box warning was added to each of the
atypical antipsychotic agents, indicating that their use in elderly patients
with dementia may be associated with a higher incidence of death. The
exact mechanism of the increased mortality risk is unknown, although it
may be due to increased infections and cardiovascular causes. Limited
information is available on the risk of death in patients with late-life bipolar
disorder. A review of the U.S. Department of Veterans Affairs registries for
older patients with bipolar disorders showed differences in mortality risks
for various atypical antipsychotics; patients given risperidone had the
highest mortality rate, whereas those given quetiapine had the lowest
(Bhalerao et al. 2012). Although the authors did not believe that the data
could currently be translated to recommendations for use of one agent over
another, they proposed that the findings suggested a cautious approach to
the use of any atypical antipsychotics in older adults and recommended
using them judiciously when traditional mood stabilizers and
psychotherapies do not fully address the patient’s needs.
Electroconvulsive Therapy
Electroconvulsive therapy (ECT) has long been known to be effective for
the treatment of bipolar disorder. However, very limited data are available
on the use of ECT in elderly patients with bipolar disorder, especially when
compared with the quantity of literature on ECT for unipolar depression.
McDonald and Thompson (2001) reported on a case series of three elderly
manic patients who also had some dementia and were resistant to
pharmacotherapy but did respond to ECT treatment. Little et al. (2004)
reported on a case series of depressed elderly patients, which included five
patients with bipolar depression, who were treated with bifrontal ECT.
Key Points
• The prevalence of bipolar disorder, like other mental illnesses, decreases
with age. However, bipolar disorder in late life does continue to be a
frequent cause for admission to psychiatric inpatient facilities and
disruption of patients’ lives.
• The mortality rate for older adults with bipolar disorder is significantly
higher than that for the general population and for patients with unipolar
depression.
• The onset of bipolar disorder at a later age may be associated with fewer
genetic associations and more neurological illnesses.
• Treatment guidelines for late-life bipolar disorder are based primarily on
case reports and extrapolation from bipolar treatment in younger adults.
As in other geriatric treatment recommendations, the maxim “start low
and go slow” is applicable to late-life bipolar treatment.
• Elderly patients may require lower doses of lithium because of decreased
renal clearance.
References
Aldenkamp AP, De Krom M, Reijs R: Newer antiepileptic drugs and
cognitive issues. Epilepsia 44(suppl 4):21–29, 2003 12823566
Alexopoulos GS, Streim J, Carpenter D, et al: Using antipsychotic agents in
older patients. J Clin Psychiatry 65(suppl 2):5–99, discussion 100–102,
quiz 103–104, 2004 14994733
Al Jurdi RK, Nguyen QX, Petersen NJ, et al: Acute bipolar I affective
episode presentation across life span. J Geriatr Psychiatry Neurol
25(1):6–14, 2012 22467840
Almeida OP, Fenner S: Bipolar disorder: similarities and differences
between patients with illness onset before and after 65 years of age. Int
Psychogeriatr 14(3):311–322, 2002 12475092
Altshuler LL, Curran JG, Hauser P, et al: T2 hyperintensities in bipolar
disorder: magnetic resonance imaging comparison and literature meta-
analysis. Am J Psychiatry 152(8):1139–1144, 1995 7625460
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Practice guideline for the treatment of
patients with bipolar disorder (revision). Am J Psychiatry 159(4
suppl):1–50, 2002 11958165
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Angst J: The course of affective disorders, II: typology of bipolar manic-
depressive illness. Arch Psychiatr Nervenkr 226(1):65–73, 1978
708228
Angst J, Preisig M: Course of a clinical cohort of unipolar, bipolar and
schizoaffective patients: results of a prospective study from 1959 to
1985. Schweiz Arch Neurol Psychiatr 146(1):5–16, 1995 7792568
Angst J, Baastrup P, Grof P, et al: The course of monopolar depression and
bipolar psychoses. Psychiatr Neurol Neurochir 76(6):489–500, 1973
4781526
Aprahamian I, Ladeira RB, Diniz BS, et al: Cognitive impairment in
euthymic older adults with bipolar disorder: a controlled study using
cognitive screening tests. Am J Geriatr Psychiatry22(4):389–397, 2014
23567429
Aylward EH, Roberts-Twillie JV, Barta PE, et al: Basal ganglia volumes
and white matter hyperintensities in patients with bipolar disorder. Am
J Psychiatry 151(5):687–693, 1994 8166310
Balanzá-Martínez V, Tabarés-Seisdedos R, Selva-Vera G, et al: Persistent
cognitive dysfunctions in bipolar I disorder and schizophrenic patients:
a 3-year follow-up study. Psychother Psychosom 74(2):113–119, 2005
15741761
Baldessarini RJ, Vázquez G, Tondo L: Treatment of cyclothymic disorder:
commentary. Psychother Psychosom 80(3):131–135, 2011 21372620
Baron M, Mendlewicz J, Klotz J: Age-of-onset and genetic transmission in
affective disorders. Acta Psychiatr Scand 64(5):373–380, 1981
7347104
Bartels SJ, Forester B, Miles KM, et al: Mental health service use by
elderly patients with bipolar disorder and unipolar major depression.
Am J Geriatr Psychiatry 8(2):160–166, 2000 10804077
Baruch Y, Tadger S, Plopski I, et al: Asenapine for elderly bipolar manic
patients. J Affect Disord 145(1):130–132, 2013 22877962
Battino D, Croci D, Rossini A, et al: Serum carbamazepine concentrations
in elderly patients: a case-matched pharmacokinetic evaluation based
on therapeutic drug monitoring data. Epilepsia 44(7):923–929, 2003
12823575
Bearden CE, Hoffman KM, Cannon TD: The neuropsychology and
neuroanatomy of bipolar affective disorder: a critical review. Bipolar
Disord 3(3):106–150, discussion 151–153, 2001 11465675
Bellivier F, Golmard JL, Rietschel M, et al: Age at onset in bipolar I
affective disorder: further evidence for three subgroups. Am J
Psychiatry 160(5):999–1001, 2003 12727708
Beyer JL, Siegal A, Kennedy JS, et al: Olanzapine, divalproex, and placebo
treatment non-head-to-head comparisons of older adult acute mania.
Presented at the annual meeting of the International Psychogeriatric
Association, Nice, France, September 2001
Beyer JL, Kuchibhatla M, Payne M, et al: Caudate volume measurement in
older adults with bipolar disorder. Int J Geriatr Psychiatry 19(2):109–
114, 2004a 14758576
Beyer JL, Kuchibhatla M, Payne ME, et al: Hippocampal volume
measurement in older adults with bipolar disorder. Am J Geriatr
Psychiatry 12(6):613–620, 2004b 15545329
Beyer J, Kuchibhatla M, Gersing K, et al: Medical comorbidity in a bipolar
outpatient clinical population. Neuropsychopharmacology 30(2):401–
404, 2005 15536492
Beyer JL, Burchitt B, Gersing K, et al: Patterns of pharmacotherapy and
treatment response in elderly adults with bipolar disorder.
Psychopharmacol Bull 41(1):102–114, 2008 18362874
Bhalerao S, Seyfried LS, Kim HM, et al: Mortality risk with the use of
atypical antipsychotics in later-life bipolar disorder. J Geriatr
Psychiatry Neurol 25(1):29–36, 2012 22467844
Botteron KN, Vannier MW, Geller B, et al: Preliminary study of magnetic
resonance imaging characteristics in 8- to 16-year-olds with mania. J
Am Acad Child Adolesc Psychiatry 34(6):742–749, 1995 7608047
Bowden CL, Lawson DM, Cunningham M, et al: The role of divalproex in
the treatment of bipolar disorder. Psychiatr Ann 32:742–750, 2002
Bradley WG, Waluch V, Brant-Zawadzki M, et al: Patchy, periventricular
white matter lesions in the elderly: a common observation during NMR
imaging. Noninv Med Imag 1:35–41, 1984
Braffman BH, Zimmerman RA, Trojanowski JQ, et al: Brain MR:
pathologic correlation with gross and histopathology. 2. Hyperintense
white-matter foci in the elderly. AJR Am J Roentgenol 151(3):559–
566, 1988 3261518
Brennan PL, Nichols KA, Moos RH: Long-term use of VA mental health
services by older patients with substance use disorders. Psychiatr Serv
53(7):836–841, 2002 12096166
Broadhead J, Jacoby R: Mania in old age: a first prospective study. Int J
Geriatr Psychiatry 5:215–222, 1990
Brown FW, Lewine RJ, Hudgins PA, et al: White matter hyperintensity
signals in psychiatric and nonpsychiatric subjects. Am J Psychiatry
149(5):620–625, 1992 1575251
Carlson GA, Davenport YB, Jamison K: A comparison of outcome in
adolescent- and later-onset bipolar manic-depressive illness. Am J
Psychiatry 134(8):919–922, 1977 879359
Carlson GA, Bromet EJ, Sievers S: Phenomenology and outcome of
subjects with early and adult-onset psychotic mania. Am J Psychiatry
157(2):213–219, 2000 10671389
Cassidy F, Ahearn E, Carroll BJ: Elevated frequency of diabetes mellitus in
hospitalized manic-depressive patients. Am J Psychiatry 156(9):1417–
1420, 1999 10484954
Cassidy F, Ahearn EP, Carroll BJ: Substance abuse in bipolar disorder.
Bipolar Disord 3(4):181–188, 2001 11552957
Chen ST, Altshuler LL, Melnyk KA, et al: Efficacy of lithium vs. valproate
in the treatment of mania in the elderly: a retrospective study. J Clin
Psychiatry 60(3):181–186, 1999 10192594
Chimowitz MI, Estes ML, Furlan AJ, et al: Further observations on the
pathology of subcortical lesions identified on magnetic resonance
imaging. Arch Neurol 49(7):747–752, 1992 1497503
Chou KL, Mackenzie CS, Liang K, et al: Three-year incidence and
predictors of first-onset of DSM-IV mood, anxiety, and substance use
disorders in older adults: results from Wave 2 of the National
Epidemiologic Survey on Alcohol and Related Conditions. J Clin
Psychiatry 72(2):144–155, 2011 21382305
Chu D, Gildengers AG, Houck PR, et al: Does age at onset have clinical
significance in older adults with bipolar disorder? Int J Geriatr
Psychiatry 25(12):1266–1271, 2010 20082348
de Asis JM, Greenwald BS, Alexopoulos GS, et al: Frontal signal
hyperintensities in mania in old age. Am J Geriatr Psychiatry
14(7):598–604, 2006 16816013
DeBattista C, Schatzberg AF: Current psychotropic dosing and monitoring
guidelines. Prim Psychiatry 13(6):61–81, 2006
Delaloye C, Moy G, de Bilbao F, et al: Longitudinal analysis of cognitive
performances and structural brain changes in late-life bipolar disorder.
Int J Geriatr Psychiatry 26(12):1309–1318, 2011 21394788
Depp CA, Jeste DV: Bipolar disorder in older adults: a critical review.
Bipolar Disord 6(5):343–367, 2004 15383127
Depp CA, Jin H, Mohamed S, et al: Bipolar disorder in middle-aged and
elderly adults: is age of onset important? J Nerv Ment Dis
192(11):796–799, 2004 15505527
Depp CA, Lindamer LA, Folsom DP, et al: Differences in clinical features
and mental health service use in bipolar disorder across the lifespan.
Am J Geriatr Psychiatry 13(4):290–298, 2005 15845754
Depp CA, Davis CE, Mittal D, et al: Health-related quality of life and
functioning of middle-aged and elderly adults with bipolar disorder. J
Clin Psychiatry 67(2): 215–221, 2006 16566616
Depp CA, Moore DJ, Sitzer D, et al: Neurocognitive impairment in middle-
aged and older adults with bipolar disorder: comparison to
schizophrenia and normal comparison subjects. J Affect Disord 101(1–
3):201–209, 2007 17224185
Dhingra U, Rabins PV: Mania in the elderly: a 5–7 year follow-up. J Am
Geriatr Soc 39(6):581–583, 1991 2037748
Dols A, Kupka RW, van Lammeren A, et al: The prevalence of late-life
mania: a review. Bipolar Disord 16(2):113–118, 2014a 23919307
Dols A, Rhebergen D, Beekman A, et al: Psychiatric and medical
comorbidities: results from a bipolar elderly cohort study. Am J Geriatr
Psychiatry 22(11):1066–1074, 2014b 24495405
Dupont RM, Jernigan TL, Gillin JC, et al: Subcortical signal
hyperintensities in bipolar patients detected by MRI. Psychiatry Res
21(4):357–358, 1987 3628618
Dupont RM, Jernigan TL, Butters N, et al: Subcortical abnormalities
detected in bipolar affective disorder using magnetic resonance
imaging. Clinical and neuropsychological significance. Arch Gen
Psychiatry 47(1):55–59, 1990 2294856
Dupont RM, Jernigan TL, Heindel W, et al: Magnetic resonance imaging
and mood disorders. Localization of white matter and other subcortical
abnormalities. Arch Gen Psychiatry 52(9):747–755, 1995 7654126
Evans DL, Byerly MJ, Greer RA: Secondary mania: diagnosis and
treatment. J Clin Psychiatry 56(suppl 3):31–37, 1995 7883741
Fazekas F, Kleinert R, Offenbacher H, et al: Pathologic correlates of
incidental MRI white matter signal hyperintensities. Neurology
43(9):1683–1689, 1993 8414012
Figiel GS, Krishnan KR, Rao VP, et al: Subcortical hyperintensities on
brain magnetic resonance imaging: a comparison of normal and bipolar
subjects. J Neuropsychiatry Clin Neurosci 3(1):18–22, 1991 7580167
Foster JR: Use of lithium in elderly psychiatric patients: a review of the
literature. Lithium 3:77–93, 1992
Frye MA, Altshuler LL, Bitran JA: Clozapine in rapid cycling bipolar
disorder. J Clin Psychopharmacol 16(1):87–90, 1996 8834431
Fujikawa T, Yanai I, Yamawaki S: Psychosocial stressors in patients with
major depression and silent cerebral infarction. Stroke 28(6):1123–
1125, 1997 9183336
George AE, de Leon MJ, Kalnin A, et al: Leukoencephalopathy in normal
and pathologic aging: 2. MRI of brain lucencies. AJNR Am J
Neuroradiol 7(4):567–570, 1986 3088934
Ghaemi SN: Antidepressants in bipolar depression: the clinical debate. Aust
N Z J Psychiatry 46(4):298–301, 2012 22508588
Gildengers AG, Butters MA, Seligman K, et al: Cognitive functioning in
late-life bipolar disorder. Am J Psychiatry 161(4):736–738, 2004
15056521
Gildengers AG, Mulsant BH, Begley A, et al: The longitudinal course of
cognition in older adults with bipolar disorder. Bipolar Disord
11(7):744–752, 2009 19719787
Gildengers AG, Mulsant BH, Al Jurdi RK, et al: The relationship of bipolar
disorder lifetime duration and vascular burden to cognition in older
adults. Bipolar Disord 12(8):851–858, 2010 21176032
Gildengers A, Tatsuoka C, Bialko C, et al: Correlates of treatment response
in depressed older adults with bipolar disorder. J Geriatr Psychiatry
Neurol 25(1):37–42, 2012 22467845
Gildengers AG, Chisholm D, Butters MA, et al: Two-year course of
cognitive function and instrumental activities of daily living in older
adults with bipolar disorder: evidence for neuroprogression? Psychol
Med 43(4):801–811, 2013 22846332
Glasser M, Rabins P: Mania in the elderly. Age Ageing 13(4):210–213,
1984 6475649
Goodwin FK, Jamison KR: The natural course of manic depressive illness,
in Neurobiology of Mood Disorders. Edited by Post RM, Ballenger JC.
Baltimore, MD, Williams & Wilkens, 1984, pp 20–37
Goodwin FK, Jamison KR: Manic-Depressive Illness. New York, Oxford
University Press, 1990
Gunning-Dixon FM, Murphy CF, Alexopoulos GS, et al: Executive
dysfunction in elderly bipolar manic patients. Am J Geriatr Psychiatry
16(6):506–512, 2008 18515695
Hays JC, Krishnan KR, George LK, et al: Age of first onset of bipolar
disorder: demographic, family history, and psychosocial correlates.
Depress Anxiety 7(2):76–82, 1998 9614596
Hickie I, Scott E, Mitchell P, et al: Subcortical hyperintensities on magnetic
resonance imaging: clinical correlates and prognostic significance in
patients with severe depression. Biol Psychiatry 37(3):151–160, 1995
7727623
Himmelhoch JM, Neil JF, May SJ, et al: Age, dementia, dyskinesias, and
lithium response. Am J Psychiatry 137(8):941–945, 1980 7416295
Hirschfeld RM, Lewis L, Vornik LA: Perceptions and impact of bipolar
disorder: how far have we really come? Results of the national
depressive and manic-depressive association 2000 survey of
individuals with bipolar disorder. J Clin Psychiatry 64(2):161–174,
2003 12633125
Hopkinson G: A genetic study of affective illness in patients over 50. Br J
Psychiatry 110:244–254, 1964 14130477
Huang SH, Chung KH, Hsu JL, et al: The risk factors for elderly patients
with bipolar disorder having cerebral infarction. J Geriatr Psychiatry
Neurol 25(1):15–19, 2012 22467841
Hussein Z, Posner J: Population pharmacokinetics of lamotrigine
monotherapy in patients with epilepsy: retrospective analysis of routine
monitoring data. Br J Clin Pharmacol 43(5):457–465, 1997 9159559
James NM: Early and late-onset bipolar affective disorder. A genetic study.
Arch Gen Psychiatry 34(6):715–717, 1977 869667
Kessing LV: Diagnostic subtypes of bipolar disorder in older versus
younger adults. Bipolar Disord 8(1):56–64, 2006 16411981
Kessler RC, Rubinow DR, Holmes C, et al: The epidemiology of DSM-III-
R bipolar I disorder in a general population survey. Psychol Med
27(5):1079–1089, 1997 9300513
Klap R, Unroe KT, Unützer J: Caring for mental illness in the United
States: a focus on older adults. Am J Geriatr Psychiatry 11(5):517–524,
2003 14506085
Kocsis JH, Shaw ED, Stokes PE, et al: Neuropsychologic effects of lithium
discontinuation. J Clin Psychopharmacol 13(4):268–275, 1993
8376614
Koenig HG, Blazer DG: Epidemiology of geriatric affective disorders. Clin
Geriatr Med 8(2):235–251, 1992 1600475
Köhler S, Allardyce J, Verhey FR, et al: Cognitive decline and dementia
risk in older adults with psychotic symptoms: a prospective cohort
study. Am J Geriatr Psychiatry 21(2):119–128, 2013 23343485
Krabbendam L, Honig A, Wiersma J, et al: Cognitive dysfunctions and
white matter lesions in patients with bipolar disorder in remission. Acta
Psychiatr Scand 101(4):274–280, 2000 10782546
Lala SV, Sajatovic M: Medical and psychiatric comorbidities among elderly
individuals with bipolar disorder: a literature review. J Geriatr
Psychiatry Neurol 25(1):20–25, 2012 22467842
Laursen TM, Munk-Olsen T, Nordentoft M, et al: Increased mortality
among patients admitted with major psychiatric disorders: a register-
based study comparing mortality in unipolar depressive disorder,
bipolar affective disorder, schizoaffective disorder, and schizophrenia. J
Clin Psychiatry 68(6):899–907, 2007 17592915
Leboyer M, Henry C, Paillere-Martinot ML, et al: Age at onset in bipolar
affective disorders: a review. Bipolar Disord 7(2): 111–118, 2005
15762851
Lewandowski KE, Sperry SH, Malloy MC, et al: Age as a predictor of
cognitive decline in bipolar disorder. Am J Geriatr Psychiatry
22(12):1462–1468, 2014 24262287
Little JD, Atkins MR, Munday J, et al: Bifrontal electroconvulsive therapy
in the elderly: a 2-year retrospective. J ECT 20(3): 139–141, 2004
15342996
Madhusoodanan S, Brenner R, Araujo L, et al: Efficacy of risperidone
treatment for psychoses associated with schizophrenia, schizoaffective
disorder, bipolar disorder, or senile dementia in 11 geriatric patients: a
case series. J Clin Psychiatry 56(11):514–518, 1995 7592504
Madhusoodanan S, Brecher M, Brenner R, et al: Risperidone in the
treatment of elderly patients with psychotic disorders. Am J Geriatr
Psychiatry 7(2):132–138, 1999 10322240
Madhusoodanan S, Brenner R, Alcantra A: Clinical experience with
quetiapine in elderly patients with psychotic disorders. J Geriatr
Psychiatry Neurol 13(1):28–32, 2000 10753004
McDonald WM: Epidemiology, etiology, and treatment of geriatric mania. J
Clin Psychiatry 61(supp 13):3–11, 2000 11153809
McDonald WM, Thompson TR: Treatment of mania in dementia with
electroconvulsive therapy. Psychopharmacol Bull 35(2):72–82, 2001
12397888
McDonald WM, Krishnan KR, Doraiswamy PM, et al: Occurrence of
subcortical hyperintensities in elderly subjects with mania. Psychiatry
Res 40(4):211–220, 1991 1811239
McDonald WM, Tupler LA, Marsteller FA, et al: Hyperintense lesions on
magnetic resonance images in bipolar disorder. Biol Psychiatry
45(8):965–971, 1999 10386178
Mendlewicz J, Fieve RR, Rainer JD, et al: Manic-depressive illness: a
comparative study of patients with and without a family history. Br J
Psychiatry 120(558):523–530, 1972 5041532
Meyer JM: A retrospective comparison of weight, lipid, and glucose
changes between risperidone- and olanzapine-treated inpatients:
metabolic outcomes after 1 year. J Clin Psychiatry 63(5):425–433,
2002 12019668
Molinari V, Marmion J: Relationship between affective disorders and Axis
II diagnoses in geropsychiatric patients. J Geriatr Psychiatry Neurol
8(1):61–64, 1995 7710650
Okuma T, Yamashita I, Takahashi R, et al: Comparison of the antimanic
efficacy of carbamazepine and lithium carbonate by double-blind
controlled study. Pharmacopsychiatry 23(3):143–150, 1990 1973844
Oshima A, Higuchi T: Treatment guidelines for geriatric mood disorders.
Psychiatry Clin Neurosci 53(suppl): S55–S59, 1999 10560900
Panjehshahin MR, Bowmer CJ, Yates MS: Effect of valproic acid, its
unsaturated metabolites and some structurally related fatty acids on the
binding of warfarin and dansylsarcosine to human albumin. Biochem
Pharmacol 41(8):1227–1233, 1991 1706921
Perlis RH, Dennehy EB, Miklowitz DJ, et al: Retrospective age at onset of
bipolar disorder and outcome during two-year follow-up: results from
the STEP-BD study. Bipolar Disord 11(4):391–400, 2009 19500092
Petterson U: Manic-depressive illness. A clinical, social and genetic study.
Acta Psychiatr Scand Suppl 269(269):1–93, 1977 343505
Phillips ML: The neural basis of mood dysregulation in bipolar disorder.
Cogn Neuropsychiatry 11(3):233–249, 2006 17354070
Ponce H, Kunik M, Molinari V, et al: Divalproex sodium treatment in
elderly male bipolar patients. J Geriatr Drug Ther 12:55–63, 1999
Posner J, Holdrich T, Crome P: Comparison of lamotrigine
pharmacokinetics in young and elderly healthy volunteers. J Pharm
Med 1:121–128, 1991
Post F: The factor of ageing in affective illness, in Recent Developments in
Affective Disorders, Special Publication 2. Edited by Coppen A, Walk
A. London, Royal Medico-Psychological Association, 1968, pp 105–
116
Prien RF, Caffey EM Jr, Klett CJ: Relationship between serum lithium level
and clinical response in acute mania treated with lithium. Br J
Psychiatry 120(557):409–414, 1972 4557271
Ramsey CM, Spira AP, Mojtabai R, et al: Lifetime manic spectrum
episodes and all-cause mortality: 26-year follow-up of the NIMH
Epidemiologic Catchment Area Study. J Affect Disord 151(1):337–
342, 2013 23835104
Regenold WT, Prasad M: Uses of intravenous valproate in geriatric
psychiatry. Am J Geriatr Psychiatry 9(3):306–308, 2001 11481141
Regenold WT, Thapar RK, Marano C, et al: Increased prevalence of type 2
diabetes mellitus among psychiatric inpatients with bipolar I affective
and schizoaffective disorders independent of psychotropic drug use. J
Affect Disord 70(1):19–26, 2002 12113916
Rej S, Butters MA, Aizenstein HJ, et al: Neuroimaging and neurocognitive
abnormalities associated with bipolar disorder in old age. Int J Geriatr
Psychiatry 29(4):421–427, 2014 24006234
Robillard M, Conn DK: Lamotrigine use in geriatric patients with bipolar
depression. Can J Psychiatry 47(8):767–770, 2002 12420655
Roose SP, Bone S, Haidorfer C, et al: Lithium treatment in older patients.
Am J Psychiatry 136(6):843–844, 1979 443475
Sajatovic M: Treatment of bipolar disorder in older adults. Int J Geriatr
Psychiatry 17(9):865–873, 2002 12221662
Sajatovic M, Popli A, Semple W: Health resource utilization over a ten-year
period by geriatric veterans with schizophrenia and bipolar disorder. J
Geriatr Psychiatry Neurol 15:128–133, 1996 12230082
Sajatovic M, Gyulai L, Calabrese JR, et al: Maintenance treatment
outcomes in older patients with bipolar I disorder. Am J Geriatr
Psychiatry 13(4):305–311, 2005a 15845756
Sajatovic M, Madhusoodanan S, Coconcea N: Managing bipolar disorder in
the elderly: defining the role of the newer agents. Drugs Aging
22(1):39–54, 2005b 15663348
Sajatovic M, Blow FC, Ignacio RV: Psychiatric comorbidity in older adults
with bipolar disorder. Int J Geriatr Psychiatry 21(6):582–587, 2006
16783798
Sajatovic M, Ramsay E, Nanry K, et al: Lamotrigine therapy in elderly
patients with epilepsy, bipolar disorder or dementia. Int J Geriatr
Psychiatry 22(10):945–950, 2007 17326238
Samamé C, Martino DJ, Strejilevich SA: A quantitative review of
neurocognition in euthymic late-life bipolar disorder. Bipolar Disord
15(6):633–644, 2013 23651122
Shulman K, Post F: Bipolar affective disorder in old age. Br J Psychiatry
136:26–32, 1980 7357218
Shulman KI, Mackenzie S, Hardy B: The clinical use of lithium carbonate
in old age: a review. Prog Neuropsychopharmacol Biol Psychiatry
11(2–3):159–164, 1987 3114827
Shulman KI, Tohen M, Satlin A, et al: Mania compared with unipolar
depression in old age. Am J Psychiatry 149(3):341–345, 1992 1536272
Shulman RW, Singh A, Shulman KI: Treatment of elderly institutionalized
bipolar patients with clozapine. Psychopharmacol Bull 33(1):113–118,
1997 9133761
Shulman KI, Rochon P, Sykora K, et al: Changing prescription patterns for
lithium and valproic acid in old age: shifting practice without evidence.
BMJ 326(7396):960–961, 2003 12727769
Sibisi CDT: Sex differences in the age of onset of bipolar affective illness.
Br J Psychiatry 156:842–845, 1990 2207514
Snowdon J: A retrospective case-note study of bipolar disorder in old age.
Br J Psychiatry 158:485–490, 1991 2054563
Spar JE, Ford CV, Liston EH: Bipolar affective disorder in aged patients. J
Clin Psychiatry 40(12):504–507, 1979 500573
Speer DC: Differences in social resources and treatment history among
diagnostic groups of older adults. Hosp Community Psychiatry
43(3):270–274, 1992 1555823
Spicer CC, Hare EH, Slater E: Neurotic and psychotic forms of depressive
illness: evidence from age-incidence in a national sample. Br J
Psychiatry 123(576):535–541, 1973 4766651
Sproule BA, Hardy BG, Shulman KI: Differential pharmacokinetics of
lithium in elderly patients. Drugs Aging 16(3):165–177, 2000
10803857
Starkstein SE, Robinson RG, Price TR: Comparison of cortical and
subcortical lesions in the production of poststroke mood disorders.
Brain 110(Pt 4):1045–1059, 1987 3651794
Starkstein SE, Mayberg HS, Berthier ML, et al: Mania after brain injury:
neuroradiological and metabolic findings. Ann Neurol 27(6):652–659,
1990 2360802
Starkstein SE, Fedoroff P, Berthier ML, et al: Manic-depressive and pure
manic states after brain lesions. Biol Psychiatry 29(2): 149–158, 1991
1995084
Stenstedt A: A study in manic-depressive psychosis; clinical, social and
genetic investigations. Acta Psychiatr Neurol Scand 28(79):1–111,
1952 13007506
Stone K: Mania in the elderly. Br J Psychiatry 155:220–224, 1989 2597918
Strakowski SM, Woods BT, Tohen M, et al: MRI subcortical signal
hyperintensities in mania at first hospitalization. Biol Psychiatry
33(3):204–206, 1993 8448268
Subramaniam H, Dennis MS, Byrne EJ: The role of vascular risk factors in
late onset bipolar disorder. Int J Geriatr Psychiatry 22(8):733–737,
2007 17146839
Swayze VW2nd, Andreasen NC, Alliger RJ, et al: Structural brain
abnormalities in bipolar affective disorder. Ventricular enlargement and
focal signal hyperintensities. Arch Gen Psychiatry 47(11):1054–1059,
1990 2241506
Tanaka Y, Hazama H, Fukuhara T, et al: Computerized tomography of the
brain in manic-depressive patients—a controlled study. Folia Psychiatr
Neurol Jpn 36(2):137–143, 1982 6982210
Tariot PN, Podgorski CA, Blazina L, et al: Mental disorders in the nursing
home: another perspective. Am J Psychiatry 150(7):1063–1069, 1993
8317577
Taylor M, Abrams R: Manic states. A genetic study of early and late onset
affective disorders. Arch Gen Psychiatry 28(5):656–658, 1973 4700678
Thase ME: STEP-BD and bipolar depression: what have we learned? Curr
Psychiatry Rep 9(6):497–503, 2007 18221631
Thase ME, Denko T: Pharmacotherapy of mood disorders. Annu Rev Clin
Psychol 4:53–91, 2008 18370614
Tohen M, Shulman KI, Satlin A: First-episode mania in late life. Am J
Psychiatry 151(1):130–132, 1994 8267112
Tsai SY, Kuo CJ, Chen CC, et al: Risk factors for completed suicide in
bipolar disorder. J Clin Psychiatry 63(6):469–476, 2002 12088157
Tsai S, Lee H, Shang C, et al: The correlates of cognitive dysfunction in
early onset elderly bipolar patients. Presented at the international
congress of the International Psychogeriatrics Association, Chicago,
IL, August 2003
Umapathy C, Mulsant BH, Pollock BG: Bipolar disorder in the elderly.
Psychiatr Ann 30:473–480, 2000
Unützer J, Simon G, Pabiniak C, et al: The treated prevalence of bipolar
disorder in a large staff-model HMO. Psychiatr Serv 49(8):1072–1078,
1998 9712215
U.S. Food and Drug Administration: Carbamazepine prescribing
information to include recommendation of genetic test for patients with
Asian ancestry. FDA News, December 12, 2007. Available at
http://www.fda.gov/bbs/topics/NEWS/2007/NEW01755.html.
Accessed February 17, 2008.
Videbech P: MRI findings in patients with affective disorder: a meta-
analysis. Acta Psychiatr Scand 96(3):157–168, 1997 9296545
Weissman MM, Leaf PJ, Tischler GL, et al: Affective disorders in five
United States communities. Psychol Med 18(1):141–153, 1988
3363034
Weissman MM, Bland RC, Canino GJ, et al: Cross-national epidemiology
of major depression and bipolar disorder. JAMA 276(4):293–299, 1996
8656541
Winokur G: The Iowa 500: heterogeneity and course in manic-depressive
illness (bipolar). Compr Psychiatry 16(2):125–131, 1975 1120412
Wylie ME, Mulsant BH, Pollock BG, et al: Age at onset in geriatric bipolar
disorder. Effects on clinical presentation and treatment outcomes in an
inpatient sample. Am J Geriatr Psychiatry 7(1):77–83, 1999 9919324
Young RC, Falk JR: Age, manic psychopathology and treatment response.
Int J Geriatr Psychiatry 4:73–78, 1989
Young RC, Kalayam B, Tsuboyama G, et al: Mania: response to lithium
across the age spectrum (abstract). Soc Neurosci 18:669, 1992
Young RC, Nambudiri DE, Jain H, et al: Brain computed tomography in
geriatric manic disorder. Biol Psychiatry 45(8):1063–1065, 1999
10386193
Young RC, Jain H, Kiosses DN, et al: Antidepressant-associated mania in
late life. Int J Geriatr Psychiatry 18(5):421–424, 2003 12766919
Young RC, Gyulai L, Mulsant BH, et al: Pharmacotherapy of bipolar
disorder in old age: review and recommendations. Am J Geriatr
Psychiatry 12(4):342–357, 2004 15249272
Young RC, Murphy CF, Heo M, et al: Cognitive impairment in bipolar
disorder in old age: literature review and findings in manic patients. J
Affect Disord 92(1):125–131, 2006 16469389
Young RC, Mulsant BH, Sajatovic M, et al: GERI-BD: A randomized,
double-blind controlled trial of lithium and divalproex in the treatment
of mania in older patients with bipolar disorder, Session presentation at
the American Association of Geriatric Psychiatrists, San Antonio, TX,
2012
Zis AP, Grof P, Goodwin FK: The natural course of affective disorders:
implications for lithium prophylaxis, in Lithium: Controversies and
Unsolved Issues. Edited by Cooper TB, Gershon S, Kline NS, Shou M.
Amsterdam, The Netherlands, Excerpta Medica, 1979, pp 381–398
Suggested Readings
Beyer JL, Burchitt B, Gersing K, et al: Patterns of pharmacotherapy and
treatment response in elderly adults with bipolar disorder.
Psychopharmacol Bull 41:102–114, 2008
Depp CA, Jeste DV: Bipolar disorder in older adults: a critical review. Bipol
Disord 6:343–367, 2004
Sajatovic M, Madhusoodanan S, Coconcea N: Managing bipolar disorder in
the elderly: defining the role of the newer agents. Drugs Aging 22:39–
54, 2005
Young RC, Gyulai L, Mulsant BH, et al: Pharmacotherapy of bipolar
disorder in old age: review and recommendations. Am J Geriatr
Psychiatry 12:342–357, 2004
CHAPTER 11
Schizophrenia
Early-Onset Schizophrenia
Typically, individuals with schizophrenia develop the disorder in the second
or third decade of life (American Psychiatric Association 2000). Although
mortality rates in general, and suicide and homicide rates in particular, are
higher among individuals with schizophrenia than in the general population
(Hannerz et al. 2001; Hiroeh et al. 2001; Joukamaa et al. 2001), many
people with early-onset schizophrenia are now living into older adulthood.
Therefore, most of the older adults with schizophrenia have had an early
onset followed by a chronic course of illness spanning several decades. The
prevalence of schizophrenia is approximately 0.6% among adults ages 45–
64 and 0.1%–0.5% among elderly individuals (Castle and Murray 1993;
Copeland et al. 1998; Keith et al. 1991).
Longitudinal follow-up of patients with schizophrenia indicates
considerable heterogeneity of outcomes. A minority of patients experience
remission of both positive and negative symptoms (Ciompi 1980; Harding
et al. 1987; Huber 1997). Auslander and Jeste (2004) reported that nearly
10% of community-dwelling older patients with schizophrenia met strict
research criteria for sustained remission. A small proportion of patients
experience deterioration of symptoms. The course in a majority of patients
is largely unchanged over time (Belitsky and McGlashan 1993; Cohen
1990; Harvey et al. 1999), although there is generally an improvement in
positive symptoms (Jeste et al. 2003). A possibility of survivor bias—that
is, that the sickest patients die young, and the hardier ones survive into
older age—should be kept in mind in studying older patients with early-
onset illness.
Factors associated with poor prognosis for early-onset schizophrenia
include chronicity, insidious onset, premorbid psychosocial or functional
deficits, and prominent negative symptoms (Ram et al. 1992). In a sample
of chronically institutionalized patients with schizophrenia, older age was
associated with lower levels of positive symptoms and higher levels of
negative symptoms (Davidson et al. 1995). However, Harding (2002) noted
that the strength of the association between predictors of outcome and
actual outcome in patients with schizophrenia weakens over time.
Functional Capacity
The level of functional impairment varies considerably among older adults
with schizophrenia. In a study of a group of middle-aged and older
schizophrenia outpatients, Palmer et al. (2002) found that 30% had been
employed at least part time since the onset of psychosis, 43% were current
drivers, and 73% were living independently. In general, worse
neuropsychological test performance, lower educational level, and negative
symptoms but not positive symptoms are associated with poorer functional
capacity in older outpatients with schizophrenia (Evans et al. 2003).
Quality of Life
Self-appraisal is considered to be essential in studies of quality of life for
patients with schizophrenia. Several studies have found poorer self-assessed
quality of life to be associated with depression, positive and negative
symptoms, cognitive deficits, financial strain, poor social support, and poor
social skills (Vahia et al. 2007). These findings suggest that a multimodal
approach to management of these patients is necessary to improve quality
of life.
Late-Onset Schizophrenia
Historically, schizophrenia has been considered a disease of younger
adulthood. Kraepelin (1971) termed schizophrenia dementia praecox to
distinguish it from organic disorders arising in late life and to indicate a
poor prognosis with a course of progressive deterioration. However, in later
years, Kraepelin himself observed that some cases arose for the first time in
older age and that progressive decline was not a universal feature of the
disease. Bleuler (1943) and Roth (1955) developed this concept further,
with studies showing the late-onset phenotype to be a distinct entity from
the early-onset form. A literature review found that approximately 23% of
patients with schizophrenia had an onset after age 40, with 3% being older
than age 60 years (Harris and Jeste 1988). An investigation involving first-
contact patients found that 29% of patients had an onset after age 44, with
12% reporting onset after age 64 (Howard et al. 1993). The consensus
statement by the International Late-Onset Schizophrenia Group suggested
that schizophrenia with an onset after age 40 should be called “late-onset
schizophrenia” and should be considered a subtype of schizophrenia rather
than a related disorder (Howard et al. 2000). Although DSM-III-R labeled
schizophrenia with onset after age 45 as a late-onset type (American
Psychiatric Association 1987), later DSM editions have not included age-
related specifiers or criteria (American Psychiatric Association 1994, 2000,
2013). DSM-5 states that “late-onset cases can still meet the diagnostic
criteria for schizophrenia, but it is not yet clear whether this is the same
condition as schizophrenia diagnosed prior to mid-life” (American
Psychiatric Association 2013, p. 103).
Risk factors and clinical presentation associated with late-onset
schizophrenia are similar to those associated with early-onset schizophrenia
(Brodaty et al. 1999; Jeste et al. 1995). Similar proportions of individuals
with early-onset or late-onset schizophrenia reported having a family
history of schizophrenia (10%–15%) (Jeste et al. 1997b). Levels of
childhood maladjustment, measured retrospectively, were similar in early-
and late-onset schizophrenia patients and higher in both groups than in
healthy subjects (Jeste et al. 1997b). Patients with early- and late-onset
schizophrenia have increased rates of minor physical anomalies relative to
healthy subjects (Lohr et al. 1997).
Women predominate among individuals with onset of schizophrenia in
middle to late life (Häfner et al. 1998; Jeste et al. 1997b). It has been
speculated that estrogen may serve as an endogenous antipsychotic,
masking schizophrenic symptoms in vulnerable women until after
menopause (Seeman 1996). However, investigations on efficacy of
hormone replacement therapy as an adjunct treatment for postmenopausal
women with psychosis have not had promising results (Kulkarni et al. 1996,
2001; Lindamer et al. 2001).
Neuroimaging studies show that compared with patients with early-onset
schizophrenia, patients with late-onset schizophrenia have more nonspecific
structural abnormalities, such as enlarged ventricles and increased white
matter hyperintensities (Sachdev et al. 1999) and a larger volume of
thalamus on magnetic resonance imaging (Corey-Bloom et al. 1995). A
single small study employing diffusion tensor imaging recently found
evidence of abnormal white matter integrity in the left parietal lobe and
right posterior cingulum in patients with late-onset schizophrenia (n = 20)
compared with age-matched healthy control subjects (n = 17), although
there were no associations between these abnormalities and symptom levels
(Chen et al. 2013). Other imaging studies have ruled out strokes, tumors, or
other abnormalities as potential causes of schizophrenia in late life (Rivkin
et al. 2000). Finally, long-term neuropsychological follow-up of a group of
patients with late-onset schizophrenia revealed no evidence of cognitive
decline, suggesting a neurodevelopmental rather than a neurodegenerative
process (Palmer et al. 2003).
Most studies have found that compared with patients with early-onset
disease, patients with late-onset schizophrenia have lower levels of positive
symptoms and a lower daily antipsychotic dose requirement (Vahia et al.
2010). Patients with late-onset schizophrenia tend to have more organized
delusions, auditory hallucinations or hallucinations with a running
commentary, and persecutory delusions with and without hallucinations
(Howard et al. 2000). Data from initial smaller studies suggested a higher
prevalence of the paranoid subtype of schizophrenia and lower levels of
negative symptoms on average among patients with late-onset
schizophrenia (approximately 75%) than among patients with early-onset
schizophrenia (approximately 50%) (Jeste et al. 1997b). However, a more
recent larger study from our center, including data collected over 20 years
comparing 744 early-onset schizophrenia and 110 late-onset schizophrenia
patients, found that the two groups had similar relative proportions of
patients with the paranoid subtype and no differences in severity of negative
symptoms (Vahia et al. 2010).
On neuropsychological testing, after correction for age, education, and
gender, patients with late-onset schizophrenia tend to have less impairment
in learning, abstraction, and flexibility in thinking than patients with early-
onset schizophrenia (Jeste et al. 1997b). Compared with patients with early-
onset schizophrenia, a greater proportion of patients with late-onset
schizophrenia have successful occupational and marital histories and
generally higher premorbid functioning.
Sensory deficits, particularly hearing loss, are associated with psychotic
symptoms in late life and have been proposed as a risk factor for late-onset
schizophrenia (Howard et al. 1994; Raghuram et al. 1980). However, other
data suggest that patients with either early- or late-onset schizophrenia may
be less likely than healthy older adults to receive appropriate correction for
vision and hearing impairments (Prager and Jeste 1993). Thus, uncorrected
sensory deficits may reflect generally poorer health care utilization by older
psychotic patients and may not be a potential cause of psychosis in the
elderly population.
In summary, early- and late-onset schizophrenia share similarities in
many key clinical characteristics. However, there are important differences,
including the greater proportion of women, lower average severity of
positive symptoms, and lower average antipsychotic dose requirement in
late-onset schizophrenia. More analyses of large data sets and studies
designed to elucidate the pathophysiological pathways underlying early-
and late-onset schizophrenia are needed to better understand how these two
groups differ from each other (Jeste et al. 2005).
Delusional Disorder
At least 6% of older individuals have paranoid symptoms such as
persecutory delusions, but most of these individuals have a neurocognitive
disorder (Christenson and Blazer 1984; Forsell and Henderson 1998;
Henderson et al. 1998). The essential feature of a delusional disorder is a
nonbizarre delusion (e.g., a persecutory, somatic, erotomanic, grandiose, or
jealous delusion) without prominent auditory or visual hallucinations. The
symptoms must be present for at least 1 month. When delusional disorder
arises in late life, basic personality features, intellectual performance, and
occupational function are preserved, but social functioning is compromised.
To diagnose delusional disorder, the clinician must rule out delirium, major
neurocognitive disorder, psychotic disorder due to another medical
condition or due to substance or medication use, schizophrenia, and mood
disorders with psychotic features. The course of persecutory delusional
disorder is typically chronic, but individuals with other types of delusions
may have partial remissions and relapses.
Treatment
The modern era of pharmacological treatment for schizophrenia and other
psychotic disorders began with the introduction of chlorpromazine in the
early 1950s. Although this and other conventional agents substantially
improved the positive symptoms of schizophrenia (e.g., hallucinations and
delusions), a number of treatment liabilities have been recognized over the
years, such as movement disorders, sedation, orthostatic hypotension, and
increased prolactin concentrations. In addition, older adults have a
significantly higher risk for developing tardive dyskinesia than do younger
adults, making use of conventional antipsychotics in this population highly
problematic.
Therefore, when atypical antipsychotics—which are associated with
significantly lower incidence of tardive dyskinesia—were introduced, they
were hailed as the drugs of choice for older adults with psychotic disorders.
However, these agents have since been linked to an increased risk of
metabolic dysfunction, including diabetes, dyslipidemia, and obesity,
thereby leading to a worsened cardiovascular risk profile. In elderly patients
with dementia, atypical antipsychotics have been associated with an
increased risk of cerebrovascular adverse events and mortality compared
with placebo; therefore, leading pharmaceutical regulatory agencies have
issued warnings about the use of these agents in patients with dementia
(Meeks and Jeste 2008). At the same time, because of the paucity of
evidence-based pharmacological treatment alternatives to antipsychotics for
patients with dementia, clinicians are restricted to off-label treatments,
which must be used with caution and close monitoring. Psychosocial
treatments for older adults with psychosis have been developed and tested
in randomized controlled trials (RCTs) and show promise as primary or
adjunctive treatments.
Pharmacological Treatments
Pharmacotherapy for older adults with chronic psychotic disorders can be
challenging. Although few randomized, placebo-controlled, double-blind
clinical trials have been conducted in this population, some information has
become available. Maintenance pharmacotherapy is usually required for
older patients with schizophrenia due to risk of relapse. Because older
patients are at higher risk of adverse antipsychotic effects, due to age-
related pharmacokinetic and pharmacodynamic factors (Hämmerlein et al.
1998), coexisting medical illnesses, and concomitant medications, the
recommended starting and maintenance doses of antipsychotics in older
adults are much lower than the usual doses in younger adults (Lehman et al.
2004). Patients with late-onset schizophrenia respond well to low-dose
antipsychotic medication, requiring about 50% of the dose typically taken
by older patients with early-onset schizophrenia and 25%–33% of the dose
used in younger patients with schizophrenia.
Use of conventional or typical antipsychotics in older adults is
problematic because of the higher incidence of tardive dyskinesia in older
patients. Aging appears to be the most important risk factor for the
development of tardive dyskinesia (American Psychiatric Association 2000;
Yassa and Jeste 1992). The cumulative 1-year incidence of tardive
dyskinesia is 29% among older patients (mean age 65 years) despite low
dosing (Jeste et al. 1999b), whereas the annual cumulative incidence of
tardive dyskinesia in young adults is 4%–5% (Kane et al. 1993). The risk of
severe tardive dyskinesia is also higher in older patients (Caligiuri et al.
1997). Other side effects of conventional neuroleptics include sedation,
anticholinergic effects, cardiovascular effects including orthostatic
hypotension, parkinsonian reactions, and neuroleptic malignant syndrome.
Despite these side effects, occasionally a conventional antipsychotic may be
the most reasonable treatment option for an individual patient, and these
agents can be used at flexible, individualized low doses to minimize side
effects (Jeste et al. 1999b).
Few efficacy comparisons have been done of conventional
antipsychotics versus atypical antipsychotics in patients with schizophrenia
over age 65. In a study of 42 elderly inpatients, Howanitz et al. (1999)
found that clozapine (≤300 mg/day) and chlorpromazine (≤600 mg/day) had
similar efficacy. Kennedy et al. (2003) compared olanzapine (5–20 mg/day)
and haloperidol (5–20 mg/day) in a 6-week trial of 117 patients ages 60
years and older who had schizophrenia and related disorders. Olanzapine
(mean modal dose 11.9 mg/day) produced significantly greater
symptomatic improvement and was associated with fewer motor side
effects than haloperidol (mean modal dose 9.4 mg/day) (Kennedy et al.
2003). The National Institute of Mental Health’s Clinical Antipsychotic
Trials of Intervention Effectiveness (CATIE) study (Lieberman et al. 2005),
which included adults ages 18–65, found no significant differences in
effectiveness between the conventional antipsychotic perphenazine and the
atypical antipsychotics risperidone, olanzapine, quetiapine, or ziprasidone,
but it is unknown how these findings would translate to patients older than
age 65.
Generally, atypical antipsychotics carry a much lower risk of tardive
dyskinesia than conventional neuroleptics, even when taken by very high-
risk patients such as middle-aged and older adults with borderline tardive
dyskinesia (Dolder and Jeste 2003; Jeste et al. 1999a). Clozapine has shown
efficacy in reducing tardive dyskinesia in patients with existing tardive
dyskinesia (Kane et al. 1993; Lieberman et al. 1991; Simpson et al. 1978;
Small et al. 1987); however, other side effects limit its use, particularly in
elderly patients. A beneficial effect of other atypical agents, specifically
risperidone and olanzapine, on preexisting tardive dyskinesia has also been
reported (Jeste et al. 1997a; Kinon et al. 2004; Littrell et al. 1998; Street et
al. 2000).
Atypical antipsychotics have a less favorable side-effect profile,
however, in terms of metabolic function. Common metabolic side effects
include excessive weight gain and obesity, glucose intolerance, new-onset
type II diabetes mellitus, diabetic ketoacidosis, and dyslipidemia (Allison et
al. 1999; Jin et al. 2002, 2004; Wirshing et al. 1998). Although there are no
guidelines for management of these side effects specifically in older
patients with schizophrenia, the monitoring recommendations developed by
the American Diabetes Association et al. (2004) are potentially applicable.
Because elderly patients tend to be at higher risk for cardiovascular disease
than younger patients, closer monitoring is necessary for older adults.
The short-term benefit of risperidone and olanzapine for treatment of
psychotic symptoms in middle-aged and older adults with schizophrenia has
been supported in several double-blind trials (Feldman et al. 2003; Jeste et
al. 2003; Riedel et al. 2009; Suzuki et al. 2011), and one short-term trial
suggests that paliperidone might be of benefit (Tzimos et al. 2008). Data
supporting the short-term benefit of other atypical antipsychotics come only
from open-label or retrospectively designed studies. Although clozapine has
been shown to have superior effectiveness than other antipsychotics in
younger adults (Jones et al. 2006; Lieberman et al. 2005), the medication is
difficult to use in elderly persons due to the risk of leukopenia and
agranulocytosis, as well as other side effects such as orthostasis, sedation,
and anticholinergic effects. The necessity of weekly blood draws also may
pose a problem for older patients.
Data from a study by Jin et al. (2012) raise serious concerns about the
longer-term safety and effectiveness of atypical antipsychotics in middle-
aged and older adults. The use of aripiprazole, olanzapine, quetiapine, and
risperidone was studied in 332 outpatients older than age 40 years who had
psychotic symptoms related to a variety of diagnoses, including
schizophrenia, mood disorders, posttraumatic stress disorder, or
neurocognitive disorder. The patients were followed for up to 2 years. The
high 1-year cumulative incidence of metabolic syndrome (36% in 1 year)
and high rates of both serious (23.7%) and nonserious (50.8%) adverse
events observed were particularly concerning given that no significant
improvement in psychopathology was detected. Over half of the study
participants discontinued their medication within 6 months, often due to
side effects (51.6%) or lack of efficacy (26%), and the quetiapine arm of the
study was discontinued early because the incidence of serious adverse
events was found to be twice that of the other three atypical antipsychotics.
The concerns about the long-term safety and efficacy of atypical
antipsychotics in middle-aged and older adults, combined with the data on
the increased risk of strokes and mortality in elderly patients with dementia
treated with atypical antipsychotics and the consequent U.S. Food and Drug
Administration (FDA) black box warnings (discussed in “Treatment of
Psychosis of Alzheimer’s Disease and Other Neurocognitive Disorders,”
later in this chapter), underscore the need for clinicians to exercise caution
when prescribing these drugs for older patients with schizophrenia. We
strongly recommend educating patients and their caregivers about the
potential risks and benefits of medications and encouraging shared decision
making. If the decision is made to use an antipsychotic medication, it is
generally best to start with a low initial dose (25%–50% of that used in a
younger patient) and titrate slowly. In patients who have been on stable
dosages of antipsychotic medications for long periods of time, clinicians
should consider gradual and incremental decreases to determine the lowest
effective dosage. Patients should be monitored closely for medication
effectiveness and for possible side effects.
Few data are available regarding the safety and effectiveness of
pharmacological treatment specifically of VLOSLP or delusional disorder
in older individuals. A single small retrospective case study of 8 outpatients
and 13 inpatients with VLOSLP concluded that atypical antipsychotics
(aripiprazole, risperidone, olanzapine, and quetiapine) could be helpful at
low dosages. Alexopoulos et al.’s (2004) survey of 48 experts in geriatric
care found antipsychotics to be the only recommended treatment for
delusional disorder in older adults, and the most favored recommendation
for older adults with delusional disorder was risperidone (0.75–2.5 mg/day),
followed by olanzapine (5–10 mg/day) and quetiapine (50–200 mg/day).
More research is required regarding pharmacological treatment of these
conditions in older adults.
Psychosocial Treatments
Over the past decade, effective psychosocial interventions for older adults
with chronic psychotic disorders have been developed. Granholm et al.
(2005) conducted an RCT to examine the effects of adding cognitive-
behavioral social skills training (CBSST) to treatment as usual for 76
middle-aged and elderly stable outpatients with schizophrenia. This training
intervention teaches cognitive and behavioral coping techniques, social
functioning skills, problem-solving techniques, and compensatory aids for
neurocognitive impairments. The investigators found that CBSST led to
significantly increased frequency of social functioning activities, greater
cognitive insight (more objectivity in reappraising psychotic symptoms),
and greater skill mastery. At 12-month follow-up, the CBSST group had
maintained their greater skill acquisition and performance of everyday
living skills. The greater cognitive insight seen in the CBSST group at the
end of the treatment was not maintained at 12-month follow-up, however,
suggesting a possible need for booster sessions (Granholm et al. 2007).
Patterson et al. (2006) conducted an RCT to compare a behavioral group
intervention called Functional Adaptation Skills Training (FAST) with a
time-equivalent attention control condition. FAST is a manualized
behavioral intervention designed to improve everyday living skills
(including medication management, social skills, communication skills,
organization and planning, transportation, and financial management) of
middle-aged and older adults with schizophrenia or schizoaffective
disorder. Compared with participants randomized to attention control, the
FAST group showed significant improvement in daily living skills and
social skills but not medication management. The FAST intervention has
also been culturally adapted and pilot-tested in middle-aged and older
Spanish-speaking Mexican American patients with schizophrenia or
schizoaffective disorder. This intervention, called Programa de
Entrenamiento para el Desarrollo de Aptitudes para Latinos (PEDAL), was
compared with a time-equivalent friendly support group in a randomized
controlled pilot study (Patterson et al. 2005). The PEDAL group
demonstrated a significant improvement in everyday living skills that was
maintained at 12-month follow-up.
Helping Older People Experience Success (HOPES), a 12-month
program combining social skills training and a nurse-administered
preventive health care program, was associated with improved community
living skills and functioning, greater self-efficacy, and lower levels of
negative symptoms in adults over age 50 with serious mental illness
including schizophrenia. Improvement in community living skills was
maintained at 3-year follow-up (Bartels et al. 2014; Mueser et al. 2010).
Following an examination of employment outcomes among middle-aged
and older adults with schizophrenia who each participated in one of three
types of work rehabilitation program, Twamley et al. (2005) reported that
the highest rates of volunteer or paid work (81%) and competitive/paid
work (69%) occurred for the patients who were placed in a job chosen with
a vocational counselor and who then received individualized on-site
support. The less successful programs (achieving at best a 44% rate of
volunteer or paid work) employed a train-then-place approach.
Conclusion
Chronic psychotic disorders of late life may include early-onset
schizophrenia (onset before age 40), late-onset schizophrenia (onset
between ages 40 and 60), very-late-onset schizophrenia-like psychosis
(onset after age 60), delusional disorder, and psychosis related to
Alzheimer’s disease or other neurocognitive disorders. Psychosocial
treatments have an important place as an adjunctive treatment for older
adults with psychosis. Aging is associated with increased risk for
antipsychotic-related adverse reactions. Compared with younger patients,
older adults have a much higher risk of developing tardive dyskinesia, and
atypical antipsychotics are associated with metabolic liabilities. There are
also concerns about the long-term safety and effectiveness of atypical
antipsychotics in older patients. No FDA-approved treatments are currently
available for psychosis and agitation in neurocognitive disorders, and
atypical antipsychotic use in this population has been associated with an
increased risk of cerebrovascular adverse events and mortality. Use of
pharmacological treatments for older adults with psychosis therefore
requires careful consideration of the potential risks and benefits as well as
shared decision making with patients and their caregivers.
Key Points
• Schizophrenia may be classified by age at onset into early-onset
schizophrenia (onset before age 40), late-onset schizophrenia (onset
between ages 40 and 60), and very-late-onset schizophrenia-like
psychosis (onset after age 60).
• There is considerable heterogeneity of course and outcome with aging in
patients with early-onset schizophrenia, although there is generally an
improvement in positive symptoms.
• Patients with late-onset schizophrenia are similar to patients with early-
onset schizophrenia in terms of risk factors, clinical presentation, family
history of schizophrenia, and response to medications. However, women
are overrepresented among the late-onset patients. Also, late-onset
schizophrenia is characterized by lower average severity of positive
symptoms, and a lower average antipsychotic dose requirement.
• Very-late-onset schizophrenia-like psychosis is a heterogeneous entity
with varied etiology.
• Patients with psychosis of Alzheimer’s disease tend to have paranoid
delusions and visual or auditory hallucinations, as well as greater risk of
agitation, faster cognitive decline, and greater likelihood of being
institutionalized than patients with Alzheimer’s disease without
psychosis.
• Compared with younger patients, older adults have a much higher risk of
developing tardive dyskinesia. Although atypical antipsychotics are
associated with significantly lower risk of tardive dyskinesia than
conventional agents, they have problematic metabolic liabilities.
• Psychosocial treatments have an important place as an adjunctive
treatment for older adults with schizophrenia.
• There are currently no FDA-approved treatments for psychosis and
agitation in dementia; however, off-label use of medications, as well as
certain psychosocial interventions, may be appropriate.
• There are concerns about longer-term safety as well as effectiveness of
atypical antipsychotics in older patients with psychotic disorders.
Additionally, atypical antipsychotic use in patients with dementia has
been associated with an increased risk of cerebrovascular adverse events
and mortality, leading the FDA to issue black box warnings for this
population.
• Principles of pharmacotherapy for older adults with psychosis include
careful consideration of indications, shared decision making, and use of
the lowest effective doses for the shortest possible time periods.
References
Alexopoulos GS, Streim J, Carpenter D, et al: Using antipsychotic agents in
older patients. J Clin Psychiatry 65 (suppl 2):5–99, discussion 100–
102, quiz 103–104, 2004 14994733
Allison DB, Mentore JL, Heo M, et al: Antipsychotic-induced weight gain:
a comprehensive research synthesis. Am J Psychiatry 156(11):1686–
1696, 1999 10553730
American Diabetes Association, American Psychiatric Association,
American Association of Clinical Endocrinologists, et al: Consensus
development conference on antipsychotic drugs and obesity and
diabetes. J Clin Psychiatry 65(2):267–272, 2004 15003083
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 3rd Edition, Revised. Washington, DC, American
Psychiatric Press, 1987
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Andreasen NC: I don’t believe in late-onset schizophrenia, in Late-Onset
Schizophrenia. Edited by Howard R, Rabins PV, Castle DJ.
Philadelphia, PA, Wrightson Biomedical, 1999, pp 111–123
Auslander LA, Jeste DV: Sustained remission of schizophrenia among
community-dwelling older outpatients. Am J Psychiatry 161(8):1490–
1493, 2004 15285980
Ayalon L, Gum AM, Feliciano L, Areán PA: Effectiveness of
nonpharmacological interventions for the management of
neuropsychiatric symptoms in patients with dementia: a systematic
review. Arch Intern Med 166(20):2182–2188, 2006 17101935
Ballard C, Corbett A: Agitation and aggression in people with Alzheimer’s
disease. Curr Opin Psychiatry 26(3):252–259, 2013 23528917
Ballard C, Waite J: The effectiveness of atypical antipsychotics for the
treatment of aggression and psychosis in Alzheimer’s disease.
Cochrane Database Syst Rev (1):CD003476, 2006 16437455
Bartels SJ, Pratt SI, Mueser KT, et al: Long-term outcomes of a randomized
trial of integrated skills training and preventive healthcare for older
adults with serious mental illness. Am J Geriatr Psychiatry
22(11):1251–1261, 2014 23954039
Baynes D, Mulholland C, Cooper SJ, et al: Depressive symptoms in stable
chronic schizophrenia: prevalence and relationship to psychopathology
and treatment. Schizophr Res 45(1-2):47–56, 2000 10978872
Belitsky R, McGlashan TH: The manifestations of schizophrenia in late
life: a dearth of data. Schizophr Bull 19(4):683–685, 1993 8303218
Bleuler M: Die spatschizophrenen Krankheitsbilder. Fortschr Neurol
Psychiatr Grenzgeb 15:259–290, 1943
Bonuccelli U, Ceravolo R, Salvetti S, et al: Clozapine in Parkinson’s
disease tremor. Effects of acute and chronic administration. Neurology
49(6):1587–1590, 1997 9409351
Brodaty H, Sachdev P, Rose N, et al: Schizophrenia with onset after age 50
years, I: phenomenology and risk factors. Br J Psychiatry 175:410–415,
1999 10789270
Burns A, Jacoby R, Levy R: Psychiatric phenomena in Alzheimer’s disease,
I: disorders of thought content. Br J Psychiatry 157:72–76, 92–94,
1990a 2397365
Burns A, Jacoby R, Levy R: Psychiatric phenomena in Alzheimer’s disease.
II: Disorders of perception. Br J Psychiatry 157:76–81, 92–94, 1990b
2397366
Caligiuri MP, Lacro JP, Rockwell E, et al: Incidence and risk factors for
severe tardive dyskinesia in older patients. Br J Psychiatry 171:148–
153, 1997 9337951
Castle DJ, Murray RM: The epidemiology of late-onset schizophrenia.
Schizophr Bull 19(4):691–700, 1993 8303220
Cervantes AN, Rabins PV, Slavney PR: Onset of schizophrenia at age 100.
Psychosomatics 47(4):356–359, 2006 16844897
Chan WC, Lam LC, Choy CN, et al: A double-blind randomised
comparison of risperidone and haloperidol in the treatment of
behavioural and psychological symptoms in Chinese dementia patients.
Int J Geriatr Psychiatry 16(12):1156–1162, 2001 11748775
Chen L, Chen X, Liu W, et al: White matter microstructural abnormalities
in patients with late-onset schizophrenia identified by a voxel-based
diffusion tensor imaging. Psychiatry Res 212(3):201–207, 2013
23146248
Chou KL, Borek LL, Friedman JH: The management of psychosis in
movement disorder patients. Expert Opin Pharmacother 8(7):935–943,
2007 17472539
Christenson R, Blazer D: Epidemiology of persecutory ideation in an
elderly population in the community. Am J Psychiatry 141(9):1088–
1091, 1984 6235752
Ciompi L: Catamnestic long-term study on the course of life and aging of
schizophrenics. Schizophr Bull 6(4):606–618, 1980 7444392
Cohen CI: Outcome of schizophrenia into later life: an overview.
Gerontologist 30(6):790–797, 1990 2286338
Cohen-Mansfield J: Nonpharmacologic interventions for inappropriate
behaviors in dementia: a review, summary, and critique. Am J Geriatr
Psychiatry 9(4):361–381, 2001 11739063
Cooper AF, Curry AR: The pathology of deafness in the paranoid and
affective psychoses of later life. J Psychosom Res 20(2):97–105, 1976
1271318
Copeland JRM, Dewey ME, Scott A, et al: Schizophrenia and delusional
disorder in older age: community prevalence, incidence, comorbidity,
and outcome. Schizophr Bull 24(1):153–161, 1998 9502553
Corey-Bloom J, Jernigan T, Archibald S, et al: Quantitative magnetic
resonance imaging of the brain in late-life schizophrenia. Am J
Psychiatry 152(3):447–449, 1995 7864275
Davidson M, Harvey PD, Powchik P, et al: Severity of symptoms in
chronically institutionalized geriatric schizophrenic patients. Am J
Psychiatry 152(2):197–207, 1995 7840352
De Deyn PP, Rabheru K, Rasmussen A, et al: A randomized trial of
risperidone, placebo, and haloperidol for behavioral symptoms of
dementia. Neurology 53(5):946–955, 1999 10496251
Devanand DP, Jacobs DM, Tang MX, et al: The course of psychopathologic
features in mild to moderate Alzheimer disease. Arch Gen Psychiatry
54(3):257–263, 1997 9075466
Diwan S, Cohen CI, Bankole AO, et al: Depression in older adults with
schizophrenia spectrum disorders: prevalence and associated factors.
Am J Geriatr Psychiatry 15(12):991–998, 2007 18056817
Dolder CR, Jeste DV: Incidence of tardive dyskinesia with typical versus
atypical antipsychotics in very high risk patients. Biol Psychiatry
53(12):1142–1145, 2003 12814866
Evans JD, Paulsen JS, Harris MJ, et al: A clinical and neuropsychological
comparison of delusional disorder and schizophrenia. J
Neuropsychiatry Clin Neurosci 8(3):281–286, 1996 8854299
Evans JD, Heaton RK, Paulsen JS, et al: The relationship of
neuropsychological abilities to specific domains of functional capacity
in older schizophrenia patients. Biol Psychiatry 53(5):422–430, 2003
12614995
Feldman PD, Kaiser CJ, Kennedy JS, et al: Comparison of risperidone and
olanzapine in the control of negative symptoms of chronic
schizophrenia and related psychotic disorders in patients aged 50 to 65
years. J Clin Psychiatry 64(9):998–1004, 2003 14628974
Forsell Y, Henderson AS: Epidemiology of paranoid symptoms in an
elderly population. Br J Psychiatry 172:429–432, 1998 9747406
Gill SS, Rochon PA, Herrmann N, et al: Atypical antipsychotic drugs and
risk of ischaemic stroke: population based retrospective cohort study.
BMJ 330(7489):445, 2005 15668211
Granholm E, McQuaid JR, McClure FS, et al: A randomized controlled
pilot study of cognitive behavioral social skills training for older
patients with schizophrenia (letter). Schizophr Res 53(1-2):167–169,
2002 11728848
Granholm E, McQuaid JR, McClure FS, et al: A randomized, controlled
trial of cognitive behavioral social skills training for middle-aged and
older outpatients with chronic schizophrenia. Am J Psychiatry
162(3):520–529, 2005 15741469
Granholm E, McQuaid JR, McClure FS, et al: Randomized controlled trial
of cognitive behavioral social skills training for older people with
schizophrenia: 12-month follow-up. J Clin Psychiatry 68(5):730–737,
2007 17503982
Häfner H, an der Heiden W, Behrens S, et al: Causes and consequences of
the gender difference in age at onset of schizophrenia. Schizophr Bull
24(1):99–113, 1998 9502549
Hämmerlein A, Derendorf H, Lowenthal DT: Pharmacokinetic and
pharmacodynamic changes in the elderly. Clinical implications. Clin
Pharmacokinet 35(1):49–64, 1998 9673834
Hannerz H, Borgå P, Borritz M: Life expectancies for individuals with
psychiatric diagnoses. Public Health 115(5):328–337, 2001 11593442
Harding C: Changes in schizophrenia across time, in Schizophrenia Into
Later Life. Edited by Cohen CI. Washington, DC, American
Psychiatric Publishing, 2002, pp 19–42
Harding CM, Brooks GW, Ashikaga T, et al: Aging and social functioning
in once-chronic schizophrenic patients 22–62 years after first
admission: the Vermont story, in Schizophrenia and Aging. Edited by
Miller NE, Cohen GD. New York, Guilford, 1987, pp 74–82
Harris MJ, Jeste DV: Late-onset schizophrenia: an overview. Schizophr
Bull 14(1): 39–55, 1988 3291094
Harvey PD, Silverman JM, Mohs RC, et al: Cognitive decline in late-life
schizophrenia: a longitudinal study of geriatric chronically hospitalized
patients. Biol Psychiatry 45(1):32–40, 1999 9894573
Heaton RK, Gladsjo JA, Palmer BW, et al: Stability and course of
neuropsychological deficits in schizophrenia. Arch Gen Psychiatry
58(1):24–32, 2001 11146755
Henderson AS, Korten AE, Levings C, et al: Psychotic symptoms in the
elderly: a prospective study in a population sample. Int J Geriatr
Psychiatry 13(7):484–492, 1998 9695039
Herrmann N, Mamdani M, Lanctôt KL: Atypical antipsychotics and risk of
cerebrovascular accidents. Am J Psychiatry 161(6):1113–1115, 2004
15169702
Hiroeh U, Appleby L, Mortensen PB, et al: Death by homicide, suicide, and
other unnatural causes in people with mental illness: a population-
based study. Lancet 358(9299):2110–2112, 2001 11784624
Howanitz E, Pardo M, Smelson DA, et al: The efficacy and safety of
clozapine versus chlorpromazine in geriatric schizophrenia. J Clin
Psychiatry 60(1):41–44, 1999 10074877
Howard R, Castle D, Wessely S, et al: A comparative study of 470 cases of
early-onset and late-onset schizophrenia. Br J Psychiatry 163:352–357,
1993 8401965
Howard R, Almeida O, Levy R: Phenomenology, demography and
diagnosis in late paraphrenia. Psychol Med 24(2):397–410, 1994
8084935
Howard RJ, Graham C, Sham P, et al: A controlled family study of late-
onset non-affective psychosis (late paraphrenia). Br J Psychiatry
170:511–514, 1997 9330015
Howard R, Rabins PV, Seeman MV, et al: Late-onset schizophrenia and
very-late-onset schizophrenia-like psychosis: an international
consensus. Am J Psychiatry 157(2):172–178, 2000 10671383
Huber G: The heterogeneous course of schizophrenia. Schizophr Res 28(2-
3):177–185, 1997 9468352
Jeste DV, Finkel SI: Psychosis of Alzheimer’s disease and related
dementias. Diagnostic criteria for a distinct syndrome. Am J Geriatr
Psychiatry 8(1):29–34, 2000 10648292
Jeste DV, Wragg RE, Salmon DP, et al: Cognitive deficits of patients with
Alzheimer’s disease with and without delusions. Am J Psychiatry
149(2):184–189, 1992 1734737
Jeste DV, Harris MJ, Krull A, et al: Clinical and neuropsychological
characteristics of patients with late-onset schizophrenia. Am J
Psychiatry 152(5):722–730, 1995 7726312
Jeste DV, Klausner M, Brecher M, et al: A clinical evaluation of risperidone
in the treatment of schizophrenia: a 10-week, open-label, multicenter
trial. ARCS Study Group. Assessment of Risperdal in a Clinical
Setting. Psychopharmacology (Berl) 131:239–247, 1997a 9203234
Jeste DV, Symonds LL, Harris MJ, et al: Nondementia nonpraecox
dementia praecox? Late-onset schizophrenia. Am J Geriatr Psychiatry
5(4):302–317, 1997b 9363287
Jeste DV, Lacro JP, Bailey A, et al: Lower incidence of tardive dyskinesia
with risperidone compared with haloperidol in older patients. J Am
Geriatr Soc 47(6):716–719, 1999a 10366172
Jeste DV, Rockwell E, Harris MJ, et al: Conventional vs. newer
antipsychotics in elderly patients. Am J Geriatr Psychiatry 7(1):70–76,
1999b 9919323
Jeste DV, Barak Y, Madhusoodanan S, et al: International multisite double-
blind trial of the atypical antipsychotics risperidone and olanzapine in
175 elderly patients with chronic schizophrenia. Am J Geriatr
Psychiatry 11(6):638–647, 2003 14609804
Jeste DV, Blazer DG, First M: Aging-related diagnostic variations: need for
diagnostic criteria appropriate for elderly psychiatric patients. Biol
Psychiatry 58(4):265–271, 2005 16102544
Jeste DV, Meeks TW, Kim DS, et al: Diagnostic categories and criteria for
neuropsychiatric syndromes in dementia: research agenda for DSM-V,
in Diagnostic Issues in Dementia: Advancing the Research Agenda for
DSM-V. Edited by Sunderlan T, Jeste DV, Baiyewu O, et al.
Washington, DC, American Psychiatric Publishing, 2007, pp 77–98
Jeste DV, Blazer D, Casey D, et al: ACNP White Paper: update on use of
antipsychotic drugs in elderly persons with dementia.
Neuropsychopharmacology 33(5):957–970, 2008 17637610
Jin H, Meyer JM, Jeste DV: Phenomenology of and risk factors for new-
onset diabetes mellitus and diabetic ketoacidosis associated with
atypical antipsychotics: an analysis of 45 published cases. Ann Clin
Psychiatry 14(1):59–64, 2002 12046641
Jin H, Meyer JM, Jeste DV: Atypical antipsychotics and glucose
dysregulation: a systematic review. Schizophr Res 71(2-3):195–212,
2004 15474892
Jin H, Shih PA, Golshan S, et al.: Comparison of longer-term safety and
effectiveness of 4 atypical antipsychotics in patients over age 40: a trial
using equipoise-stratified randomization. J Clin Psychiatry 74(1):10–
18, 2012 23218100
Jones DK, Catani M, Pierpaoli C, et al: A diffusion tensor magnetic
resonance imaging study of frontal cortex connections in very-late-
onset schizophrenia-like psychosis. Am J Geriatr Psychiatry 13(12):
1092–1099, 2005 16319302
Jones PB, Barnes TR, Davies L, et al: Randomized controlled trial of the
effect on Quality of Life of second- vs first-generation antipsychotic
drugs in schizophrenia: Cost Utility of the Latest Antipsychotic Drugs
in Schizophrenia Study (CUtLASS 1). Arch Gen Psychiatry
63(10):1079–1087, 2006 17015810
Joukamaa M, Heliövaara M, Knekt P, et al: Mental disorders and cause-
specific mortality. Br J Psychiatry 179:498–502, 2001 11731351
Kammoun S, Gold G, Bouras C, et al: Immediate causes of death of
demented and non-demented elderly. Acta Neurol Scand Suppl 176:96–
99, 2000 11261812
Kane JM, Woerner MG, Pollack S, et al: Does clozapine cause tardive
dyskinesia? J Clin Psychiatry 54(9):327–330, 1993 8104929
Keene J, Hope T, Fairburn CG, Jacoby R: Death and dementia. Int J Geriatr
Psychiatry 16(10):969–974, 2001 11607941
Keith SJ, Regier DA, Rae DS: Schizophrenic disorders, in Psychiatric
Disorders in America: the Epidemiologic Catchment Area Study.
Edited by Robins LN, Regier DA. New York, Free Press, 1991, pp 33–
52
Kendler KS, Davis KL: The genetics and biochemistry of paranoid
schizophrenia and other paranoid psychoses. Schizophr Bull 7(4):689–
709, 1981 7034192
Kennedy JS, Jeste D, Kaiser CJ, et al: Olanzapine vs haloperidol in geriatric
schizophrenia: analysis of data from a double-blind controlled trial. Int
J Geriatr Psychiatry 18(11):1013–1020, 2003 14618553
Kindermann SS, Dolder CR, Bailey A, et al: Pharmacological treatment of
psychosis and agitation in elderly patients with dementia: four decades
of experience. Drugs Aging 19(4):257–276, 2002 12038878
Kinon BJ, Jeste DV, Kollack-Walker S, et al: Olanzapine treatment for
tardive dyskinesia in schizophrenia patients: a prospective clinical trial
with patients randomized to blinded dose reduction periods. Prog
Neuropsychopharmacol Biol Psychiatry 28(6):985–996, 2004
15380859
Kraepelin E: Dementia Praecox and Paraphrenia (1919). Translated by
Barclay RM. Huntington, NY, Krieger, 1971
Kulkarni J, de Castella A, Smith D, et al: A clinical trial of the effects of
estrogen in acutely psychotic women. Schizophr Res 20(3):247–252,
1996 8827850
Kulkarni J, Riedel A, de Castella AR, et al: Estrogen—a potential treatment
for schizophrenia. Schizophr Res 48(1):137–144, 2001 11278160
Lehman AF, Lieberman JA, Dixon LB, et al: Practice guideline for the
treatment of patients with schizophrenia, second edition. Am J
Psychiatry 161 (2 suppl):1–56, 2004 15000267
Lieberman JA, Saltz BL, Johns CA, et al: The effects of clozapine on
tardive dyskinesia. Br J Psychiatry 158:503–510, 1991 1675900
Lieberman JA, Stroup TS, McEvoy JP, et al: Effectiveness of antipsychotic
drugs in patients with chronic schizophrenia. N Engl J Med
353(12):1209–1223, 2005 16172203
Lindamer LA, Buse DC, Lohr JB, et al: Hormone replacement therapy in
postmenopausal women with schizophrenia: positive effect on negative
symptoms? Biol Psychiatry 49:47–51, 2001 11163779
Littrell KH, Johnson CG, Littrell S, Peabody CD: Marked reduction of
tardive dyskinesia with olanzapine. Arch Gen Psychiatry 55(3):279–
280, 1998 9510225
Livingston G, Johnston K, Katona C, et al: Systematic review of
psychological approaches to the management of neuropsychiatric
symptoms of dementia. Am J Psychiatry 162(11):1996–2021, 2005
16263837
Lohr JB, Alder M, Flynn K, et al: Minor physical anomalies in older
patients with late-onset schizophrenia, early-onset schizophrenia,
depression, and Alzheimer’s disease. Am J Geriatr Psychiatry
5(4):318–323, 1997 9363288
Maher B: Delusional thinking and cognitive disorder. Integr Physiol Behav
Sci 40(3):136–146, 2005 17477206
Masand PS: Atypical antipsychotics for elderly patients with
neurodegenerative disorders and medical conditions. Psychiatr Ann
30:203–208, 2000
Mazeh D, Zemishlani C, Aizenberg D, Barak Y: Patients with very-late-
onset schizophrenia-like psychosis: a follow-up study. Am J Geriatr
Psychiatry 13(5):417–419, 2005 15879591
McKeith I, Del Ser T, Spano P, et al: Efficacy of rivastigmine in dementia
with Lewy bodies: a randomised, double-blind, placebo-controlled
international study. Lancet 356(9247):2031–2036, 2000 11145488
McQuaid JR, Granholm E, McClure FS, et al: Development of an
integrated cognitive-behavioral and social skills training intervention
for older patients with schizophrenia. J Psychother Pract Res 9(3):149–
156, 2000 10896740
Meeks TW, Jeste DV: Beyond the black box: What is the role for
antipsychotics in dementia? Curr Psychiatr 7(6):50–65, 2008 19221607
Mitter P, Reeves S, Romero-Rubiales F, et al: Migrant status, age, gender
and social isolation in very late-onset schizophrenia-like psychosis. Int
J Geriatr Psychiatry 20(11):1046–1051, 2005 16250076
Miyasaki JM, Shannon K, Voon V, et al: Practice Parameter: evaluation and
treatment of depression, psychosis, and dementia in Parkinson disease
(an evidence-based review): report of the Quality Standards
Subcommittee of the American Academy of Neurology. Neurology
66(7):996–1002, 2006 16606910
Montross L, Kasckow J, Golshan S, et al: The continued need for suicide
assessments among middle-aged and older patients with schizophrenia.
Paper presented at the annual meeting of the American Association for
Geriatric Psychiatry, San Juan, Puerto Rico, March 2006
Moore NC: Is paranoid illness associated with sensory defects in the
elderly? J Psychosom Res 25(2):69–74, 1981 7277274
Moore R, Blackwood N, Corcoran R, et al: Misunderstanding the intentions
of others: an exploratory study of the cognitive etiology of persecutory
delusions in very late-onset schizophrenia-like psychosis. Am J Geriatr
Psychiatry 14(5):410–418, 2006 16670245
Mueser KT, Pratt SI, Bartels SJ, et al: Randomized trial of social
rehabilitation and integrated health care for older people with severe
mental illness. J Consult Clin Psychol 78(4):561–573, 2010 20658812
Mukaetova-Ladinska EB, Harrington CR, Xuereb J, et al: Biochemical,
neuropathological, and clinical correlations of neurofibrillary
degeneration in Alzheimer’s disease, in Treating Alzheimer’s and
Other Dementias. Edited by Bergener M, Finkel SI. New York,
Springer, 1995, pp 57–80
Naimark D, Jackson E, Rockwell E, et al: Psychotic symptoms in
Parkinson’s disease patients with dementia. J Am Geriatr Soc
44(3):296–299, 1996 8600200
Ostling S, Skoog I: Psychotic symptoms and paranoid ideation in a
nondemented population-based sample of the very old. Arch Gen
Psychiatry 59(1):53–59, 2002 11779282
Palmer BW, McClure FS, Jeste DV: Schizophrenia in late life: findings
challenge traditional concepts. Harv Rev Psychiatry 9(2):51–58, 2001
11266402
Palmer BW, Nayak GV, Jeste DV: A comparison of early- and late-onset
schizophrenia, in Schizophrenia Into Later Life: Treatment, Research,
and Policy. Edited by Cohen CI. Washington, DC, American
Psychiatric Publishing, 2002, pp 43–55
Palmer BW, Bondi MW, Twamley EW, et al: Are late-onset schizophrenia
spectrum disorders neurodegenerative conditions? Annual rates of
change on two dementia measures. J Neuropsychiatry Clin Neurosci
15(1):45–52, 2003 12556570
Parkinson Study Group: Low-dose clozapine for the treatment of drug-
induced psychosis in Parkinson’s disease. N Engl J Med 340(10):757–
763, 1999 10072410
Patterson TL, Bucardo J, McKibbin CL, et al: Development and pilot
testing of a new psychosocial intervention for older Latinos with
chronic psychosis. Schizophr Bull 31(4):922–930, 2005 16037481
Patterson TL, Mausbach BT, McKibbin C, et al: Functional adaptation
skills training (FAST): a randomized trial of a psychosocial
intervention for middle-aged and older patients with chronic psychotic
disorders. Schizophr Res 86(1–3):291–299, 2006 16814526
Paulsen JS, Salmon DP, Thal LJ, et al: Incidence of and risk factors for
hallucinations and delusions in patients with probable AD. Neurology
54(10):1965–1971, 2000 10822438
Pollak P, Tison F, Rascol O, et al: Clozapine in drug induced psychosis in
Parkinson’s disease: a randomised, placebo controlled study with open
follow up. J Neurol Neurosurg Psychiatry 75(5):689–695, 2004
15090561
Prager S, Jeste DV: Sensory impairment in late-life schizophrenia.
Schizophr Bull 19(4):755–772, 1993 8303225
Putnam KM, Harvey PD: Cognitive impairment and enduring negative
symptoms: a comparative study of geriatric and nongeriatric
schizophrenia patients. Schizophr Bull 26(4):867–878, 2000 11087019
Raghuram R, Keshavan MD, Channabasavanna SM: Musical hallucinations
in a deaf middle-aged patient. J Clin Psychiatry 41(10):357, 1980
7430079
Ram R, Bromet EJ, Eaton WW, et al: The natural course of schizophrenia: a
review of first-admission studies. Schizophr Bull 18(2):185–207, 1992
1621068
Riedel M, Eich FX, Möller HJ: A pilot study of the safety and efficacy of
amisulpride and risperidone in elderly psychotic patients. Eur
Psychiatry 24(3):149–153, 2009 19070995
Rivkin P, Kraut M, Barta P, et al: White matter hyperintensity volume in
late-onset and early-onset schizophrenia. Int J Geriatr Psychiatry
15(12):1085–1089, 2000 11180463
Rockwell E, Krull AJ, Dimsdale J, Jeste DV: Late-onset psychosis with
somatic delusions. Psychosomatics 35(1):66–72, 1994 8134531
Ropacki SA, Jeste DV: Epidemiology of and risk factors for psychosis of
Alzheimer’s disease: a review of 55 studies published from 1990 to
2003. Am J Psychiatry 162(11):2022–2030, 2005 16263838
Roth M: The natural history of mental disorder in old age. J Ment Sci
101(423):281–301, 1955 13243044
Sachdev P, Brodaty H, Rose N, Cathcart S: Schizophrenia with onset after
age 50 years, 2: neurological, neuropsychological and MRI
investigation. Br J Psychiatry 175:416–421, 1999 10789271
Schneider LS: Pharmacologic management of psychosis in dementia. J Clin
Psychiatry 60(suppl 8):54–60, 1999 10335671
Schneider LS, Pollock VE, Lyness SA: A metaanalysis of controlled trials
of neuroleptic treatment in dementia. J Am Geriatr Soc 38(5):553–563,
1990 1970586
Schneider LS, Dagerman KS, Insel P: Risk of death with atypical
antipsychotic drug treatment for dementia: meta-analysis of
randomized placebo-controlled trials. JAMA 294(15):1934–1943, 2005
16234500
Schneider LS, Dagerman K, Insel PS: Efficacy and adverse effects of
atypical antipsychotics for dementia: meta-analysis of randomized,
placebo-controlled trials. Am J Geriatr Psychiatry 14(3):191–210,
2006a 16505124
Schneider LS, Tariot PN, Dagerman KS, et al: Effectiveness of atypical
antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med
355(15):1525–1538, 2006b 17035647
Seeman MV: The role of estrogen in schizophrenia. J Psychiatry Neurosci
21(2):123–127, 1996 8820178
Simpson GM, Lee JH, Shrivastava RK: Clozapine in tardive dyskinesia.
Psychopharmacology (Berl) 56(1):75–80, 1978 415329
Sink KM, Holden KF, Yaffe K: Pharmacological treatment of
neuropsychiatric symptoms of dementia: a review of the evidence.
JAMA 293(5):596–608, 2005 15687315
Small JG, Milstein V, Marhenke JD, et al: Treatment outcome with
clozapine in tardive dyskinesia, neuroleptic sensitivity, and treatment-
resistant psychosis. J Clin Psychiatry 48(7):263–267, 1987 2885310
Srikanth S, Nagaraja AV, Ratnavalli E: Neuropsychiatric symptoms in
dementia-frequency, relationship to dementia severity and comparison
in Alzheimer’s disease, vascular dementia and frontotemporal
dementia. J Neurol Sci 236(1–2):43–48, 2005 15964021
Stern Y, Albert M, Brandt J, et al: Utility of extrapyramidal signs and
psychosis as predictors of cognitive and functional decline, nursing
home admission, and death in Alzheimer’s disease: prospective
analyses from the Predictors Study. Neurology 44(12):2300–2307,
1994 7991116
Stoppe G, Brandt CA, Staedt JH: Behavioural problems associated with
dementia: the role of newer antipsychotics. Drugs Aging 14(1):41–54,
1999 10069407
Street JS, Tollefson GD, Tohen M, et al: Olanzapine for psychotic
conditions in the elderly. Psychiatr Ann 30:191–196, 2000
Suh GH, Son HG, Ju YS, et al: A randomized, double-blind, crossover
comparison of risperidone and haloperidol in Korean dementia patients
with behavioral disturbances. Am J Geriatr Psychiatry 12(5): 509–516,
2004 15353389
Sultzer DL: Neuroimaging and the origin of psychiatric symptoms in
dementia. Int Psychogeriatr 8 (suppl 3):239–243, discussion 269–272,
1996 9154570
Suzuki T, Remington G, Uchida H, et al: Management of schizophrenia in
late life with antipsychotic medications: a qualitative review. Drugs
Aging 28(12):961–980, 2011 22117095
Tariot PN, Schneider L, Katz IR, et al: Quetiapine treatment of psychosis
associated with dementia: a double-blind, randomized, placebo-
controlled clinical trial. Am J Geriatr Psychiatry 14(9):767–776, 2006
16905684
Twamley EW, Padin DS, Bayne KS, et al: Work rehabilitation for middle-
aged and older people with schizophrenia: a comparison of three
approaches. J Nerv Ment Dis 193(9):596–601, 2005 16131942
Tzimos A, Samokhvalov V, Kramer M, et al: Safety and tolerability of oral
paliperidone extended-release tablets in elderly patients with
schizophrenia: a double-blind, placebo-controlled study with six-month
open-label extension. Am J Geriatr Psychiatry 16(1):31–43, 2008
18165460
Vahia I, Bankole AO, Reyes P, et al: Schizophrenia in later life. Aging
Health 3:383–396, 2007
Vahia IV, Palmer BW, Depp C, et al: Is late-onset schizophrenia a subtype
of schizophrenia? Acta Psychiatr Scand 122(5): 414–426, 2010
20199491
Weiss E, Hummer M, Koller D, et al: Off-label use of antipsychotic drugs. J
Clin Psychopharmacol 20(6):695–698, 2000 11106144
Wilkosz PA, Miyahara S, Lopez OL, et al: Prediction of psychosis onset in
Alzheimer disease: the role of cognitive impairment, depressive
symptoms, and further evidence for psychosis subtypes. Am J Geriatr
Psychiatry 14(4):352–360, 2006 16582044
Wirshing DA, Spellberg BJ, Erhart SM, et al: Novel antipsychotics and new
onset diabetes. Biol Psychiatry 44(8):778–783, 1998 9798083
Yassa R, Jeste DV: Gender differences in tardive dyskinesia: a critical
review of the literature. Schizophr Bull 18(4):701–715, 1992 1359633
Yeung PP, Tariot PN, Schneider LS, et al: Quetiapine for elderly patients
with psychotic disorders. Psychiatr Ann 30:197–201, 2000
Zisook S, Montross L, Kasckow J, et al: Subsyndromal depressive
symptoms in middle-aged and older persons with schizophrenia. Am J
Geriatr Psychiatry 15(12):1005–1014, 2007 18056819
Zubenko GS, Moossy J, Martinez AJ, et al: Neuropathologic and
neurochemical correlates of psychosis in primary dementia. Arch
Neurol 48(6):619–624, 1991 1710105
Suggested Readings
Auslander LA, Jeste DV: Sustained remission of schizophrenia among
community-dwelling older outpatients. Am J Psychiatry 161:1490–
1493, 2004
Chou KL, Borek LL, Friedman JH: The management of psychosis in
movement disorder patients. Expert Opin Pharmacother 8:935–943,
2007
Granholm E, McQuaid JR, McClure FS, et al: A randomized, controlled
trial of cognitive behavioral social skills training for middle-aged and
older outpatients with chronic schizophrenia. Am J Psychiatry
162:520–529, 2005
Howard R, Rabins PV, Seeman MV, et al: Late-onset schizophrenia and
very-late-onset schizophrenia-like psychosis: an international
consensus. Am J Psychiatry 157:172–178, 2000
Jeste DV, Harris MJ, Krull A, et al: Clinical and neuropsychological
characteristics of patients with late-onset schizophrenia. Am J
Psychiatry 152:722–730, 1995
Jeste DV, Blazer D, Casey D, et al: ACNP White Paper: update on the use
of antipsychotic drugs in elderly persons with dementia.
Neuropsychopharmacology 33:957–970, 2008
Kendler KS, Tsuang MT, Hays P: Age at onset in schizophrenia: a familial
perspective. Arch Gen Psychiatry 44:881–890, 1987
Ropacki SA, Jeste DV: Epidemiology of and risk factors for psychosis of
Alzheimer’s disease: a review of 55 studies published from 1990 to
2003. Am J Psychiatry 162(11):2022–2030, 2005
Vahia I, Bankole AO, Reyes P, et al: Schizophrenia in later life. Aging
Health 3:383–396, 2007
CHAPTER 12
Epidemiology
Prevalence estimates of late-life anxiety disorders vary widely, ranging from the view
that they are relatively uncommon and, if present, are typically comorbid conditions
stemming from depression and/or cognitive decline to the opinion that anxiety disorders
are common and in fact the most prevalent disorders in older adults. Such disparate
opinions may stem from the wide variability in published prevalence estimates of
anxiety disorders and related conditions in community-dwelling older adults (Table 12–
3). For example, the National Comorbidity Survey Replication (NCS-R) found much
lower rates of all anxiety disorders in individuals ages 65 years and older compared
with their peak prevalence in individuals ages 18–44 years (Gum et al. 2009). In
contrast, the Longitudinal Aging Study Amsterdam (LASA; Beekman et al. 1998), one
of the largest epidemiological studies to focus on psychiatric disorders in elderly
persons, demonstrated that more than 10% of elderly adults had any anxiety disorder,
including generalized anxiety disorder (GAD), any phobic disorder, panic disorder,
and/or OCD, closer to estimates in younger adults. Other studies have found that all of
the anxiety disorders are less common in older adults, with many studies finding panic
disorder rare or nonexistent (Flint 1994). One study, the Australian National Mental
Health and Well-Being Survey (NMHWS), found that the prevalence of anxiety
disorders in elderly people varied almost threefold, depending on whether DSM-IV-TR
(American Psychiatric Association 2000) or ICD-10 (World Health Organization 1992)
criteria were used (Trollor et al. 2007).
As a whole, these epidemiological studies suggest that GAD is about as common in
older adults as in younger adults, whereas other anxiety disorders (as well as OCD and
possibly PTSD) are rarer in older adults. However, we view the prevalence estimates as
being limited by the well-known challenges in identifying anxiety disorders in this age
group (Wolitzky-Taylor et al. 2010). Chief among these challenges is that older adults
are less able as a group than younger adults to accurately identify anxiety symptoms
(Wetherell et al. 2009). Furthermore, avoidance and the “excessive” nature of anxiety,
which are two distinguishing features in DSM-IV (American Psychiatric Association
1994), are quite challenging to detect in older people, as described in Table 12–2.
Additionally, anxiety conditions germane to elderly persons—such as anxiety in the
context of dementia (Starkstein et al. 2007), anxiogenic medical conditions such as
heart disease (Todaro et al. 2007), fear of falling (Gagnon et al. 2005), and hoarding
syndrome (Saxena 2007)—may not be discerned by standard epidemiological
assessments or methodology. Thus, the prevalence and significance of such conditions
are currently unknown.
TABLE 12–1. DSM-5 changes of most relevance to older adults: anxiety, obsessive-
compulsive, and trauma-related disorders
Disorder Changes in DSM-5
Anxiety disorders
Specific phobia “Excessive or unreasonable” requirement has been removed
in favor of “out of proportion to the actual danger.”
Social anxiety disorder “Excessive or unreasonable” criterion has been removed in
(social phobia) favor of “out of proportion to the actual threat”; fear of
negative evaluation has been added.
Panic disorder, Panic disorder and agoraphobia are now diagnosed
agoraphobia separately. Panic disorder diagnosis is unchanged.
Agoraphobic diagnosis is more specific about feared
situations (public transportation, open spaces, enclosed
places, standing in line or being in a crowd, and being
outside home alone) and requires fear/anxiety “out of
proportion to the actual danger.”
Generalized anxiety “During depression” exclusion has been removed.
disorder
Obsessive-compulsive and related disorders
Obsessive-compulsive Disorder has been reclassified into obsessive-compulsive
disorder and related disorders. “Recognized as excessive or
unreasonable” requirement has been removed.
Hoarding disorder Disorder has been added in DSM-5, in obsessive-
compulsive and related disorders.
Trauma- and stressor-related disorders
Posttraumatic stress Disorder has been reclassified into trauma- and stressor-
disorder related disorders. Changes include greater specificity
regarding the nature of the traumatic experience (see
Table 12–12, criterion A). Diagnosis requires symptoms
of intrusion, avoidance, negative alterations in cognitions
or mood, and heightened arousal/reactivity. “Sense of
foreshortened future” symptom has been removed.
Age at Onset and Risk Factors for Anxiety Disorders and
Related Conditions
Published reviews of the literature have taken two different stances on the question of
age at onset of anxiety disorders. One camp describes them as rarely of late onset (Flint
2005a), pointing to studies indicating onset in childhood or early adulthood (Kessler et
al. 2007). Another camp has found in clinical samples of elderly persons that many
individuals have a later onset of anxiety disorder (Lenze et al. 2005a; Le Roux et al.
2005; Sheikh et al. 2004; van Zelst et al. 2003). For example, in a sample of 103
treatment-seeking elderly patients with GAD, the mean age at onset was 49 years, with
46% of cases being of late onset (Lenze et al. 2005a). Other evidence suggests a
bimodal distribution of onset, with approximately two-thirds of patients having onset in
childhood or adolescence and one-third developing the disorder for the first time at age
50 years or later (Blazer et al. 1991; Le Roux et al. 2005). The largest epidemiological
study to specifically examine the question of incident mental disorders in elderly found
that late-onset anxiety disorders are not rare (Chou et al. 2011).
In a sense, both camps may be correct: aging may either protect from anxiety or
cause anxiety, depending on circumstances. Congruent with this assertion, some reports
have noted a decline in the propensity for negative affect (which underlies anxiety as
well as depression) from adulthood to “early” elderly years, with a subsequent rise in
the mid-70s (Charles et al. 2001; Teachman 2006). It has been suggested that aging
provides the opportunity to inoculate against the anxiety-producing nature of stressors
and to practice emotion regulation (Jarvik and Russell 1979). Additionally, aging may
reduce the propensity for anxiety because of degeneration in anxiety-producing brain
regions such as the locus coeruleus (Flint 1994). On the other hand, aging is associated
with a number of new stressors, such as chronic illness and disability (in self or loved
ones), that may be particularly anxiogenic. It is perhaps for this reason that GAD,
centrally defined by worry-related distress, seems to peak in prevalence in middle age
and frequently have a late age at onset. Also, age-related degeneration in brain regions
associated with adaptive responses to anxiety (e.g., the dorsolateral prefrontal cortex)
may reduce the ability of some to manage anxiogenic situations; findings of poststroke
GAD (Aström 1996) and cerebral lesions associated with late-onset OCD (Swoboda
and Jenike 1995) are congruent with this assertion. Along these lines, clinicians should
be aware that many prescription medications can induce anxiety (e.g., anticholinergics,
psychostimulants, steroids), and this possibility should be a part of any differential
diagnosis of new-onset anxiety in an elderly person. However, it is our clinical
experience that elderly persons, unless cognitively impaired, are usually quite insightful
when a medication they take causes clinically significant anxiety.
Another consideration is that some anxiety disorders presenting in later life may
have a different nature (Flint 2005b), owing to a potentially different etiology (much as
late-onset depression, often in the context of cerebrovascular disease, has different
phenomenology than the early-onset variety). Research has generally not found that age
at onset affects qualitative presentation of anxiety disorders (Sheikh et al. 2004;
Wetherell et al. 2003b), yet this in itself may be due to the use of DSM diagnoses
created for younger adults. In summary, late-onset anxiety disorders are probably more
common than appreciated (Chou et al. 2011), but the disorders may present differently
than in younger adults.
Risk factors for anxiety disorders have been reported in elderly persons (Vink et al.
2008). They include female gender, neurotic personality, early life stress, and chronic
disability in self or spouse. However, research uncovering neurobiological risk factors
for late-life, or late-onset, anxiety has not been done. Therefore, the basic question of
who develops anxiety in older age is largely unanswered.
TABLE 12–4. Shared and distinct clinical features of anxiety disorders and related
conditions
Situational Situational Autonomic Anticipatory Obsessions Panic
fear avoidance arousal worry and attacks
compulsions
Specific X X X X X
phobia,
social
anxiety
disorder
(social
phobia), and
agoraphobia
Panic disorder X X X X X
Generalized X
anxiety
disorder
Obsessive- X X X X
compulsive
disorder
Hoarding X
disorder
Posttraumatic X X X X
stress
disorder
Phobias
Phobias are marked by an excessive and unreasonable situational fear that is distressing
or impairing. Exposure to the feared situation is either avoided or endured only with
distress, even precipitating a panic attack in some instances.
Phobia may be difficult to detect in an older adult—particularly if the phobia has
been lifelong—because of lifestyle accommodation around it (e.g., avoiding social
situations in the case of social phobia). The available evidence suggests that specific
phobias (Table 12–5) are quite common in elderly persons, whereas social phobia is less
so.
The difficulty of diagnosing phobias is exemplified by fear of falling, a common
problem in elderly persons, which by its very name might be considered a phobia.
However, under DSM-IV criteria, this condition usually could not be diagnosable as a
phobia, in part because individuals who fear falling typically do not feel their fear to be
excessive or unreasonable (Gagnon et al. 2005). Under DSM-5 criteria, fear that is “out
of proportion to actual danger posed,” such as severe levels of fear and avoidance in
individuals who are at low objective fall risk, could be diagnosed as a phobia.
Social phobia (Table 12–6) has received little study in elderly persons. In the largest
examination of this condition, Cairney et al. (2007) found that aging was associated
with reduced prevalence of social phobia, although the condition still remained fairly
common in old age and qualitatively appeared strikingly similar to that in young adults.
Agoraphobia (Table 12–7) can present within the context of panic attacks but
frequently does not do so in elderly persons (McCabe et al. 2006). It can appear de novo
in the context of a stroke or other medical event, in which case it can be highly
disabling, leading to inhibition of activities necessary for restoration after the event
(Burvill et al. 1995). The condition itself might lead to decreased ability to detect it;
accordingly, studies of adults who are homebound or receiving home-based aging
services have found high rates of anxiety symptoms (Gum et al. 2009; Jayasinghe et al.
2013).
Panic Disorder
Case Example
A 72-year-old woman was referred to geriatric psychiatry after she presented with complaints of “passing out” to
her primary care doctor. She described how, without warning, she would feel like she was losing consciousness.
The feeling was subjective and not tied to exertion, physical activity, or orthostatic changes. On further interview,
she reported that these episodes would last for approximately 15 minutes and would be associated with shortness
of breath, palpitations, tingling in her hands, and abdominal distress. She did not have any active cardiac
problems. These episodes occurred approximately monthly.
Since the onset of these symptoms, the woman’s family describes her as more “needy.” She unnecessarily
pays lots of attention to and closely monitors interoceptive sensations. She leaves her home less often, and when
she does, it is only with a family member present. She describes this change as related to crime in the
neighborhood and realistic worries about falling without someone to help her in case of an injury.
TABLE 12–6. DSM-5 diagnostic criteria for social anxiety disorder (social phobia)
A. Marked fear or anxiety about one or more social situations in which the individual is
exposed to possible scrutiny by others. Examples include social interactions (e.g.,
having a conversation, meeting unfamiliar people), being observed (e.g., eating or
drinking), and performing in front of others (e.g., giving a speech).
Note: In children, the anxiety must occur in peer settings and not just during
interactions with adults.
B. The individual fears that he or she will act in a way or show anxiety symptoms that
will be negatively evaluated (i.e., will be humiliating or embarrassing; will lead to
rejection or offend others).
C. The social situations almost always provoke fear or anxiety.
Note: In children, the fear or anxiety may be expressed by crying, tantrums,
freezing, clinging, shrinking, or failing to speak in social situations.
D. The social situations are avoided or endured with intense fear or anxiety.
E. The fear or anxiety is out of proportion to the actual threat posed by the social
situation and to the sociocultural context.
F. The fear, anxiety, or avoidance is persistent, typically lasting for 6 months or more.
G. The fear, anxiety, or avoidance causes clinically significant distress or impairment in
social, occupational, or other important areas of functioning.
H. The fear, anxiety, or avoidance is not attributable to the physiological effects of a
substance (e.g., a drug of abuse, a medication) or another medical condition.
I. The fear, anxiety, or avoidance is not better explained by the symptoms of another
mental disorder, such as panic disorder, body dysmorphic disorder, or autism
spectrum disorder.
J. If another medical condition (e.g., Parkinson’s disease, obesity, disfigurement from
burns or injury) is present, the fear, anxiety, or avoidance is clearly unrelated or is
excessive.
Specify if:
Performance only
Note. Criteria set above contains only the diagnostic criteria and specifiers; refer to DSM-5 for the full criteria set,
including specifier descriptions and coding and reporting procedures.
Source. DSM-5 criteria for social anxiety disorder (social phobia) reprinted from Diagnostic and Statistical Manual
of Mental Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, pp. 202–203. Used with
permission. Copyright © 2013 American Psychiatric Association.
Panic disorder (Table 12–8) is the syndrome of recurrent panic attacks (at least some
of which are unexpected) associated with concern about having further attacks and,
often, avoidance of situations associated in the patient’s mind with attacks. This
avoidance can often lead to agoraphobia.
Most studies have found a low prevalence of panic disorder in older adults in the
community. This has led to a line of thinking that perhaps the aging process reduces
panic disorder prevalence or that mortality due to panic disorder reduces the likelihood
of its being present in older age groups (Flint 1994). Late-onset panic disorder appears
to be rare and may often be a prodrome of a medical or neurological problem. However,
this assertion is based on the disorder as defined in young adults and cannot exclude the
possibility that panic symptoms that do not meet criteria for panic disorder may be more
common in late life. Even this assertion has been questioned by epidemiological data
suggesting that incident panic disorder in late life, although not common, is not rare
(Chou et al. 2011).
When panic disorder does occur in elderly persons, it tends to be associated with less
severe or less frequent panic attacks than in young adults, who frequently have daily
attacks (Sheikh et al. 2004). The lower severity of attacks may be because older adults
have less ability to mount a strong autonomic response (Flint et al. 2002). Nevertheless,
because the disability due to panic disorder tends to be related to the behavioral
avoidance rather than the frequency of attacks themselves, panic disorder can still be a
highly disabling condition in elderly persons.
GAD is a common and impairing disorder in older adults; it is highly comorbid with
personality, mood, and other anxiety disorders (Mackenzie et al. 2011). The central
feature of GAD is worry or anticipation of future negative events. Also known as
anxious apprehension, worry is thought to be a separate form of anxiety from fear or
anxious arousal, as seen in the fear disorders (e.g., panic disorder). Phenomenologically,
they are quite different; worry may wax and wane but does not have the intense
episodic nature of panic attacks (although worry can itself precipitate panic attacks).
GAD has evolved diagnostically to be considered first a residual state of phobic/panic
anxiety, then a disorder in its own right, and now, in the DSM-5 era, as still an anxiety
disorder but a close relative to major depressive disorder (MDD). Worry in DSM-5–
diagnosed GAD must be excessive, be difficult to control, and cause distress (Table 12–
9). The actual content of worry in late-life GAD has been noted to be similar to that in
older adults without GAD—that is, concerns about health or disability, family
relationships, or finances (Diefenbach et al. 2001).
Associated features of GAD are muscle tension, restlessness (or feeling keyed up or
on edge), sleep difficulties, concentration problems (often thought by older adults to be
memory problems), fatigue, and irritability. GAD may wax and wane but is typically
chronic, unlike depressive episodes (Lenze et al. 2005a). Late-life GAD is associated
with impaired quality of life but low levels of professional help seeking in mental health
settings (Mackenzie et al. 2011). In our clinical experience, elderly persons with GAD
often present to primary care or specialty medical care with these associated symptoms,
which then become the targets of treatment (e.g., benzodiazepines for sleep, muscle
relaxants or even physical therapy referral for muscle tension, cholinesterase inhibitors
for cognitive complaints) rather than detection of the underlying anxiety syndrome
(Figure 12–1). As the figure depicts, this focus on treating symptoms may lead to
additional complications from the pharmacological or other management, in addition to
lack of treatment of the underlying disorder.
In any event, GAD is one of the most common mental disorders in elderly persons,
and as noted earlier in the section “Age at Onset and Risk Factors for Anxiety
Disorders,” late-onset GAD is not uncommon (Chou et al. 2011). The disorder is even
more common in medical settings, as might be expected for any affective or anxiety
disorder (Todaro et al. 2007; Tolin et al. 2005). These findings, together with data
showing that elderly persons with anxiety disorders rarely present to specialty mental
health clinics (Ettner and Hermann 1997), suggest that primary and specialty medical
care, not psychiatric settings, are the places to find late-life GAD that is not comorbid
with depression (with attendant problems from this, as shown in Figure 12–1).
Symptoms that are common to GAD and MDD include impaired sleep and
concentration, low energy, and irritability. Additionally, low mood is a frequent feature
of GAD, and excessive worry is frequently seen in MDD. The underlying genetic
predisposition to both disorders is thought to be similar in older and younger adults
(Kendler et al. 1992, 2007). As a whole, there are several reasons for high comorbidity
between GAD and MDD, including bidirectional risk and shared underlying
neurocircuitry (Andreescu et al. 2013).
As previously noted in this section, GAD tends to be chronic rather than episodic.
Studies have found that the mean length of GAD symptoms in elderly persons
presenting for treatment ranged from 20 (Lenze et al. 2005a) to more than 30 years
(Stanley et al. 2003a; Wetherell et al. 2003a), whereas 9 years is the average among
younger adults (Wang et al. 2005). This chronicity fits with evidence showing that GAD
is one of the least likely mental disorders in the anxiety-affective spectrum to remit
spontaneously over time (Bruce et al. 2005). Other reports have suggested that in
elderly persons, chronic GAD tends to develop into a somatization condition (Rubio
and López-Ibor 2007) or mixed GAD and MDD (Schoevers et al. 2003).
Hoarding Disorder
Hoarding can be a disorder in its own right (Table 12–11) or can be a symptom of OCD.
Hoarding disorder is a common, often highly impairing, and sometimes dangerous
condition in older adults. Hoarding behavior is increasingly common and severe in old
age (Teachman 2006). Because hoarding is commonly seen in association with
dementia, debilitating physical conditions, and comorbid mental illness (Ayers et al.
2014a; Snowdon et al. 2007), clinicians should be sensitive to detecting hoarding
behavior in these contexts. In patients with dementia, clinicians need to delineate
repetitive behaviors from hoarding. True late-onset hoarding disorder is unlikely, but
because the disorder is progressive and rarely remits with age, its prevalence is highest
in oldest age groups (Ayers et al. 2010). For example, 15% of nursing home residents
and 25% of senior day care attendees display clinically significant hoarding behaviors.
Hoarding disorder may be difficult to detect in many older adults, who may deny
“urges” to save but may describe their hoarding by other phrases (e.g., need to save,
cannot part with). Similarly, they may not describe their hoarding as a psychiatric
problem but rather as a consequence of early life events or learned behaviors. Older
adults are often unaware that this is a treatable psychiatric problem and have not sought
or received treatment (Ayers et al. 2014b). Accurate recognition of and treatment for
hoarding are of growing concern, because hoarding is now considered a form of elder
self-abuse in several U.S. states (Dong et al. 2012). Therefore, hoarding disorder merits
greater efforts by clinicians to diagnose and manage it and by researchers to develop
better treatments.
Note. Criteria set above contains only the diagnostic criteria and specifiers; refer to DSM-5 for the full criteria set,
including specifier descriptions and coding and reporting procedures.
Source. DSM-5 criteria for obsessive-compulsive disorder reprinted from Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, p. 237. Used with
permission. Copyright © 2013 American Psychiatric Association.
Case Example
A World War II veteran presented to the Veterans Affairs hospital for complaints related to a possible
diagnosis of PTSD. He said that during the invasion of Normandy in 1941, he was a land-sea transport
captain who delivered soldiers to Omaha Beach. Many of these soldiers, especially in the initial waves, were
killed by gunfire in front of him. He describes still visualizing bodies lapped against his ship by the waves.
He describes several symptoms related to this visualization: elevated anxiety and distress, nightmares, poor
concentration, irritability, and feelings of being detached or numb. These symptoms have worsened recently
as a number of his friends have passed away from illness.
PTSD is a chronic, often fluctuating disorder that adversely affects the quality of life
of older adults (Chopra et al. 2014) and may be a risk factor for dementia (Yaffe et al.
2010). PTSD is no longer categorized as an anxiety disorder in DSM-5; rather, it is now
one of the trauma- and stressor-related disorders. PTSD is characterized by exposure to
a traumatic event (an accident, violence, or natural event) (Table 12–12). The event
must have produced at least 1 month of symptoms in each of four clusters: 1) intrusion
(reexperiencing of the event via thoughts, flashbacks, or dreams), 2) avoidance
(avoiding thoughts of the event or situations that bring it to mind), 3) negative
alterations in cognitions or mood (feeling detached, having diminished interest in or
responsiveness to activities, being unable to experience positive emotions), and 4)
heightened arousal/reactivity (e.g., sleep disturbances, exaggerated startle, irritability).
If this syndrome is present but symptoms last less than 1 month, acute stress disorder is
diagnosed.
PTSD has received little study in older adults, despite the association of the disorder
with many types of traumatic experiences common in this age group, such as combat
experiences for military veterans (Frueh et al. 2007) or traumatic medical events (Dew
et al. 2001). In a study by Pietrzak et al. (2012a), the combined lifetime prevalence of
full or partial PTSD in older individuals was 10%. Also of note is an epidemiological
study that found that PTSD had the highest 12-month prevalence of any mental disorder
in older African American individuals (Ford et al. 2007). This finding may have
reflected methodological differences from other studies or real differences of minorities
from Caucasians in this age group. Overall, the research in late-life PTSD is far behind
its overall impact in terms of prevalence and morbidity (van Zelst et al. 2003).
In older adults, partial PTSD needs to be considered when diagnosing patients.
Although partial PTSD does not meet full PTSD criteria, it appears to be more than a
subsyndromal problem because it is associated with impaired psychosocial functioning
(Pietrzak et al. 2012).
Fear of Falling
A prime example of how DSM criteria have not mapped well onto geriatric syndromes
is fear of falling (e.g., Mohlman et al. 2012). Fear of falling is a common syndrome in
community-dwelling older adults; estimates are 7%–14%. Fear of falling consists of
moderate to severe fear with avoidance of multiple situations and activities (e.g.,
approximately 3% of older adults avoid leaving their houses or yards as a result of fear
of falling; Arfken et al. 1994).
The fear of falling is frequently excessive relative to objective fall risk. Many who
report fear have never experienced a fall and do not report any fall risk factors;
therefore, the diagnosis of agoraphobia could properly define many older adults with
fear of falling (McCabe et al. 2006; van Haastregt et al. 2008). The fear of falling is also
impairing, limiting mobility and contributing to functional decline and
institutionalization (Zijlstra et al. 2007). However, fear of falling does not map well
onto the pre–DSM-5 requirement of insight into excessiveness of the fear, which many
older patients lack. For example, Gagnon et al. (2005) found that only 1 of 48 subjects
with moderate or severe fear of falling considered their fear to be unreasonable, even
though the fear frequently resulted in the individuals’ avoidance of activities, in some
cases to the point of their being housebound (Flint 2005a). Fear of falling is common
and impairing and therefore hopefully will be encompassed more readily by the changes
with DSM-5, such as removal of the requirement of the patient’s insight into
“excessiveness” for many of the anxiety disorders.
A. Exposure to actual or threatened death, serious injury, or sexual violence in one (or
more) of the following ways:
1. Directly experiencing the traumatic event(s).
2. Witnessing, in person, the event(s) as it occurred to others.
3. Learning that the traumatic event(s) occurred to a close family member or close
friend. In cases of actual or threatened death of a family member or friend, the
event(s) must have been violent or accidental.
4. Experiencing repeated or extreme exposure to aversive details of the traumatic
event(s) (e.g., first responders collecting human remains; police officers repeatedly
exposed to details of child abuse).
Note: Criterion A4 does not apply to exposure through electronic media, television,
movies, or pictures, unless this exposure is work related.
B. Presence of one (or more) of the following intrusion symptoms associated with the
traumatic event(s), beginning after the traumatic event(s) occurred:
1. Recurrent, involuntary, and intrusive distressing memories of the traumatic
event(s).
Note: In children older than 6 years, repetitive play may occur in which themes or
aspects of the traumatic event(s) are expressed.
2. Recurrent distressing dreams in which the content and/or affect of the dream are
related to the traumatic event(s).
Note: In children, there may be frightening dreams without recognizable content.
3. Dissociative reactions (e.g., flashbacks) in which the individual feels or acts as if
the traumatic event(s) were recurring. (Such reactions may occur on a continuum,
with the most extreme expression being a complete loss of awareness of present
surroundings.)
Note: In children, trauma-specific reenactment may occur in play.
4. Intense or prolonged psychological distress at exposure to internal or external cues
that symbolize or resemble an aspect of the traumatic event(s).
5. Marked physiological reactions to internal or external cues that symbolize or
resemble an aspect of the traumatic event(s).
C. Persistent avoidance of stimuli associated with the traumatic event(s), beginning
after the traumatic event(s) occurred, as evidenced by one or both of the following:
1. Avoidance of or efforts to avoid distressing memories, thoughts, or feelings about
or closely associated with the traumatic event(s).
2. Avoidance of or efforts to avoid external reminders (people, places, conversations,
activities, objects, situations) that arouse distressing memories, thoughts, or
feelings about or closely associated with the traumatic event(s).
D. Negative alterations in cognitions and mood associated with the traumatic event(s),
beginning or worsening after the traumatic event(s) occurred, as evidenced by two
(or more) of the following:
1. Inability to remember an important aspect of the traumatic event(s) (typically due
to dissociative amnesia and not to other factors such as head injury, alcohol, or
drugs).
2. Persistent and exaggerated negative beliefs or expectations about oneself, others,
or the world (e.g., “I am bad,” “No one can be trusted,” “The world is completely
dangerous,” “My whole nervous system is permanently ruined”).
3. Persistent, distorted cognitions about the cause or consequences of the traumatic
event(s) that lead the individual to blame himself/herself or others.
4. Persistent negative emotional state (e.g., fear, horror, anger, guilt, or shame).
5. Markedly diminished interest or participation in significant activities.
6. Feelings of detachment or estrangement from others.
7. Persistent inability to experience positive emotions (e.g., inability to experience
happiness, satisfaction, or loving feelings).
E. Marked alterations in arousal and reactivity associated with the traumatic event(s),
beginning or worsening after the traumatic event(s) occurred, as evidenced by two
(or more) of the following:
1. Irritable behavior and angry outbursts (with little or no provocation) typically
expressed as verbal or physical aggression toward people or objects.
2. Reckless or self-destructive behavior.
3. Hypervigilance.
4. Exaggerated startle response.
5. Problems with concentration.
6. Sleep disturbance (e.g., difficulty falling or staying asleep or restless sleep).
F. Duration of the disturbance (Criteria B, C, D, and E) is more than 1 month.
G. The disturbance causes clinically significant distress or impairment in social,
occupational, or other important areas of functioning.
H. The disturbance is not attributable to the physiological effects of a substance (e.g.,
medication, alcohol) or another medical condition.
Specify whether:
With dissociative symptoms:
1. Depersonalization
2. Derealization
Specify if:
With delayed expression
Note. Criteria set above contains only the diagnostic criteria and specifiers; refer to DSM-5 for the full criteria set,
including specifier descriptions and coding and reporting procedures.
Source. DSM-5 criteria for posttraumatic stress disorder reprinted from Diagnostic and Statistical Manual of Mental
Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, pp. 271–272. Used with
permission. Copyright © 2013 American Psychiatric Association.
TABLE 12–13. DSM-5 diagnostic criteria for anxiety disorder due to another
medical condition
A. Panic attacks or anxiety is predominant in the clinical picture.
B. There is evidence from the history, physical examination, or laboratory findings that
the disturbance is the direct pathophysiological consequence of another medical
condition.
C. The disturbance is not better explained by another mental disorder.
D. The disturbance does not occur exclusively during the course of a delirium.
E. The disturbance causes clinically significant distress or impairment in social,
occupational, or other important areas of functioning.
Note. Criteria set above contains only the diagnostic criteria and specifiers; refer to DSM-5 for the full criteria set,
including specifier descriptions and coding and reporting procedures.
Source. DSM-5 criteria for anxiety disorder due to another medical condition reprinted from Diagnostic and
Statistical Manual of Mental Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013, p.
230. Used with permission. Copyright © 2013 American Psychiatric Association.
TABLE 12–14. Suggestions for delineating anxiety in the context of medical illness
and medication use
Factors implicating a physiological cause of anxiety in medically ill older adults
Anxiety symptoms arise de novo in absence of psychiatric history and risk factors
after diagnosis of physical illness.
Symptoms of a medical condition are known to mimic those of anxiety (e.g.,
hyperthyroidism, cardiac conditions).
Anxiety symptoms are a known side effect of recently started medication.
The patient is, or was until recently, taking sedatives or antianxiety medication
(leading to withdrawal symptoms).
Anxiety symptoms have acute onset in the absence of other life events.
Factors implicating a psychological cause of anxiety in medically unwell older
adults
Anxiety symptoms predate physical illness.
Affective disorders predate physical illness.
Anxiety symptoms arise in the context of a chronic illness.
The patient has no recent new medications, medication changes, or issues with
adherence.
The patient has experienced a major life event or change in his or her circumstances.
The patient has one or more present risk factors for an anxiety disorder (e.g., high
score on measures of neuroticism, anxiety sensitivity).
Effect on functioning is characterized by avoidance or by increased anxiety when in
feared situation.
Source. Adapted from Mohlman J, Bryant C, Lenze EJ, et al.: “Improving Recognition of Late Life Anxiety
Disorders in Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition: Observations and
Recommendations of the Advisory Committee to the Lifespan Disorders Work Group.” International Journal of
Geriatric Psychiatry 27(6):549–556, 2012. Used with permission.
Treatment
Anxiety disorders have a waning and waxing course in elderly adults, as in young
adults, but these disorders are unlikely to remit completely in older patients
(Schuurmans et al. 2005). Additionally, anxiety disorders produce quality-of-life
impairments and disability on par with that of late-life depression (de Beurs et al. 1999;
Wetherell et al. 2004) and may be risk factors for the development of anxious
depression, a severe and treatment-resistant illness. These issues suggest the importance
of treatment.
Despite the relative lack of treatment trials, the treatment of late-life anxiety
disorders has been the subject of reviews and meta-analyses (e.g., Pinquart and
Duberstein 2007; Wetherell et al. 2005b). The available evidence suggests that both
psychotherapy (primarily CBT) and pharmacotherapy are effective. However,
considerable gaps exist in the understanding of these treatments. As a result, various
authors have concluded that psychotherapy (Mohlman and Price 2006) or that
pharmacotherapy (Pinquart and Duberstein 2007) should be the first-line treatment, but
this issue is not yet resolved.
Psychotherapy
There is evidence that older adults with GAD can learn new skills in CBT and use them
effectively over time (Wetherell et al. 2005a). In the only study to simultaneously
compare CBT with a credible attention control condition and a waiting list for patients
with late-life GAD, CBT produced large effects on measures of anxiety, depression, and
quality of life relative to the waiting list, whereas the control condition produced small
to medium effects (Wetherell et al. 2003a). Other research has found that CBT is
effective compared with minimal-contact control subjects (Stanley et al. 2003a) or usual
care (Stanley et al. 2003b, 2009). These data show that CBT is an efficacious treatment
for GAD in older adults (although they do not prove that CBT is more effective than
other therapies). It is likely that relaxation therapy is the most effective part of CBT
(Thorp et al. 2009; Yoder et al. 2013); clinicians need to ensure the inclusion of this
relatively easily instructed component with older adults. In addition, in older patients
with PTSD, prolonged exposure therapy appears preliminarily to be effective (Thorp et
al. 2012), suggesting that exposure is an effective component of CBT at all ages.
TABLE 12–15. Guidelines for use of assessment tools for anxiety in older adults
Avoid a yes/no format for screening questions in favor of open-ended questions. A
yes/no screening question requires that subjects know exactly what the symptom is
referring to in context of their own experience.
Avoid numeric frequency/severity format of standard assessments, or supplement with
additional probes.
Avoid sets of questions whose response formats alternate between narrative and
numerical.
Use the term endorsed by the individual to describe anxiety as a concept (e.g.,
“nerves,” “concerns”).
Avoid lengthy, complex, or reverse-coded items.
Use personal timelines to aid in recall. Include informant reports.
Use a lower number of symptoms to meet cutoff or diagnostic criteria compared with
young adults.
CBT for late-life anxiety typically includes education about anxiety and its
symptoms, relaxation skills, cognitive therapy, problem-solving skills training, exposure
exercises, and sleep hygiene instruction (Stanley et al. 2004). Adaptations for CBT in
elderly persons include increased repetition, increased time reviewing previous sessions
(Mohlman and Price 2006), and combining CBT with cognitive rehabilitation training
to enhance logic and reasoning skills (Mohlman 2008; Mohlman et al. 2013).
The evidence is mixed as to whether CBT is a superior treatment to other types of
psychotherapy for late-life anxiety (Barrowclough et al. 2001; Stanley et al. 1996;
Wetherell et al. 2003a). One study directly comparing CBT and pharmacotherapy for
late-life anxiety (mostly GAD and panic disorder) found a greater effect size for
pharmacotherapy (sertraline), although this study had a relatively small sample size
(Schuurmans et al. 2006). In contrast, a study comparing CBT to paroxetine for late-life
panic disorder found no differences in response rate between the treatments, with both
treatments leading to better outcomes than a waitlist control condition (Hendriks et al.
2010). This question of whether medication or psychotherapy is better remains
unresolved, but a more clinically salient question may be whether further enhancements
or a sequential approach of using medications and CBT would be more effective in
older adults. Addressing this issue, Wetherell et al. (2013b) found that augmenting SSRI
treatment with CBT improved worry symptoms and reduced relapse rates in older
adults with GAD. This report suggests that a combination of pharmacotherapy and
psychotherapy approaches may provide the greatest benefits for patients with this
disorder.
No controlled research on psychotherapy that focuses on anxiety conditions other
than GAD exists. Several studies have included patient samples with mixed anxiety
disorders, including panic disorder and social phobia, but none have published disorder-
specific results.
Pharmacotherapy
Some studies have found that benzodiazepines are efficacious in reducing anxiety
symptoms in late-life anxiety (Bresolin et al. 1988; Koepke et al. 1982). However, these
medications increase the risk of falls (e.g., Landi et al. 2005; Lenze et al. 2009a) and
cognitive impairment (Billioti de Gage et al. 2014). This is troubling, given their
widespread use in older adults (Olfson et al. 2014).
Antidepressants—particularly SSRIs, such as escitalopram, citalopram, and
sertraline, and serotonin-norepinephrine reuptake inhibitors, such as duloxetine and
venlafaxine—are considered first-line treatment for anxiety disorders in younger adults
because of these drugs’ lower potential for toxicity or abuse. To our knowledge, three
randomized controlled trials (of which two are pilot studies) have examined the efficacy
of the SSRIs for late-life anxiety disorders (Lenze et al. 2005b, 2009b; Schuurmans et
al. 2006). All of these demonstrated efficacy of SSRIs, although the largest (Lenze et al.
2009b) found only a small effect size. However, the maintenance (i.e., relapse-
prevention) efficacy of maintenance SSRI has been demonstrated, with an apparent
large effect size similar to what has been seen in late-life depression (Wetherell et al.
2013b).
Few studies have been published examining second-line strategies. Of these, the two
largest studies are multisite industry-led studies involving older patients with GAD:
Montgomery et al. (2008) found that pregabalin was efficacious in older adults (similar
to findings in young adults), and Mezhebovsky et al. (2013) found that extended-release
quetiapine was efficacious. Additionally, a small study of risperidone ([Moríñigo et al.
2005]) showed it to be efficacious as an augmentation strategy. None of these
medications, however, has been approved by the U.S. Food and Drug Administration
for treating GAD.
No published studies have focused exclusively on late-life PTSD, but a few studies
involving mixed-age populations, with mean ages ranging from 55 to 60 years, have
suggested that the antidepressants citalopram (English et al. 2006) and mirtazapine
(Chung et al. 2004) and the α-adrenergic blocker prazosin (Raskind et al. 2007) are
effective. Further research is needed to determine optimal treatment for PTSD in elderly
persons.
Clinical Management
Although medication, psychotherapy, and their combination have been shown to be
efficacious in acute and long-term treatment of late-life anxiety disorders, having this
knowledge base is only a small part of what therapists need for managing these
common and debilitating disorders. A list of important management issues for clinicians
is provided in Table 12–16 (Lenze and Wetherell 2011).
Detecting and diagnosing the patient’s anxiety (whether primary or comorbid) are of
utmost importance. Elderly persons with anxiety disorders may not be insightful or
conversant with terms used to define anxiety (e.g., panic, social anxiety). They often
feel that anxiety or fear is a realistic response to their environment or current stressors.
However, undetected anxiety is likely to persist and have a detrimental effect on
treatment of comorbid conditions, such as depression. Anecdotally, we have found that
asking about reactions to stress (e.g., “How do you feel in times of stress?”) is a useful
opener when asking elderly persons about anxiety symptoms. Patients who report
anxiety using terms such as anxious, concerned, or worried can then be asked
additional questions, such as “How often does it occur?” and “What do you do to
manage these feelings?” Asking whether anxiety is excessive or uncontrollable is
unlikely to be fruitful; instead, asking about a patient’s mechanisms to control anxiety
will elicit a more pertinent response. Patients with panic symptoms may not endorse
“panic attacks” when questioned, but they may admit to experiencing brief periods with
multiple physical symptoms (particularly autonomic symptoms such as palpitations).
The clinician should discuss management with the patient while keeping in mind that
anxiety may affect the patient’s views about treatment. Patients’ reactions are
particularly an issue in relation to medications, especially antidepressants, which are
well known to have a stimulating or mildly anxiogenic effect initially before an
antianxiety effect occurs. Anecdotally, we have observed that most patients with late-
life anxiety report being sensitive to or intolerant of antidepressant medications if they
have tried them. Their claim of intolerance likely results from anticipatory concern
about side effects, vigilance toward interoceptive stimuli, and a tendency to
catastrophize about any interoceptive sensations they detect (even if unrelated to the
antidepressant). Therefore, clinicians need to counsel patients in advance about
medication side effects explicitly to reduce patients’ concerns that these might
somehow be toxic or incapacitating. Additionally, management requires close follow-up
after the onset or dosage titration of an antidepressant medication to ensure that the
patient is taking the medication and to address the patient’s perception of side effects.
Clinicians should realize that when a patient claims to have side effects, the patient may
actually be describing anxiety-related symptoms that preceded the start of the medicine
and that are now being attributed to the medicine. It is useful to inform the patient that
such symptoms are expected to get better and are not harmful.
The early use of benzodiazepines may produce a fast anxiolytic action, which could
improve compliance; however, the long-term use of these medications is discouraged in
favor of alternative treatments, such as SSRIs, other antidepressants, or psychotherapy.
Also, it should be noted that benzodiazepines may counteract the effects of
psychotherapy, particularly CBT (Westra and Stewart 1998).
In summary, the detection and management of anxiety disorders in elderly persons is
challenging, but the high prevalence and significant morbidity associated with these
disorders highlight the importance of understanding them. It is expected that as the
knowledge base (regarding diagnosis, etiology, and treatment) of these disorders
improves, anxiety disorders will become a core clinical issue in geriatric psychiatry,
much like dementia and depression.
Key Points
• Late-life anxiety disorders are more common than previously thought but may
escape detection unless specific strategies are used to enhance recognition.
• Antidepressants and psychotherapeutic treatments as well as their combination are
effective in older adults, but probably less so than in young adults.
• Increased vigilance is required of the clinician to manage anxiety in this age group.
• More research is needed to examine the nature, course, and treatment of these
common but still understudied syndromes.
References
Alexopoulos GS, Katz IR, Bruce ML, et al: Remission in depressed geriatric primary
care patients: a report from the PROSPECT study. Am J Psychiatry 162(4):718–
724, 2005 15800144
Allgulander C, Lavori PW: Causes of death among 936 elderly patients with “pure”
anxiety neurosis in Stockholm County, Sweden, and in patients with depressive
neurosis or both diagnoses. Compr Psychiatry 34(5):299–302, 1993 8306638
American Psychiatric Association: Diagnostic and Statistical Manual of Mental
Disorders, 4th Edition. Washington, DC, American Psychiatric Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of Mental
Disorders, 4th Edition, Text Revision. Washington, DC, American Psychiatric
Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of Mental
Disorders, 5th Edition. Washington, DC, American Psychiatric Association, 2013
Andreescu C, Lenze EJ, Dew MA, et al: Effect of comorbid anxiety on treatment
response and relapse risk in late-life depression: controlled study. Br J Psychiatry
190:344–349, 2007 17401042
Andreescu C, Wu M, Butters MA, et al: The default mode network in late-life anxious
depression. Am J Geriatr Psychiatry 19(11):980–983, 2011 21765344
Andreescu C, Sheu LK, Tudorascu D, et al: The ages of anxiety: differences across the
lifespan in the default mode network functional connectivity in generalized anxiety
disorder. Int J Geriatr Psychiatry 2013 Epub ahead of print
Andreescu C, Teverovsky E, Fu B, et al: Old worries and new anxieties: behavioral
symptoms and mild cognitive impairment in a population study. Am J Geriatr
Psychiatry 22(3):274–284, 2014 23759435
Arfken CL, Lach HW, Birge SJ, et al: The prevalence and correlates of fear of falling in
elderly persons living in the community. Am J Public Health 84(4):565–570, 1994
8154557
Aström M: Generalized anxiety disorder in stroke patients: a 3-year longitudinal study.
Stroke 27(2):270–275, 1996 8571422
Ayers CR, Saxena S, Golshan S, et al: Age at onset and clinical features of late life
compulsive hoarding. Int J Geriatr Psychiatry 25(2):142–149, 2010 19548272
Ayers CR, Wetherell JL, Schiehser D, et al: Executive functioning in older adults with
hoarding disorder. Int J Geriatr Psychiatry 28(11):1175–1181, 2013 23440720
Ayers CR, Iqbal Y, Strickland K: Medical conditions in geriatric hoarding disorder
patients. Aging Ment Health 18(2):148–151, 2014a 23863040
Ayers CR, Saxena S, Espejo E, et al: Novel treatment for geriatric hoarding disorder: an
open trial of cognitive rehabilitation paired with behavior therapy. Am J Geriatr
Psychiatry22(3):248–252 2014b 23831173
Ballard C, Neill D, O’Brien J, et al: Anxiety, depression and psychosis in vascular
dementia: prevalence and associations. J Affect Disord 59(2):97–106, 2000
10837878
Barrowclough C, King P, Colville J, et al: A randomized trial of the effectiveness of
cognitive-behavioral therapy and supportive counseling for anxiety symptoms in
older adults. J Consult Clin Psychol 69(5):756–762, 2001 11680552
Beaudreau SA, O’Hara R: Late-life anxiety and cognitive impairment: a review. Am J
Geriatr Psychiatry 16(10):790–803, 2008 18827225
Beekman AT, Bremmer MA, Deeg DJ, et al: Anxiety disorders in later life: a report
from the Longitudinal Aging Study Amsterdam. Int J Geriatr Psychiatry 13(10):
717–726, 1998 9818308
Beekman ATF, de Beurs E, van Balkom AJLM, et al: Anxiety and depression in later
life: co-occurrence and communality of risk factors. Am J Psychiatry 157(1):89–
95, 2000 10618018
Billioti de Gage S, Moride Y, Ducruet T, et al: Benzodiazepine use and risk of
Alzheimer’s disease: case-control study. BMJ 349:g5205, 2014 25208536
Blazer D, George KL, Hughes D: The epidemiology of anxiety disorders: an age
comparison, in Anxiety in the Elderly: Treatment and Research. Edited by Salzman
C, Lebowitz BD. New York, Springer, 1991, pp 17–30
Bresolin N, Monza G, Scarpini E, et al: Treatment of anxiety with ketazolam in elderly
patients. Clin Ther 10(5):536–542, 1988 2908803
Bruce SE, Yonkers KA, Otto MW, et al: Influence of psychiatric comorbidity on
recovery and recurrence in generalized anxiety disorder, social phobia, and panic
disorder: a 12-year prospective study. Am J Psychiatry 162(6):1179–1187, 2005
15930067
Bryant C, Mohlman J, Gum A, et al: Anxiety disorders in older adults: looking to
DSM5 and beyond. Am J Geriatr Psychiatry 21(9):872–876, 2013 23567396
Burvill PW, Johnson GA, Jamrozik KD, et al: Anxiety disorders after stroke: results
from the Perth Community Stroke Study. Br J Psychiatry 166(3):328–332, 1995
7788124
Butters MA, Bhalla RK, Andreescu C, et al: Changes in neuropsychological functioning
following treatment for late-life generalised anxiety disorder. Br J Psychiatry
199(3):211–218, 2011 21727232
Byers AL, Yaffe K, Covinsky KE, et al: High occurrence of mood and anxiety disorders
among older adults: the National Comorbidity Survey Replication. Arch Gen
Psychiatry 67(5):489–496, 2010 20439830
Cairney J, McCabe L, Veldhuizen S, et al: Epidemiology of social phobia in later life.
Am J Geriatr Psychiatry 15(3):224–233, 2007 17213375
Caudle DD, Senior AC, Wetherell JL, et al: Cognitive errors, symptom severity, and
response to cognitive behavior therapy in older adults with generalized anxiety
disorder. Am J Geriatr Psychiatry 15(8):680–689, 2007 17670997
Chacko RC, Corbin MA, Harper RG: Acquired obsessive-compulsive disorder
associated with basal ganglia lesions. J Neuropsychiatry Clin Neurosci 12(2):269–
272, 2000 11001608
Charles ST, Reynolds CA, Gatz M: Age-related differences and change in positive and
negative affect over 23 years. J Pers Soc Psychol 80(1):136–151, 2001 11195886
Chopra MP, Zhang H, Pless Kaiser A, et al: PTSD is a chronic, fluctuating disorder
affecting the mental quality of life in older adults. Am J Geriatr Psychiatry
22(1):86–97, 2014 24314889
Chou KL, Mackenzie CS, Liang K, et al: Three-year incidence and predictors of first-
onset of DSM-IV mood, anxiety, and substance use disorders in older adults:
results from Wave 2 of the National Epidemiologic Survey on Alcohol and Related
Conditions. J Clin Psychiatry 72(2):144–155, 2011 21382305
Chung MY, Min KH, Jun YJ, et al: Efficacy and tolerability of mirtazapine and
sertraline in Korean veterans with posttraumatic stress disorder: a randomized open
label trial. Hum Psychopharmacol 19(7):489–494, 2004 15378676
Cuthbert BN: Translating intermediate phenotypes to psychopathology: the NIMH
Research Domain Criteria. Psychophysiology 51(12):1205–1206, 2014 25387702
de Beurs E, Beekman ATF, van Balkom AJLM, et al: Consequences of anxiety in older
persons: its effect on disability, well-being and use of health services. Psychol Med
29(3):583–593, 1999 10405079
DeLuca AK, Lenze EJ, Mulsant BH, et al: Comorbid anxiety disorder in late life
depression: association with memory decline over four years. Int J Geriatr
Psychiatry 20(9):848–854, 2005 16116585
Dew MA, Reynolds CF 3rd, Houck PR, et al: Temporal profiles of the course of
depression during treatment: predictors of pathways toward recovery in the elderly.
Arch Gen Psychiatry 54(11):1016–1024, 1997 9366658
Dew MA, Kormos RL, DiMartini AF, et al: Prevalence and risk of depression and
anxiety-related disorders during the first three years after heart transplantation.
Psychosomatics 42(4):300–313, 2001 11496019
Diefenbach GJ, Stanley MA, Beck JG: Worry content reported by older adults with and
without generalized anxiety disorder. Aging Ment Health 5(3):269–274, 2001
11575066
Dombrovski AY, Mulsant BH, Houck PR, et al: Residual symptoms and recurrence
during maintenance treatment of late-life depression. J Affect Disord 103(1–3): 77–
82, 2007 17321595
Dong X, Simon MA, Mosqueda L, Evans DA: The prevalence of elder self-neglect in a
community-dwelling population: hoarding, hygiene, and environmental hazards. J
Aging Health 24(3):507–524, 2012 22187089
English BA, Jewell M, Jewell G, et al: Treatment of chronic posttraumatic stress
disorder in combat veterans with citalopram: an open trial. J Clin Psychopharmacol
26(1):84–88, 2006 16415713
Ettner SL, Hermann RC: Provider specialty choice among Medicare beneficiaries
treated for psychiatric disorders. Health Care Financ Rev 18(3):43–59, 1997
10173123
Ferretti L, McCurry SM, Logsdon R, et al: Anxiety and Alzheimer’s disease. J Geriatr
Psychiatry Neurol 14(1):52–58, 2001 11281317
Flint AJ: Epidemiology and comorbidity of anxiety disorders in the elderly. Am J
Psychiatry 151(5):640–649, 1994 8166303
Flint AJ: Anxiety and its disorders in late life: moving the field forward. Am J Geriatr
Psychiatry 13(1):3–6, 2005a 15653934
Flint AJ: Generalised anxiety disorder in elderly patients: epidemiology, diagnosis and
treatment options. Drugs Aging 22(2):101–114, 2005b 15733018
Flint AJ, Rifat SL: Two-year outcome of elderly patients with anxious depression.
Psychiatry Res 66(1):23–31, 1997 9061801
Flint A, Bradwejn J, Vaccarino F, et al: Aging and panicogenic response to
cholecystokinin tetrapeptide: an examination of the cholecystokinin system.
Neuropsychopharmacology 27(4):663–671, 2002 12377403
Ford BC, Bullard KM, Taylor RJ, et al: Lifetime and 12-month prevalence of
Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition disorders
among older African Americans: findings from the National Survey of American
Life Am J Geriatr Psychiatry 15(8):652–659, 2007 17504908
Frueh BC, Grubaugh AL, Acierno R, et al: Age differences in posttraumatic stress
disorder, psychiatric disorders, and healthcare service use among veterans in
Veterans Affairs primary care clinics. Am J Geriatr Psychiatry 15(8):660–672,
2007 17670996
Gagnon N, Flint AJ, Naglie G, et al: Affective correlates of fear of falling in elderly
persons. Am J Geriatr Psychiatry 13(1):7–14, 2005 15653935
Grant JE, Mancebo MC, Pinto A, et al: Late-onset obsessive compulsive disorder:
clinical characteristics and psychiatric comorbidity. Psychiatry Res 152(1):21–27,
2007 17363071
Gum AM, Petkus A, McDougal SJ, et al: Behavioral health needs and problem
recognition by older adults receiving home-based aging services. Int J Geriatr
Psychiatry 24(4):400–408, 2009 18836987
Hek K, Direk N, Newson RS, et al: Anxiety disorders and salivary cortisol levels in
older adults: a population-based study. Psychoneuroendocrinology 38(2):300–305,
2013 22776419
Hendriks GJ, Keijsers GP, Kampman M, et al: A randomized controlled study of
paroxetine and cognitive-behavioural therapy for late-life panic disorder. Acta
Psychiatr Scand 122(1):11–19, 2010 19958308
Hettema JM: What is the genetic relationship between anxiety and depression? Am J
Med Genet C Semin Med Genet 148C(2): 140–146, 2008 18412101
Hettema JM, Kuhn JW, Prescott CA, et al: The impact of generalized anxiety disorder
and stressful life events on risk for major depressive episodes. Psychol Med
36(6):789–795, 2006 16515735
Hwang SH, Kwon JS, Shin YW, et al: Neuropsychological profiles of patients with
obsessive-compulsive disorder: early onset versus late onset. J Int Neuropsychol
Soc 13(1):30–37, 2007 17166301
Jarvik LF, Russell D: Anxiety, aging and the third emergency reaction. J Gerontol
34(2):197–200, 1979 438473
Jayasinghe N, Rocha LP, Sheeran T, et al: Anxiety symptoms in older home health care
recipients: prevalence and associates. Home Health Care Serv Q 32(3):163–177,
2013 23937710
Jeste ND, Hays JC, Steffens DC: Clinical correlates of anxious depression among
elderly patients with depression. J Affect Disord 90(1):37–41, 2006 16325261
Kendler KS, Neale MC, Kessler RC, et al: Major depression and generalized anxiety
disorder: same genes, (partly) different environments? Arch Gen Psychiatry
49(9):716–722, 1992 1514877
Kendler KS, Gardner CO, Gatz M, Pedersen NL: The sources of co-morbidity between
major depression and generalized anxiety disorder in a Swedish national twin
sample. Psychol Med 37(3):453–462, 2007 17121688
Kessler RC, Amminger GP, Aguilar-Gaxiola S, et al: Age of onset of mental disorders:
a review of recent literature. Curr Opin Psychiatry 20(4):359–364, 2007 17551351
King-Kallimanis B, Gum AM, Kohn R: Comorbidity of depressive and anxiety
disorders for older Americans in the National Comorbidity Survey-Replication.
Am J Geriatr Psychiatry 17(9):782–792, 2009 19700950
Koepke HH, Gold RL, Linden ME, et al: Multicenter controlled study of oxazepam in
anxious elderly outpatients. Psychosomatics 23(6):641–645, 1982 6750675
Landi F, Onder G, Cesari M, et al: Psychotropic medications and risk for falls among
community-dwelling frail older people: an observational study. J Gerontol A Biol
Sci Med Sci 60(5):622–626, 2005 15972615
Lenze EJ, Wetherell JL: Bringing the bedside to the bench, and then to the community:
a prospectus for intervention research in late-life anxiety disorders. Int J Geriatr
Psychiatry 24(1):1–14, 2009 18613267
Lenze EJ, Wetherell JL: A lifespan view of anxiety disorders. Dialogues Clin Neurosci
13(4):381–399, 2011 22275845
Lenze EJ, Mulsant BH, Shear MK, et al: Comorbid anxiety disorders in depressed
elderly patients. Am J Psychiatry 157(5): 722–728, 2000 10784464
Lenze EJ, Mulsant BH, Dew MA, et al: Good treatment outcomes in late-life depression
with comorbid anxiety. J Affect Disord 77(3):247–254, 2003 14612224
Lenze EJ, Mulsant BH, Mohlman J, et al: Generalized anxiety disorder in late life:
lifetime course and comorbidity with major depressive disorder. Am J Geriatr
Psychiatry 13(1):77–80, 2005a 15653943
Lenze EJ, Mulsant BH, Shear MK, et al: Efficacy and tolerability of citalopram in the
treatment of late-life anxiety disorders: results from an 8-week randomized,
placebo-controlled trial. Am J Psychiatry 162(1):146–150, 2005b 15625213
Lenze EJ, Iaboni A, Wetherell J: Benzodiazepines in older adults: definite harms,
doubtful benefits [letter]. BMJ 349:g5205, 2009a. Available at:
http://www.bmj.com/content/349/bmj.g5205/rr/777842. Accessed December 4,
2014.
Lenze EJ, Rollman BL, Shear MK, et al: Escitalopram for older adults with generalized
anxiety disorder: a randomized controlled trial. JAMA 301(3):295–303, 2009b
19155456
Lenze EJ, Mantella RC, Shi P, et al: Elevated cortisol in older adults with generalized
anxiety disorder is reduced by treatment: a placebo-controlled evaluation of
escitalopram. Am J Geriatr Psychiatry 19(5):482–490, 2011 20808146
Lenze EJ, Dixon D, Mantella RC, et al: Treatment-related alteration of cortisol predicts
change in neuropsychological function during acute treatment of late-life anxiety
disorder. Int J Geriatr Psychiatry 27(5):454–462, 2012 21681817
Lenze EJ, Hickman S, Hershey T, et al: Mindfulness-based stress reduction for older
adults with worry symptoms and co-occurring cognitive dysfunction. Int J Geriatr
Psychiatry 29(10):991–1000, 2014a 24677282
Lenze EJ, Hershey T, Newcomer JW, et al: Antiglucocorticoid therapy for older adults
with anxiety and co-occurring cognitive dysfunction: results from a pilot study with
mifepristone. Int J Geriatr Psychiatry 29(9):962–969, 2014b 24633761
Le Roux H, Gatz M, Wetherell JL: Age at onset of generalized anxiety disorder in older
adults. Am J Geriatr Psychiatry 13(1):23–30, 2005 15653937
Mackenzie CS, Reynolds K, Chou KL, et al: Prevalence and correlates of generalized
anxiety disorder in a national sample of older adults. Am J Geriatr Psychiatry
19(4):305–315, 2011 21427639
Mantella RC, Butters MA, Dew MA, et al: Cognitive impairment in late-life
generalized anxiety disorder. Am J Geriatr Psychiatry 15(8):673–679, 2007
17426260
Mantella RC, Butters MA, Amico JA, et al: Salivary cortisol is associated with
diagnosis and severity of late-life generalized anxiety disorder.
Psychoneuroendocrinology 33(6):773–781, 2008 18407426
McCabe L, Cairney J, Veldhuizen S, et al: Prevalence and correlates of agoraphobia in
older adults. Am J Geriatr Psychiatry 14(6):515–522, 2006 16731720
Mewton L, Sachdev PS, Andrews G: A naturalistic study of the acceptability and
effectiveness of internet-delivered cognitive behavioural therapy for psychiatric
disorders in older Australians. PLoS ONE 8(8):e71825, 2013 23951253
Mezhebovsky I, Mägi K, She F, et al: Double-blind, randomized study of extended
release quetiapine fumarate (quetiapine XR) monotherapy in older patients with
generalized anxiety disorder. Int J Geriatr Psychiatry 28(6):615–625, 2013
23070803
Mintzer JE, Brawman-Mintzer O: Agitation as a possible expression of generalized
anxiety disorder in demented elderly patients: toward a treatment approach. J Clin
Psychiatry 57 (suppl 7):55–63; discussion 73–75, 1996 8690698
Mohlman J: More power to the executive? A preliminary test of CBT plus executive
skills training for treatment of late-life GAD. Cognitive and Behavioral Practice
15(3):306–316, 2008
Mohlman J, Price R: Recognizing and treating late-life generalized anxiety disorder:
distinguishing features and psychosocial treatment. Expert Rev Neurother
6(10):1439–1445, 2006 17078785
Mohlman J, Price RB, Eldreth DA, et al: The relation of worry to prefrontal cortex
volume in older adults with and without generalized anxiety disorder. Psychiatry
Res 173(2):121–127, 2009 19559575
Mohlman J, Bryant C, Lenze EJ, et al: Improving recognition of late life anxiety
disorders in Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition:
observations and recommendations of the Advisory Committee to the Lifespan
Disorders Work Group. Int J Geriatr Psychiatry 27(6):549–556, 2012 21773996
Mohlman J, Sirota KG, Papp LA, et al: Clinical interviewing with older adults. Cognit
Behav Pract 19:89–100, 2013
Montgomery S, Chatamra K, Pauer L, et al: Efficacy and safety of pregabalin in elderly
people with generalised anxiety disorder. Br J Psychiatry 193(5):389–394, 2008
18978320
Moríñigo A, Blanco M, Labrador J, et al: Risperidone for resistant anxiety in elderly
persons. Am J Geriatr Psychiatry 13(1): 81–82, 2005 15653944
Mulsant BH, Reynolds CF 3rd, Shear MK, et al: Comorbid anxiety disorders in late-life
depression. Anxiety 2(5):242–247, 1996 9160629
Nelson JC, Delucchi K, Schneider LS: Anxiety does not predict response to
antidepressant treatment in late life depression: results of a meta-analysis. Int J
Geriatr Psychiatry 24(5):539–544, 2009 19334041
Nestadt G, Bienvenu OJ, Cai G, et al: Incidence of obsessive-compulsive disorder in
adults. J Nerv Ment Dis 186(7):401–406, 1998 9680040
Nestadt G, Samuels J, Riddle M, et al: A family study of obsessive-compulsive
disorder. Arch Gen Psychiatry 57(4):358–363, 2000 10768697
Olfson M, King M, Schoenbaum M: Benzodiazepine use in the United States. JAMA
Psychiatry. 2014 Dec 17. [Epub ahead of print] 25517224
Pietrzak RH, Goldstein RB, Southwick SM, et al: Psychiatric comorbidity of full and
partial posttraumatic stress disorder among older adults in the United States: results
from wave 2 of the National Epidemiologic Survey on Alcohol and Related
Conditions. Am J Geriatr Psychiatry 20(5):380–390, 2012a 22522959
Pietrzak RH, Maruff P, Woodward M, et al: Mild worry symptoms predict decline in
learning and memory in healthy older adults: a 2-year prospective cohort study. Am
J Geriatr Psychiatry 20(3):266–275, 2012b 22354117
Pinquart M, Duberstein PR: Treatment of anxiety disorders in older adults: a meta-
analytic comparison of behavioral and pharmacological interventions. Am J Geriatr
Psychiatry 15(8):639–651, 2007 17670995
Pomara N, Willoughby LM, Sidtis JJ, et al: Cortisol response to diazepam: its
relationship to age, dose, duration of treatment, and presence of generalized anxiety
disorder. Psychopharmacology (Berl) 178(1):1–8, 2005 15338100
Price RB, Mohlman J: Inhibitory control and symptom severity in late life generalized
anxiety disorder. Behav Res Ther 45(11):2628–2639, 2007 17662240
Price RB, Eldreth DA, Mohlman J: Deficient prefrontal attentional control in late-life
generalized anxiety disorder: an fMRI investigation. Transl Psychiatr 1:e46, 2011
22833192
Raskind MA, Peskind ER, Hoff DJ, et al: A parallel group placebo controlled study of
prazosin for trauma nightmares and sleep disturbance in combat veterans with post-
traumatic stress disorder. Biol Psychiatry 61(8):928–934, 2007 17069768
Regier DA, Narrow WE, Rae DS: The epidemiology of anxiety disorders: the
Epidemiologic Catchment Area (ECA) experience. J Psychiatr Res 24 (suppl 2):3–
14, 1990 2280373
Rosenberg PB, Mielke MM, Appleby BS, et al: The association of neuropsychiatric
symptoms in MCI with incident dementia and Alzheimer disease. Am J Geriatr
Psychiatry 21(7):685–695, 2013 23567400
Rubio G, López-Ibor JJ: Generalized anxiety disorder: a 40-year follow-up study. Acta
Psychiatr Scand 115(5):372–379, 2007 17430415
Saxena S: Is compulsive hoarding a genetically and neurobiologically discrete
syndrome? Implications for diagnostic classification. Am J Psychiatry 164(3):380–
384, 2007 17329459
Schoevers RA, Beekman ATF, Deeg DJH, et al: Comorbidity and risk-patterns of
depression, generalised anxiety disorder and mixed anxiety-depression in later life:
results from the AMSTEL study. Int J Geriatr Psychiatry 18(11):994–1001, 2003
14618550
Schuitevoerder S, Rosen JW, Twamley EW, et al: A meta-analysis of cognitive
functioning in older adults with PTSD. J Anxiety Disord 27(6):550–558, 2013
23422492
Schuurmans J, Comijs HC, Beekman ATF, et al: The outcome of anxiety disorders in
older people at 6-year follow-up: results from the Longitudinal Aging Study
Amsterdam. Acta Psychiatr Scand 111(6):420–428, 2005 15877708
Schuurmans J, Comijs H, Emmelkamp PM, et al: A randomized, controlled trial of the
effectiveness of cognitive-behavioral therapy and sertraline versus a waitlist control
group for anxiety disorders in older adults. Am J Geriatr Psychiatry 14(3):255–263,
2006 16505130
Seignourel PJ, Kunik ME, Snow L, et al: Anxiety in dementia: a critical review. Clin
Psychol Rev 28(7):1071–1082, 2008 18555569
Sharma RP, Pandey GN, Janicak PG, et al: The effect of diagnosis and age on the DST:
a metaanalytic approach. Biol Psychiatry 24(5):555–568, 1988 3167144
Sheikh JI, Swales PJ, Carlson EB, et al: Aging and panic disorder: phenomenology,
comorbidity, and risk factors. Am J Geriatr Psychiatry 12(1):102–109, 2004
14729565
Sinoff G, Werner P: Anxiety disorder and accompanying subjective memory loss in the
elderly as a predictor of future cognitive decline. Int J Geriatr Psychiatry
18(10):951–959, 2003 14533128
Snowdon J, Shah A, Halliday G: Severe domestic squalor: a review. Int Psychogeriatr
19(1):37–51, 2007 16973099
Stanley MA, Beck JG, Glassco JD: Treatment of generalized anxiety in older adults: a
preliminary comparison of cognitive-behavioral and supportive approaches. Behav
Ther 27:565–581, 1996
Stanley MA, Beck JG, Novy DM, et al: Cognitive-behavioral treatment of late-life
generalized anxiety disorder. J Consult Clin Psychol 71(2):309–319, 2003a
12699025
Stanley MA, Hopko DR, Diefenbach GJ, et al: Cognitive-behavior therapy for late-life
generalized anxiety disorder in primary care: preliminary findings. Am J Geriatr
Psychiatry 11(1):92–96, 2003b 12527544
Stanley MA, Diefenbach GJ, Hopko DR: Cognitive behavioral treatment for older
adults with generalized anxiety disorder: a therapist manual for primary care
settings. Behav Modif 28(1):73–117, 2004 14710708
Stanley MA, Wilson NL, Novy DM, et al: Cognitive behavior therapy for generalized
anxiety disorder among older adults in primary care: a randomized clinical trial.
JAMA 301(14):1460–1467, 2009 19351943
Stanley MA, Calleo J, Bush AL, et al: The peaceful mind program: a pilot test of a
cognitive-behavioral therapy-based intervention for anxious patients with
dementia. Am J Geriatr Psychiatry 21(7):696–708, 2013 23567399
Starkstein SE, Jorge R, Petracca G, et al: The construct of generalized anxiety disorder
in Alzheimer disease. Am J Geriatr Psychiatry 15(1):42–49, 2007 17194814
Steffens DC, McQuoid DR: Impact of symptoms of generalized anxiety disorder on the
course of late-life depression. Am J Geriatr Psychiatry 13(1):40–47, 2005
15653939
Streiner DL, Cairney J, Veldhuizen S: The epidemiology of psychological problems in
the elderly. Can J Psychiatry 51(3):185–191, 2006 16618010
Swoboda KJ, Jenike MA: Frontal abnormalities in a patient with obsessive-compulsive
disorder: the role of structural lesions in obsessive-compulsive behavior. Neurology
45(12):2130–2134, 1995 8848180
Sylvester CM, Corbetta M, Raichle ME, et al: Functional network dysfunction in
anxiety and anxiety disorders. Trends Neurosci 35(9):527–535, 2012 22658924
Teachman BA: Aging and negative affect: the rise and fall and rise of anxiety and
depression symptoms. Psychol Aging 21(1): 201–207, 2006 16594806
Thorp SR, Ayers CR, Nuevo R, et al: Meta-analysis comparing different behavioral
treatments for late-life anxiety. Am J Geriatr Psychiatry 17(2):105–115, 2009
19155744
Thorp SR, Stein MB, Jeste DV, et al: Prolonged exposure therapy for older veterans
with posttraumatic stress disorder: a pilot study. Am J Geriatr Psychiatry
20(3):276–280, 2012 22273763
Todaro JF, Shen BJ, Raffa SD, et al: Prevalence of anxiety disorders in men and women
with established coronary heart disease. J Cardiopulm Rehabil Prev 27(2):86–91,
2007 17558244
Tolin DF, Robison JT, Gaztambide S, et al: Anxiety disorders in older Puerto Rican
primary care patients. Am J Geriatr Psychiatry 13(2):150–156, 2005 15703324
Trollor JN, Anderson TM, Sachdev PS, et al: Prevalence of mental disorders in the
elderly: the Australian National Mental Health and Well-Being Survey. Am J
Geriatr Psychiatry 15(6):455–466, 2007 17545446
van Haastregt JC, Zijlstra GA, van Rossum E, et al: Feelings of anxiety and symptoms
of depression in community-living older persons who avoid activity for fear of
falling. Am J Geriatr Psychiatry 16(3):186–193, 2008 18310549
van Zelst WH, de Beurs E, Beekman AT, et al: Prevalence and risk factors of
posttraumatic stress disorder in older adults. Psychother Psychosom 72(6):333–
342, 2003 14526136
Velayudhan L, Katz AW: Late-onset obsessive-compulsive disorder: the role of stressful
life events. Int Psychogeriatr 18(2):341–344, 2006 16420720
Vink D, Aartsen MJ, Schoevers RA: Risk factors for anxiety and depression in the
elderly: a review. J Affect Disord 106(1–2):29–44, 2008 17707515
Wang PS, Berglund P, Olfson M, et al: Failure and delay in initial treatment contact
after first onset of mental disorders in the National Comorbidity Survey
Replication. Arch Gen Psychiatry 62(6):603–613, 2005 15939838
Watson D: Rethinking the mood and anxiety disorders: a quantitative hierarchical
model for DSM-V. J Abnorm Psychol 114(4):522–536, 2005 16351375
Weiss AP, Jenike MA: Late-onset obsessive-compulsive disorder: a case series. J
Neuropsychiatry Clin Neurosci 12(2):265–268, 2000 11001607
Westra HA, Stewart SH: Cognitive behavioural therapy and pharmacotherapy:
complementary or contradictory approaches to the treatment of anxiety? Clin
Psychol Rev 18(3):307–340, 1998 9564583
Wetherell JL, Gatz M, Pedersen NL: A longitudinal analysis of anxiety and depressive
symptoms. Psychol Aging 16(2):187–195, 2001 11405307
Wetherell JL, Reynolds CA, Gatz M, et al: Anxiety, cognitive performance, and
cognitive decline in normal aging. J Gerontol B Psychol Sci Soc Sci 57(3):P246–
P255, 2002 11983736
Wetherell JL, Gatz M, Craske MG: Treatment of generalized anxiety disorder in older
adults. J Consult Clin Psychol 71(1):31–40, 2003a 12602423
Wetherell JL, Le Roux H, Gatz M: DSM-IV criteria for generalized anxiety disorder in
older adults: distinguishing the worried from the well. Psychol Aging 18(3):622–
627, 2003b 14518822
Wetherell JL, Thorp SR, Patterson TL, et al: Quality of life in geriatric generalized
anxiety disorder: a preliminary investigation. J Psychiatr Res 38(3):305–312, 2004
15003436
Wetherell JL, Hopko DR, Diefenbach GJ, et al: Cognitive-behavioral therapy for late-
life generalized anxiety disorder: who gets better? Behav Ther 36:147–156, 2005a
Wetherell JL, Lenze EJ, Stanley MA: Evidence-based treatment of geriatric anxiety
disorders. Psychiatr Clin North Am 28:871–896, ix, 2005b
Wetherell JL, Petkus AJ, McChesney K, et al: Older adults are less accurate than
younger adults at identifying symptoms of anxiety and depression. J Nerv Ment Dis
197(8):623–626, 2009 19684501
Wetherell JL, Johnson K, Chang DG, et al: Activity, Balance, Learning, and Exposure
(ABLE): a new intervention for excessive fear of falling. Poster session presented
at the annual meeting of the American Association for Geriatric Psychiatry, Los
Angeles, CA, March 2013a
Wetherell JL, Petkus AJ, White KS, et al: Antidepressant medication augmented with
cognitive-behavioral therapy for generalized anxiety disorder in older adults. Am J
Psychiatry 170(7):782–789, 2013b 23680817
Wilson RS, Begeny CT, Boyle PA, et al: Vulnerability to stress, anxiety, and
development of dementia in old age. Am J Geriatr Psychiatry 19(4):327–334, 2011
21427641
Wolitzky-Taylor KB, Castriotta N, Lenze EJ, et al: Anxiety disorders in older adults: a
comprehensive review. Depress Anxiety 27(2):190–211, 2010 20099273
World Health Organization: International Statistical Classification of Diseases and
Related Health Problems, 10th Revision. Geneva, Switzerland, World Health
Organization, 1992
Yaffe K, Vittinghoff E, Lindquist K, et al: Posttraumatic stress disorder and risk of
dementia among U.S. veterans. Arch Gen Psychiatry 67(6):608–613, 2010
20530010
Yehuda R, Golier JA, Tischler L, et al: Hippocampal volume in aging combat veterans
with and without post-traumatic stress disorder: relation to risk and resilience
factors. J Psychiatr Res 41(5):435–445, 2007 16445942
Yoder MS, Lozano B, Center KB, et al: Effectiveness of prolonged exposure for PTSD
in older veterans. Int J Psychiatry Med 45(2):111–124, 2013 23977816
Zijlstra GA, van Haastregt JC, van Eijk JT, et al: Prevalence and correlates of fear of
falling, and associated avoidance of activity in the general population of
community-living older people. Age Ageing 36(3):304–309, 2007 17379605
Suggested Readings
Beaudreau SA, O’Hara R: Late-life anxiety and cognitive impairment: a review. Am J
Geriatr Psychiatry 16(10):790–803, 2008
Lenze EJ, Wetherell JL: A lifespan view of anxiety disorders. Dialogues Clin Neurosci
13(4):381–399, 2011
Lenze EJ, Iaboni A, Wetherell J: Benzodiazepines in older adults: definite harms,
doubtful benefits [letter]. BMJ 349:g5205, 2009. Available at:
http://www.bmj.com/content/349/bmj.g5205/rr/777842. Accessed December 4,
2014.
Mohlman J, Bryant C, Lenze EJ, et al: Improving recognition of late life anxiety
disorders in Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition:
observations and recommendations of the Advisory Committee to the Lifespan
Disorders Work Group. Int J Geriatr Psychiatry 27(6):549–556, 2012
Wetherell JL, Petkus AJ, White KS, et al: Antidepressant medication augmented with
cognitive-behavioral therapy for generalized anxiety disorder in older adults. Am J
Psychiatry 170(7):782–789, 2013
CHAPTER 13
Clinical Features
Prominent somatic symptoms that cause significant distress and impairment
are seen commonly in outpatient settings. When these symptoms shift from
transient expressions of somatic concern to more serious bodily
preoccupation and impairment, and begin to cause excessive emotional or
behavioral responses that are out of proportion to what would be expected
given the physical symptoms, somatic symptom disorder becomes a more
likely diagnosis. Somatic symptoms are experienced by the affected
individual as real physical sensations, pain, or discomfort, usually
indistinguishable from symptoms of actual medical disorders and frequently
coexisting with them. Associated psychological factors are presumed but
not always apparent, and patients vary in their degree of insight into such
factors. In many but not all cases, no clear organic causes emerge from
appropriate workup. Three important factors associated with all of the
disorders in this class are high sensitivity to somatic sensations and pain;
catastrophizing, in which there are excessive and unrealistic fears regarding
one’s health status; and excess functional disability associated with the
symptoms (Bortz 2008; Egloff et al. 2014).
Patients with somatic symptom and related disorders are often able to
accept that their symptoms may be functional and have psychological roots.
Unlike malingering, however, the disorders in this class do not represent
intentional, conscious attempts by patients to present physical symptoms in
order to achieve a specific goal (e.g., to get out of work). In DSM-IV,
somatoform disorders were classified separately from factitious disorders
such as Munchausen’s disease, because the etiologies of the former were
considered to be wholly unconscious and not always aimed at assuming the
sick role (as noted in the introduction to this chapter, factitious disorder
now falls under somatic symptom and related disorders). Neither do
somatoform disorders represent delusional thinking as found in psychotic
states. Somatic symptom and related disorders also differ from
psychosomatic disorders, which are characterized by actual disease states
with presumed psychological triggers.
Conversion Disorder
Conversion disorder, also labeled functional neurological symptom disorder
in DSM-5 (Table 13–3), is characterized by one or more symptoms of
altered voluntary motor or sensory function, which do not appear
compatible with recognized neurological or medical conditions. The
symptoms cause significant distress and/or impairment in important areas of
functioning, and may involve weakness or paralysis, abnormal movements
(e.g., tremor), swallowing or speech symptoms, pseudoseizures, or sensory
loss. As with other somatic symptoms, however, the presence of true
medical comorbidity can cloud the picture. Some researchers have argued
that conversion disorder is actually a form of a dissociative disorder (Brown
et al. 2007). Previous DSM nomenclature for conversion symptoms was
rooted in psychodynamic theory and assumed the presence of a
psychological conflict that seemed to be prompting the symptom.
Etiology
The causes of somatic symptom and related disorders are usually
multifactorial and are often rooted in both early developmental experiences
and personality traits. For example, these disorders have been associated
with the experience of serious illness early in life (Stuart and Noyes 1999),
childhood abuse (Roelofs et al. 2002; Samelius et al. 2007; Waldinger et al.
2006), dissociative amnesia (Brown et al. 2005), significant psychological
stress (Hollifield et al. 1999; Ritsner et al. 2000), and the personality traits
of alexithymia and neuroticism (Bailey and Henry 2007; De Gucht 2003).
As noted throughout the chapter, somatic symptom and related disorders are
also highly associated with comorbid depression, anxiety and panic
disorders, substance abuse, and personality disorders, especially paranoid,
avoidant, and obsessive-compulsive personality disorders (Fallon et al.
2012; Noyes et al. 2001; Rabinowitz and Laek 2005; Sar et al. 2004).
Somatization may be more common in women and in older individuals,
although the prevalence of actual somatic symptom and related disorders
has not been associated with increased age, with the exception of
hypochondriasis. When present in late life, especially with late onset,
somatic symptom and related disorders may be associated with
neuropsychological impairment and/or comorbid neurological illness
(Sheehan and Banerjee 1999). Functional magnetic resonance imaging
scans have found distinctive patterns of increased cerebral activation in the
left inferior frontal lobe and in left limbic structures associated with
somatoform symptoms (Stone et al. 2007; Vuilleumier 2005). Specific
neuroimaging studies of patients with conversion symptoms have found
decreased activity in motor or somatosensory cortex, along with increased
activation in orbitofrontal and anterior cingulate cortex (Vuilleumier et al.
2001; Yazici and Kostakoglu 1998). Nonetheless, there is not yet sufficient
evidence to accurately identify any somatic symptom disorder using
neuroimaging (Browning et al. 2011). Overall factors associated with
somatic symptom and related disorders are summarized in Table 13–4.
Psychodynamic approaches suggest that somatic symptom and related
disorders result from unconscious conflict in which intolerable impulses or
affects are believed to be expressed through more tolerable somatic
symptoms or complaints. The classic example of this phenomenon is found
in conversion disorder, in which intolerable, unconscious impulses are
believed to be converted into motor or sensory dysfunction. Freud first
wrote about such a mechanism based on his studies of young women who
had what was then termed hysteria (Breuer and Freud 1893–1895/1955).
Specifically, psychodynamic theory suggests that excessive and intolerable
guilt or hostility is the psychological source of somatization—in particular,
hypochondriasis (Barsky and Klerman 1983). In such cases, physical
symptoms serve as a means of self-punishment for unacceptable
unconscious impulses. Anger directed toward caregivers is indirectly
expressed through distrust of and dissatisfaction with multiple physicians.
Some researchers have suggested that underlying and complicating this
psychodynamic rechannelization of anger or guilt is the trait of alexithymia,
in which an individual has a relative inability to identify and express
emotional states (Bailey and Henry 2007; Waller and Scheidt 2004). The
experiencing and reporting of bodily sensations thus substitute for
emotional expression. Although alexithymia has long been postulated to
play a role in both somatoform and psychosomatic illnesses, not all
empirical research has supported the correlation of alexithymia with
somatic complaints (Lundh and Simonsson-Sarnecki 2001).
Treatment
By definition, patients with somatic symptom and related disorders present
to clinicians with what appear to be legitimate somatic complaints. It is only
after repeated but fruitless workups, multiple and persistent complaints and
requests, and sometimes angry and inappropriate reactions to treatment that
clinicians begin to suspect a somatic symptom disorder. In some cases, the
manner of presentation and the symptom complex are more immediately
suggestive of a particular disorder. In any event, it is important for the
clinician to remember that the reported symptoms and complaints are quite
real and disturbing for the patient. Even after workups have indicated that
psychological factors are involved, it is never wise for a clinician to
challenge the patient or suggest that the symptoms are “all in your mind.”
The typical response to such a suggestion is for the patient to seek
additional opinions and medical tests, which in turn can perpetuate a cycle
of somatization in which underlying issues are never addressed.
Instead, the role of the clinician must be to foster a supportive,
consistent, and professional relationship with the affected individual. Such a
relationship will provide reassurance as well as protect the patient from
excessive and unnecessary medical visits and procedures. The clinician
should focus on responding to individual complaints, perhaps with periodic
but regularly scheduled appointments, and setting limits on workup and
treatment in a firm but empathic manner. This can be difficult to do when
patients become demanding and attempt to consume excessive clinic time,
but the clinician must endeavor to remain professional and to not
personalize the situation or feel as though he or she were failing the patient.
The clinician should focus on symptom reduction and rehabilitation and not
attempt to force the patient to gain insight into the potential psychological
nature of his or her symptoms.
It would obviously be hazardous for a clinician to diagnose a somatic
symptom disorder prematurely, because underlying organic pathology
might have eluded diagnosis. For example, multiple sclerosis, systemic
lupus erythematosus, and acute intermittent porphyria often have complex
presentations that elude initial diagnostic workup (Kellner 1987). Somatic
symptom and related disorders may coexist with actual disease states; for
example, many individuals with pseudoseizures also have a seizure disorder
(D’Alessio et al. 2006; Mari et al. 2006). Moene et al. (2000) found that
slightly more than 10% of patients who received an initial diagnosis of
conversion disorder actually had a true neurological disorder. At the same
time, it is important for the clinician to set limits on what he or she can offer
and to make appropriate referrals to specialists and/or mental health
clinicians.
In contrast to a primary care practitioner or a medical specialist, the
geriatric psychiatrist will play a more active role in addressing the disorder
itself rather than the actual physical complaints. Because many
presentations of somatic symptom and related disorders are chronic, the
goal of treatment is often to control symptoms rather than to cure the
patient. To facilitate this, the clinician must form a therapeutic alliance
through empathic listening and acknowledging of physical discomfort,
without trivializing the somatic complaints. An offer to review all available
medical records can sometimes be a tangible way of conveying one’s
seriousness to the patient. Educating the patient about various symptom
complexes and involving him or her in part of the decision making can be
empowering for the patient, especially a patient with chronic pain.
Individual therapy using a psychodynamic approach will focus on
helping the patient identify and then discuss psychological conflict and
associated emotions. Cognitive-behavioral therapy (CBT) will focus on
identifying distorted thought patterns and anxiety triggers and replacing
them with more realistic and adaptive strategies, as well as integrating
behavioral techniques to desensitize anxious reactions. There is an
impressive body of research supporting the efficacy of psychotherapy,
particularly CBT, for treating somatoform disorders (Kroenke 2007;
Thomson and Page 2007; Tyrer et al. 2014; Woolfolk et al. 2007).
Pharmacotherapy is a central component of treatment for somatic
symptom and related disorders. It can be targeted at a specific disorder or at
underlying anxiety and depression. Studies have documented successful
treatment of somatization disorder with either antidepressants or mood
stabilizers (García-Campayo and Sanz-Carrillo 2001). Hypochondriacal
symptoms were found to respond to a variety of antidepressant medications
—in particular, selective serotonin reuptake inhibitors—as well as to
anxiolytics (Barsky 2001; Fallon et al. 1996; Oosterbaan et al. 2001;
Somashekar et al. 2013). Antidepressants and/or anticonvulsants have long
been a mainstay of treatment for pain disorder (Eisenberg et al. 2007;
Fishbain et al. 1998; Karp and Weiner 2011; Kroenke 2007; Somashekar et
al. 2013).
The tendency of many psychiatrists to focus primarily on
pharmacotherapy can become a trap with somatic symptom and related
disorders, because the therapeutic relationship is such a key element. Given
the chronic nature of somatoform symptoms, it is unlikely that
pharmacotherapy will be a quick fix. When this narrow focus on treatment
with medications fails to result in rapid control of symptoms, the patient
may abandon the therapist for alternative treatment. Other patients may
welcome such a focus because it keeps them from having to face underlying
psychological issues. Instead, clinicians must be prepared for the long haul
and strike a balance between reasonable pharmacotherapy that targets
specific symptoms of anxiety or depression and a supportive alliance in
which the most appropriate therapy for the patient is used. If another
clinician serves as the therapist, frequent communication between
psychiatrist and therapist is necessary to coordinate treatment.
For the older patient with a somatic symptom disorder, the greatest
challenge is always trying to separate out actual medical disease from
somatic symptoms. The two are sometimes so intertwined that the line
between where one begins and the other ends cannot be reasonably
discerned without successful response of a discrete symptom to either a
medical or a psychiatric intervention. Moreover, many patients are quite
resistant to psychiatric care because they feel it delegitimizes their physical
suffering. Teamwork between internist and psychiatrist is key, allowing
both to identify the most important symptoms of concern to the patient, to
provide appropriate attention and workup, and to coordinate medical and
psychiatric interventions.
Key Points
• Somatic symptom and related disorders represent a heterogeneous group
of disorders characterized by prominent physical symptoms or
complaints that are associated with significant distress and impairment in
excess of what would be expected.
• The high degree of both medical and psychiatry comorbidity in late life
poses a unique diagnostic challenge for disorders with prominent
somatic symptoms.
• The main somatic symptom and related disorders seen in the elderly are
somatic symptom disorder, illness anxiety disorder, and conversion
disorder. Previous categories of pain disorder and undifferentiated
somatoform disorder are now subsumed within somatic symptom
disorder.
• Somatic symptom and related disorders are most commonly diagnosed
in younger individuals. Major risk factors include childhood abuse,
female gender, chronic illness or pain, lower education and
socioeconomic status, and comorbid anxiety, depression, personality
disorders, and substance abuse.
• Management of somatic symptom and related disorders requires a
consistent, empathic approach that focuses on symptomatic improvement
and rehabilitation, does not challenge the veracity of the patient’s
reports, and provides efficacious cognitive-behavioral therapy and
appropriate pharmacotherapy.
References
Aigner M, Bach M: Clinical utility of DSM-IV pain disorder. Compr
Psychiatry 40(5): 353–357, 1999 10509617
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition, DSM-5. Washington, DC, American
Psychiatric Association, 2013
Bailey PE, Henry JD: Alexithymia, somatization and negative affect in a
community sample. Psychiatry Res 150(1):13–20, 2007 17258817
Barsky AJ: Clinical practice: the patient with hypochondriasis. N Engl J
Med 345(19): 1395–1399, 2001 11794173
Barsky AJ, Klerman GL: Overview: hypochondriasis, bodily complaints,
and somatic styles. Am J Psychiatry 140(3):273–283, 1983 6338747
Behrouz R, Heriaud L, Benbadis SR: Late-onset psychogenic nonepileptic
seizures. Epilepsy Behav 8(3):649–650, 2006 16531122
Bortz JJ: Medically unexplained symptoms in older adults, in Clinical
Neurology of the Older Adult. Edited by Sirven JI, Malamut BL.
Philadelphia, PA, Lippincott Williams & Wilkins, 2008, pp 561–573
Breuer J, Freud S: Studies on hysteria (1893–1895), in The Standard
Edition of the Complete Psychological Works of Sigmund Freud, Vol 2.
Translated and edited by Strachey J. London, Hogarth, 1955, pp 1–319
Brown RJ, Schrag A, Trimble MR: Dissociation, childhood interpersonal
trauma, and family functioning in patients with somatization disorder.
Am J Psychiatry 162(5):899–905, 2005 15863791
Brown RJ, Cardeña E, Nijenhuis E, et al: Should conversion disorder be
reclassified as a dissociative disorder in DSM V? Psychosomatics
48(5):369–378, 2007 17878494
Browning M, Fletcher P, Sharpe M: Can neuroimaging help us to
understand and classify somatoform disorders? A systematic and
critical review. Psychosom Med 73(2):173–184, 2011 21217095
Chabolla DR, Krahn LE, So EL, et al: Psychogenic nonepileptic seizures.
Mayo Clin Proc 71(5):493–500, 1996 8628032
D’Alessio L, Giagante B, Oddo S, et al: Psychiatric disorders in patients
with psychogenic non-epileptic seizures, with and without comorbid
epilepsy. Seizure 15(5):333–339, 2006 16720097
Deen HG: Back and neck pain, in Clinical Neurology of the Older Adult.
Edited by Sirven JI, Malamut BL. Philadelphia, PA, Lippincott
Williams & Wilkins, 2008, pp 213–221
De Gucht V: Stability of neuroticism and alexithymia in somatization.
Compr Psychiatry 44(6):466–471, 2003 14610725
de Waal MWM, Arnold IA, Eekhof JAH, et al: Somatoform disorders in
general practice: prevalence, functional impairment and comorbidity
with anxiety and depressive disorders. Br J Psychiatry 184(6):470–476,
2004 15172939
de Waal MWM, Arnold IA, Eekhof JA, et al: Follow-up study on health
care use of patients with somatoform, anxiety and depressive disorders
in primary care. BMC Fam Pract 9:5, 2008 18218070
Egloff N, Cámara RJ, von Känel R, et al: Hypersensitivity and hyperalgesia
in somatoform pain disorders. Gen Hosp Psychiatry 36(3):284–290,
2014 24650586
Eisenberg E, River Y, Shifrin A, et al: Antiepileptic drugs in the treatment
of neuropathic pain. Drugs 67(9):1265–1289, 2007 17547471
El-Gabalawy R, Mackenzie CS, Thibodeau MA, et al: Health anxiety
disorders in older adults: conceptualizing complex conditions in late
life. Clin Psychol Rev 33(8):1096–1105, 2013 24091001
Escobar JI, Gara M, Waitzkin H, et al: DSM-IV hypochondriasis in primary
care. Gen Hosp Psychiatry 20(3):155–159, 1998 9650033
Fallon BA, Schneier FR, Marshall R, et al: The pharmacotherapy of
hypochondriasis. Psychopharmacol Bull 32(4):607–611, 1996 8993081
Fallon BA, Harper KM, Landa A, et al: Personality disorders in
hypochondriasis: prevalence and comparison with two anxiety
disorders. Psychosomatics 53(6):566–574, 2012 22658329
Faravelli C, Salvatori S, Galassi F, et al: Epidemiology of somatoform
disorders: a community survey in Florence. Soc Psychiatry Psychiatr
Epidemiol 32(1):24–29, 1997 9029984
Fink P, Hansen MS, Oxhøj ML: The prevalence of somatoform disorders
among internal medical inpatients. J Psychosom Res 56(4):413–418,
2004 15094025
Fishbain DA, Cutler RB, Rosomoff HL, et al: Do antidepressants have an
analgesic effect in psychogenic pain and somatoform pain disorder? A
meta-analysis. Psychosom Med 60(4):503–509, 1998 9710298
García-Campayo J, Sanz-Carrillo C: Gabapentin for the treatment of
patients with somatization disorder (letter). J Clin Psychiatry
62(6):474, 2001 11465526
Haftgoli N, Favrat B, Verdon F, et al: Patients presenting with somatic
complaints in general practice: depression, anxiety and somatoform
disorders are frequent and associated with psychosocial stressors. BMC
Fam Pract 11:67, 2010 20843358
Hasin D, Katz H: Somatoform and substance use disorders. Psychosom
Med 69(9):870–875, 2007 18040097
Hollifield M, Tuttle L, Paine S, et al: Hypochondriasis and somatization
related to personality and attitudes toward self. Psychosomatics
40(5):387–395, 1999 10479943
Jankovic J, Vuong KD, Thomas M: Psychogenic tremor: long-term
outcome. CNS Spectr 11(7):501–508, 2006 16816790
Karp JF, Weiner DK: Persistent pain and older adults, in Principles and
Practice of Geriatric Psychiatry, 2nd Edition. Edited by Agronin ME,
Maletta G. Philadelphia, PA, Lippincott Williams & Wilkins 2011, pp
763–782
Kellner R: Hypochondriasis and somatization. JAMA 258(19):2718–2722,
1987 3312664
Klengel T, Heck A, Pfister H, et al: Somatization in major depression—
clinical features and genetic associations. Acta Psychiatr Scand
124(4):317–328, 2011 21838737
Krautwurst S, Gerlach AL, Gomille L, et al: Health anxiety—an indicator
of higher interoceptive sensitivity? J Behav Ther Exp Psychiatry
45(2):303–309, 2014 24584036
Krem MM: Motor conversion disorders reviewed from a neuropsychiatric
perspective. J Clin Psychiatry 65(6):783–790, 2004 15291655
Kroenke K: Efficacy of treatment for somatoform disorders: a review of
randomized controlled trials. Psychosom Med 69(9): 881–888, 2007
18040099
Kushwaha V, Sinha Deb K, Chadda RK, et al: A study of disability and its
correlates in somatization disorder. Asian J Psychiatr 8:56–58, 2014
24655628
Leveille SG, Ling S, Hochberg MC, et al: Widespread musculoskeletal pain
and the progression of disability in older disabled women. Ann Intern
Med 135(12):1038–1046, 2001 11747382
Liskow B, Othmer E, Penick EC, et al: Is Briquet’s syndrome a
heterogeneous disorder? Am J Psychiatry 143(5):626–629, 1986
3963251
Lundh LG, Simonsson-Sarnecki M: Alexithymia, emotion, and somatic
complaints. J Pers 69(3):483–510, 2001 11478734
Mari F, Di Bonaventura C, Vanacore N, et al: Video-EEG study of
psychogenic non-epileptic seizures: differential characteristics in
patients with and without epilepsy. Epilepsia 47(suppl 5):64–67, 2006
17239109
Miller AR, North CS, Clouse RE, et al: The association of irritable bowel
syndrome and somatization disorder. Ann Clin Psychiatry 13(1):25–30,
2001 11465682
Moene FC, Landberg EH, Hoogduin KA, et al: Organic syndromes
diagnosed as conversion disorder: identification and frequency in a
study of 85 patients. J Psychosom Res 49(1):7–12, 2000 11053598
Ness D: Physical therapy management for conversion disorder: case series.
J Neurol Phys Ther 31(1):30–39, 2007 17419887
Noyes RJr, Langbehn DR, Happel RL, et al: Personality dysfunction among
somatizing patients. Psychosomatics 42(4):320–329, 2001 11496021
Oosterbaan DB, van Balkom AJ, van Boeijen CA, et al: An open study of
paroxetine in hypochondriasis. Prog Neuropsychopharmacol Biol
Psychiatry 25(5):1023–1033, 2001 11444675
O’Sullivan SS, Spillane JE, McMahon EM, et al: Clinical characteristics
and outcome of patients diagnosed with psychogenic nonepileptic
seizures: a 5-year review. Epilepsy Behav 11(1):77–84, 2007 17517535
Otis JAD, McGeeney B: Managing pain in the elderly. Clin Geriatr 8:48–
62, 2000
Pribor EF, Smith DS, Yutzy SH: Somatization disorder in elderly patients.
Am J Geriatr Psychiatry 2:109–117, 1994
Puri PR, Dimsdale JE: Health care utilization and poor reassurance:
potential predictors of somatoform disorders. Psychiatr Clin North Am
34(3):525–544, 2011 21889677
Rabinowitz T, Laek J: An approach to the patient with physical complaints
or irrational anxiety about an illness or their appearance, in The 10-
Minute Guide to Psychiatric Diagnosis and Treatment. Edited by Stern
TA. New York, Professional Publishing Group, Ltd, 2005, pp 225–238
Rabinowitz T, Hirdes JP, Desjardins I: Somatoform disorders in late life, in
Principles and Practice of Geriatric Psychiatry, 2nd Edition. Edited by
Agronin ME, Maletta G. Philadelphia, PA, Lippincott Williams &
Wilkins 2011, pp 565–581
Rief W, Hessel A, Braehler E: Somatization symptoms and
hypochondriacal features in the general population. Psychosom Med
63(4):595–602, 2001 11485113
Ritsner M, Ponizovsky A, Kurs R, et al: Somatization in an immigrant
population in Israel: a community survey of prevalence, risk factors,
and help-seeking behavior. Am J Psychiatry 157(3):385–392, 2000
10698814
Roelofs K, Keijsers GP, Hoogduin KA, et al: Childhood abuse in patients
with conversion disorder. Am J Psychiatry 159(11): 1908–1913, 2002
12411227
Sack M, Lahmann C, Jaeger B, et al: Trauma prevalence and somatoform
symptoms: are there specific somatoform symptoms related to
traumatic experiences? J Nerv Ment Dis 195(11):928–933, 2007
18000455
Samelius L, Wijma B, Wingren G, et al: Somatization in abused women. J
Womens Health (Larchmt) 16(6):909–918, 2007 17678462
Sar V, Akyüz G, Kundakçi T, et al: Childhood trauma, dissociation, and
psychiatric comorbidity in patients with conversion disorder. Am J
Psychiatry 161(12):2271–2276, 2004 15569899
Sawyer P, Lillis JP, Bodner EV, et al: Substantial daily pain among nursing
home residents. J Am Med Dir Assoc 8(3):158–165, 2007 17349944
Scherder EJ, Plooij B: Assessment and management of pain, with particular
emphasis on central neuropathic pain, in moderate to severe dementia.
Drugs Aging 29(9):701–706, 2012 23018606
Scherder E, Bouma A, Slaets J, et al: Repeated pain assessment in
Alzheimer’s disease. Dement Geriatr Cogn Disord 12(6):400–407,
2001 11598312
Scudds RJ, Ostbye T: Pain and pain-related interference with function in
older Canadians: the Canadian Study of Health and Aging. Disabil
Rehabil 23(15):654–664, 2001 11720115
Sheehan B, Banerjee S: Review: Somatization in the elderly. Int J Geriatr
Psychiatry 14(12):1044–1049, 1999 10607972
Simon GE, Gureje O, Fullerton C: Course of hypochondriasis in an
international primary care study. Gen Hosp Psychiatry 23(2):51–55,
2001 11313070
Smith RC, Gardiner JC, Lyles JS, et al: Exploration of DSM-IV criteria in
primary care patients with medically unexplained symptoms.
Psychosom Med 67(1):123–129, 2005 15673634
Somashekar B, Jainer A, Wuntakal B: Psychopharmacotherapy of somatic
symptoms disorders. Int Rev Psychiatry 25(1):107–115, 2013
23383672
Stone J, Zeman A, Simonotto E, et al: fMRI in patients with motor
conversion symptoms and controls with simulated weakness.
Psychosom Med 69(9):961–969, 2007 17991812
Stuart S, Noyes R Jr: Attachment and interpersonal communication in
somatization. Psychosomatics 40(1):34–43, 1999 9989119
Thomson AB, Page LA: Psychotherapies for hypochondriasis. Cochrane
Database Syst Rev (4):CD006520, 2007 17943915
Tomenson B, McBeth J, Chew-Graham CA, et al: Somatization and health
anxiety as predictors of health care use. Psychosom Med 74(6):656–
664, 2012 22753632
Tyrer P, Cooper S, Salkovskis P, et al: Clinical and cost-effectiveness of
cognitive behaviour therapy for health anxiety in medical patients: a
multicentre randomised controlled trial. Lancet 383(9913):219–225,
2014 24139977
Voigt K, Wollburg E, Weinmann N, et al: Predictive validity and clinical
utility of DSM-5 Somatic Symptom Disorder—comparison with DSM-
IV somatoform disorders and additional criteria for consideration. J
Psychosom Res 73(5):345–350, 2012 23062807
Vuilleumier P: Hysterical conversion and brain function. Prog Brain Res
150:309–329, 2005 16186033
Vuilleumier P, Chicherio C, Assal F, et al: Functional neuroanatomical
correlates of hysterical sensorimotor loss. Brain 124(Pt 6):1077–1090,
2001 11353724
Waldinger RJ, Schulz MS, Barsky AJ, et al: Mapping the road from
childhood trauma to adult somatization: the role of attachment.
Psychosom Med 68(1):129–135, 2006 16449423
Waller E, Scheidt CE: Somatoform disorders as disorders of affect
regulation: a study comparing the TAS-20 with non-self-report
measures of alexithymia. J Psychosom Res 57(3):239–247, 2004
15507250
Woolfolk RL, Allen LA, Tiu JE: New directions in the treatment of
somatization. Psychiatr Clin North Am 30(4):621–644, 2007 17938037
Yazici KM, Kostakoglu L: Cerebral blood flow changes in patients with
conversion disorder. Psychiatry Res 83(3):163–168, 1998 9849725
Yoshimasu K: Substance-related disorders and somatic symptoms: how
should clinicians understand the associations? Curr Drug Abuse Rev
5(4):291–303, 2012 23244342
Suggested Readings
El-Gabalawy R, Mackenzie CS, Thibodeau MA, et al: Health anxiety
disorders in older adults: conceptualizing complex conditions in late
life. Clin Psychol Rev 33(8):1096–1105, 2013
Kroenke K: Efficacy of treatment for somatoform disorders: a review of
randomized controlled trials. Psychosom Med 69(9): 881–888, 2007
Puri PR, Dimsdale JE: Health care utilization and poor reassurance:
potential predictors of somatoform disorders. Psychiatr Clin North Am
34(3):525–544, 2011
Rabinowitz T, Laek J: An approach to the patient with physical complaints
or irrational anxiety about an illness or their appearance, in The 10-
Minute Guide to Psychiatric Diagnosis and Treatment. Edited by Stern
TA. New York, Professional Publishing Group, Ltd, 2005, pp 225–238
Somashekar B, Jainer A, Wuntakal B: Psychopharmacotherapy of somatic
symptoms disorders. Int Rev Psychiatry 25(1):107–115, 2013
CHAPTER 14
People are living longer and healthier lives, and many expect sexuality to
continue to play an important role. As a result, sexual issues and disorders
are an important part of assessment and treatment by the geriatric
psychiatrist, in both outpatient and long-term-care settings. The idea of
sexuality in late life, once commonly regarded with denial, humor, or even
disgust, too often led clinicians to view sexual dysfunction as a normal and
untreatable part of aging. Such distorted attitudes have changed widely,
however, starting with the sexual and feminist revolutions in the 1960s and
1970s and buoyed by the widespread use of hormone replacement therapy
and then the advent of oral erectogenic agents for erectile dysfunction. This
widespread openness toward late-life sexuality coupled with reliable
treatments for sexual dysfunction has made sexuality a more common and
comfortable topic of conversation among aged individuals and their
clinicians, and can now ensure the persistence of enjoyable sexual function
in later years. In addition, the destigmatization of sexual dysfunction has no
doubt encouraged many older couples to seek treatment who otherwise
might have suffered in silence and shame.
Compared with women, the sexual changes in aging men occur more
gradually with a less predictable time frame (Morley 2003; Westheimer and
Lopater 2002). As men age, desire may involve less anticipatory physical
arousal, and sexual arousal and orgasm may take longer to achieve. Older
men require more physical stimulation to achieve erections, which tend to
be less frequent, less durable, and less reliable. The volume of ejaculate
during orgasm is decreased. In older men, the resolution or refractory stage
is much longer, lasting hours to days instead of minutes to hours as in
younger men. Testosterone levels in men decline 35% on average by age 80
years, although some men have more significant declines, with levels
dropping below 200 ng/dL, the level at which they are diagnosed with
hypogonadism (Morley 2003). Some researchers have suggested the
existence of a male menopause or andropause resulting from declining
testosterone levels and involving a symptom complex that includes
decreased libido and sexual function; diminished bone and muscle mass,
muscle power, and body hair; and decreased lean body mass (Haider et al.
2014; Heaton and Morales 2001; Morley and Perry 2003; Pines 2011).
Research has suggested, however, that these changes are quite variable, and
that testosterone replacement therapy has inconsistent results (Harman
2005).
In both sexes, the effects of physiological changes in sexual function are
mediated by a number of psychosocial factors. The more an individual
knows about what constitutes normal age-associated changes in sexual
function, the easier it may be for him or her to accept these changes. For
example, a man who does not understand the normal changes in erectile
function may misinterpret them and believe that he has a sexual problem.
Similarly, a woman may misinterpret vaginal dryness as an indication that
she does not want to have sex. Such overreactions to normal changes can
lead an individual to engage in less frequent or more limited sexual activity.
In addition, some older individuals may accept ageist stereotypes about
sexuality and view their behaviors as inappropriate or potentially harmful,
despite the individuals’ relatively normal sexual desire and capacity. Other
individuals may lose self-confidence and feel less sexy, especially as they
struggle to cope with age-associated changes in physical appearance,
strength, and endurance. Such attitudinal barriers may be more damaging to
sexuality than actual physiological changes.
The quality of an individual’s relationship with a partner is also
influential. Couples often have to adapt sexual technique and spend more
time on foreplay to preserve previous levels of sexual function and
enjoyment. Partners who are unable to work together may experience
difficulty with sex and perhaps even sexual dysfunction. On the other hand,
aging can bring new possibilities for sexuality in later life. Partners may
have more time to spend with each other once children have left home or
during retirement. For postmenopausal women, sex may be associated with
a reduced level of anxiety because of the impossibility of pregnancy.
Sexually Transmitted Diseases
None of the published surveys about late-life sexuality asked respondents
about sexually transmitted diseases (STDs), even though older people are
certainly at risk for contracting them. According to surveillance data from
the Centers for Disease Control and Prevention (2012), rates of STDs
including chlamydia, gonorrhea, and syphilis in individuals ages 65 years
and older were the lowest of any group and had not changed appreciably in
the previous 5 years. In a study from Washington State, the most common
STDs in older individuals were nongonococcal urethritis in men and genital
herpes in women—representing 1.3% of all reported cases of STDs (Xu et
al. 2001). With respect to HIV and AIDS, the largest increase in individuals
living with HIV infection from 2008 to 2010 was among individuals older
than 65 years, with a rate at the end of 2010 of 85.7 per 100,000 persons in
the United States. Although rates of new infections among the elderly have
been decreasing, rates of stage 3 AIDS and death have increased (Centers
for Disease Control and Prevention 2011). It should be noted that not all of
these cases are due to sexual transmission.
Despite these low prevalence rates for STDs, older individuals remain at
risk because they continue to be sexually active. Adding to this risk is the
fact that many older people never received the sex education provided to
today’s younger population. Thus, they may neglect safe-sex practices due
to lack of knowledge, the absence of pregnancy risk, and a false sense of
safety from knowing that STDs are more prevalent in younger people. One
survey of a representative sample of 1,670 individuals 45 years and older
found that less than 20% of men and women who were single and dating
reported using a condom or any other form of protection on a regular basis
during sexual intercourse (AARP 2010). Given these lapses in safe-sex
measures, education about sexuality, STDs, and safe-sex practices remains
critical throughout the entire adult life cycle.
Assessment
The assessment of sexual dysfunction in late life involves identifying the
specific problem and then obtaining a comprehensive medical, psychiatric,
and sexual history to determine potential causes. A comprehensive sexual
history involves asking an individual about prior sexual experiences,
current sexual functioning, and attitudes toward sexuality and toward any
current partner. With older couples, interviewers must be able to identify
relevant age-appropriate issues (Agronin 2014; Agronin and Westheimer
2011). It is important to balance the need to gather sexual history with the
responsibility to be sensitive to the fact that sexual data may be some of the
most personal information that a patient will ever divulge. Finally, accurate
assessment of sexual dysfunction in late life depends to a large degree on a
comfortable and productive doctor-patient relationship, one in which the
patient and his or her partner feel secure enough to disclose adequate
history and the physician asks the right questions and has sufficient testing
performed. Partner involvement is crucial to a successful outcome.
The medical workup for sexual dysfunction may involve a physical
examination, laboratory testing, and specialized diagnostic testing. The
focus of the physical examination is on genital and urological anatomy and
function, including underlying vascular and neurological function.
Laboratory testing typically involves examination of routine blood
chemistry (e.g., blood count, electrolyte levels, glucose levels, lipid profile),
testosterone and prolactin levels, thyroid function, and, in men, prostate-
specific antigen levels. Specialized diagnostic tests for ED may include
nocturnal penile tumescence and rigidity testing (to determine whether
natural erections occur during sleep) and penile duplex ultrasonography (to
assess blood flow in the penis). This workup is typically conducted by a
urologist.
Treatment
Preservation and enhancement of sexual activity in geriatric patients require
recognition of and sensitivity to the fact that many of these individuals want
and intend to continue having sex, despite changes in physical and sexual
function. Once an evaluation is complete, both partners should be educated
about normal and dysfunctional sexuality (Bitzer et al. 2008). This
information helps to reassure the affected individual that he or she is not the
only person with the particular problem, that the problem has specific
causes, and that it can be treated. In addition, clinicians can help patients
recognize sexuality as a form of physical and psychological intimacy and
not solely as sexual intercourse. This discussion will build trust between the
patient and the clinician, and will help the patient feel comfortable about
seeking follow-up and being open about emotional reactions to the problem.
Many treatments fail at this point, not because the treatments cannot work
but because the patient and the clinician never establish a solid working
relationship. Treatment can also fail when one partner refuses to cooperate
with treatment or when problems within the couple’s entire relationship
become insurmountable.
Unique challenges are faced by couples in which one or both partners
have a chronic medical illness or disability. These couples often need to
shift their focus from intercourse to foreplay and to adapt sexual practices
to account for physical limitations such as fatigue, loss of muscle strength,
and pain (Agronin and Westheimer 2011; Morley and Tariq 2003; Schover
and Jensen 1988). Education is key. Organizations such as the American
Cancer Society, the United Ostomy Associations of America, National
Jewish Health (on respiratory disorders), and others have developed helpful
Internet guides to maintaining sexual function despite specific medical
illnesses. Physicians should work to maximize both rehabilitative and
palliative treatments—for example, making use of analgesics for pain,
inhalers for shortness of breath, or physical therapy for joint immobility and
muscle weakness. In addition, appropriate treatment of depression, anxiety,
or psychosis can often lead to significant improvement in sexual function,
assuming that the medications used to treat these disorders do not
themselves cause problems. Some ways in which an older couple can
enhance sexual function and cope with disability are outlined in Table 14–5.
Medication Effects
When medication side effects impair sexual function, physicians can
consider several options (Goodwin and Agronin 1997; Labbate et al. 2003;
Zajecka 2003). The first step is to continue administering the medication
and wait for tolerance to develop; many side effects diminish or disappear
after several weeks. If no change occurs, dose reduction can be tried.
Simplifying the overall regimen might also be helpful, given that
combinations of medications can cause more sexual side effects than each
medication alone. For certain medications, such as antidepressants that have
short half-lives, a drug holiday in which administration of the medication is
temporarily stopped for a day or two (such as for a weekend) can result in
transient improvement in sexual function (Rothschild 1995). However,
there is a risk of recurrence of psychiatric symptoms during this holiday.
Ultimately, the clinician may have to consider replacing the medication
with an agent that has less potential for sexual side effects, such as
bupropion or mirtazapine (Clayton et al. 2014; Reichenpfader et al. 2014).
With regard to antipsychotic medications, more potent agents with fewer
anticholinergic side effects and prolactin-sparing agents may cause less
dysfunction (Baggaley 2008; Serretti and Chiesa 2011).
When sexual dysfunction is due to antidepressant medication, the
clinician can also consider using antidotes to reverse sexual side effects
(Taylor et al. 2013; Thomas 2003; Zajecka 2003). Antidotes include
yohimbine, amantadine, cyproheptadine (a note of caution: cyproheptadine
can also reverse the antidepressant effects of SSRIs), bethanechol,
methylphenidate, buspirone, bromocriptine (for antipsychotic-induced
sexual dysfunction), and the antidepressants bupropion, nefazodone,
mirtazapine, and trazodone. The oral erectogenic agents sildenafil, tadalafil,
and vardenafil have also been shown to reverse antidepressant-induced ED
(Berigan 2004; Fava et al. 2006;Segraves et al. 2007; Taylor et al. 2013).
Depending on the chosen antidote for sexual side effects, the patient can
take a dose anywhere from 30 to 60 minutes before anticipated sex and can
take increasing doses until success is achieved. If intermittent use of an
antidote does not work, a regularly scheduled daily dose should be
considered.
If none of these strategies work, the clinician must consider the trade-off
between the benefits of the original medication and the sexual side effects.
For some individuals, stopping the medication poses too great a risk of
recurrent psychiatric symptoms, and adequate alternatives may not exist. In
this frustrating situation, patients must choose either to discontinue a
needed medication or to cope with persistent sexual dysfunction.
Sex Therapy
In some older couples, sexual dysfunction has clear psychological roots; for
example, sexual dysfunction often occurs in the context of a dysfunctional
relationship. Sex therapy is always best done conjointly, where both
partners participate because both are an integral part of the problem and
solution. Historically, a psychodynamic model was used in sex therapy to
uncover underlying unconscious conflicts, but that approach is now viewed
as less successful, and cognitive-behavioral techniques are used in current
treatment models (Brotto and Luria 2014; Kaplan 1983, 1974; Westheimer
and Lopater 2002). Brief supportive and educational counseling is a first
step in treatment and can help dispel distorted and uninformed attitudes
toward sexuality in general and toward a sexual problem in particular.
Counseling can also help an individual or couple change sexual practices to
resolve a problem. In other cases, more intensive couples therapy is needed
to resolve long-standing relationship issues before work on a sexual
problem can begin.
Sex therapy involves both cognitive and behavioral techniques, with an
overall goal of building an association between relaxed and sensual
physical intimacy and sexual relations. The same principles can be applied
across the life span, with several refinements in late life. Using cognitive
therapy techniques, the therapist attempts to change distorted cognitive
attitudes toward sexual activity into more practical attitudes. For example,
many men with ED find it difficult not to assume the role of a spectator
during sex—that is, not to watch themselves with their partners and be
preoccupied with the status of their erections. This spectator role can
increase anxiety and distract the man from concentrating on pleasurable
sensations, with the result that ED is reinforced (Masters and Johnson
1970). To counter this, the man is taught to shift his mental focus from his
erection to pleasurable aspects of the encounter (Kaplan 1974). ED may
also be perpetuated by cognitive distortions such as catastrophizing, in
which the man thinks that if he does not achieve an erection during sex, he
will be rejected not only by his partner but by all women. Another common
cognitive distortion is all-or-nothing thinking, in which the man thinks that
he must achieve an instant erection during sex or else the whole thing is
pointless. The problem with such unrealistic cognitive distortions is that
they often become self-fulfilling prophesies. The therapist helps the patient
to gain insight into the negative effects of such thoughts and then to practice
replacing them with more realistic and hopeful ones, sometimes even with
positive assertions or affirmations of success (Goodwin and Agronin 1997).
Behavioral techniques used during sex therapy begin with exercises
called sensate focus, in which a couple practices physical relaxation
techniques during nonpressured sensual touching. Sensate focus helps to
reduce performance anxiety and restore the natural flow of the sexual
response cycle. Once the partners are able to feel relaxed and physically
intimate together without sexual stimulation, they gradually progress to
genital stimulation and then intercourse. Several adjustments in these
exercises may be required for the older couple. For example, older patients
with physical problems that involve some degree of disability may express
concerns about being able to exert themselves adequately during sexual
activity. The therapist might recommend one of several positions that
minimize exertion, such as lying side by side or having one partner kneel on
pillows and support himself or herself on a low bed. Other suggestions
outlined in Table 14–5 might also apply. Such simple suggestions may
remove some of the most anxiety-provoking barriers for an older couple,
especially the common but unfounded belief that older persons lack the
stamina or dexterity for sexual activity.
During sex therapy, the therapist continues to work with the couple on
their relationship and tries to identify and confront resistance that inevitably
arises during treatment. Such resistance to these seemingly innocuous
exercises often reveals key problems in the relationship that are either
causing the sexual dysfunction or impeding its treatment. Regardless of age,
many couples find that sexual interest and pleasure reemerge and sexual
function improves during sex therapy, allowing them to enjoy once again
such a fundamental component of their relationship.
Erectile Disorder
ED, the most common sexual dysfunction in older men, historically was
seen as a psychological problem. More recent data, however, indicate that
ED is primarily caused by a problem with erectile physiology in the
majority of cases (Ludwig and Phillips 2014; Tariq et al. 2003). There are,
however, important psychological components of ED in terms of both cause
and effect. Many men equate erections with masculinity, potency, and
vitality. As a result, ED in late life is often experienced by men as a
harbinger of physical and sexual decline. Performance anxiety, stress,
depression, and relationship problems can trigger or exacerbate ED. In turn,
ED is associated with feelings of anger, anxiety, powerlessness, shame, and
humiliation in front of one’s partner. Recurrent ED can lead to depression.
Treatment of ED in geriatric patients involves the same approaches as
those used in younger men and has been revolutionized with the advent of
oral erectogenic agents. For men with hypogonadism as the likely cause of
ED, testosterone replacement therapy—in the form of a pill, transdermal gel
or patch, intramuscular injection, or subcutaneous implant—may be helpful
(Howell and Shalet 2001; Jacob 2011; Morley 2003). This treatment should
be avoided in men with a history of prostate or bladder cancer or with
bladder outlet obstruction. Some men have ED as a result of vascular
damage and may benefit from microsurgical revascularization. Peyronie’s
disease, characterized by scarring-caused curvature of the penis during
erection, can also be treated, with resultant improvement in erectile
function.
For many years, the only viable treatments for chronic ED involved
either the use of a vacuum constriction device prior to intercourse or the
surgical placement of a penile prosthetic device (Dutta and Eid 1999; Sison
et al. 1997). Initial pharmacological approaches to ED involved either
penile intracavernosal injection or urethral suppositories using alprostadil, a
synthetic form of prostaglandin E1, which worked by increasing smooth
muscle relaxation and arterial dilatation in the penis (Althof 1995; Padma-
Nathan et al. 1997). All of these approaches have given way, however, to
the use of the oral erectogenic agents sildenafil, tadalafil, and vardenafil.
These agents improve erectile function in men with both organic and
psychogenic ED by serving as selective inhibitors of phosphodiesterase
type 5 (PDE5). Sildenafil and vardenafil can be taken 30 minutes to 4 hours
before anticipated sexual activity, and tadalafil can be taken up to 30 hours
prior. Erections do not occur spontaneously on taking these medications but
require adequate physical stimulation. The obvious advantages of PDE5
inhibitors are ease of use and success in up to 70%–80% of affected men
(Boolell et al. 1996; Porst et al. 2003a, 2003b). Potential side effects include
headache, skin flushing, dizziness, gastrointestinal discomfort, blurred
vision, and the potential for blood pressure decreases when combined with
nitrates (e.g., sublingual nitroglycerin, isosorbide). In addition, the PDE5
inhibitors should be used with caution in men with abnormal penile shape, a
history of orthostatic hypotension, severe renal or hepatic disease,
concomitant use of certain antiviral and antifungal medications, and
diseases that increase the risk of priapism, such as sickle cell anemia,
multiple myeloma, and leukemia. An extremely rare but potentially
devastating side effect of PDE5 inhibitors that has emerged is nonarteritic
anterior ischemic optic neuropathy, or NOIAN (the acronym for the French
name for the condition), characterized by the rapid onset of visual loss.
Although the exact role of PDE5 inhibitors in the pathogenesis of NOIAN
has not been fully established, any changes in visual acuity while taking a
PDE5 inhibitor require immediate assessment (Bella et al. 2006).
Key Points
• Because individuals are living longer, healthier lives, sexuality continues
to play an important role, facilitated by increasingly positive attitudes and
newer and more effective treatments for sexual dysfunction.
• Sexual surveys indicate that although a majority of individuals ages 65
years and older continue to be sexually active, there are declines in both
the rate and the frequency of sexual activity, particularly in older single
women.
• The main predictors of sexual activity in late life include previous sexual
behaviors; the availability, health, and interest of a partner; and the
individual’s overall physical health.
• Rates of sexual dysfunction increase with age, with erectile disorder being
the most common disorder in older men and sexual interest/arousal
disorder being the most common disorder in older women.
• Depending on its form, sexual dysfunction in late life can be treated with a
variety of approaches, including treatment of causative medical or
medication-related factors, psychoeducation, individual or couples
counseling, sex therapy, and the use of disorder-specific medications, such
as oral erectogenic agents for erectile dysfunction.
References
AARP: Sexuality at Midlife and Beyond: 2004 Update of Attitudes and
Behaviors, 2005. Available at:
http://assets.aarp.org/rgcenter/general/2004_sexuality.pdf. Accessed
December 8, 2014.
AARP: Sex, Romance, and Relationships. AARP Survey of Midlife and
Older Adults (publication D19234). Washington, DC, AARP, April
2010. Available at: http://assets.aarp.org/rgcenter/general/srr_09.pdf.
Accessed December 1, 2014.
Adelman M: Stigma, gay lifestyles, and adjustment to aging: a study of
later-life gay men and lesbians. J Homosex 20(3–4):7–32, 1990
2086652
Agronin ME: Sexuality and aging, in Principles and Practice of Sex
Therapy, 5th Edition. Edited by Binik YM, Hall KSK. New York,
Guilford, 2014, pp 525–539
Agronin ME, Westheimer RK: Sexuality and sexual disorders in late life, in
Principles and Practice of Geriatric Psychiatry, 2nd Edition. Edited by
Agronin ME, Maletta G. Philadelphia, PA, Lippincott, Williams &
Wilkins 2011, pp 603–625
Alexander JL, Kotz K, Dennerstein L, et al: The effects of postmenopausal
hormone therapies on female sexual functioning: a review of double-
blind, randomized controlled trials. Menopause 11(6 Pt 2):749–765,
2004 15543027
Althof SE: Pharmacologic treatment of rapid ejaculation. Psychiatr Clin
North Am 18(1):85–94, 1995 7761310
Althof SE: Treatment of premature ejaculation: psychotherapy,
pharmacotherapy and combined therapy, in Principles and Practice of
Sex Therapy, 5th Edition. Edited by Binik YM, Hall KSK. New York,
Guilford, 2014, pp 112–137
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition, DSM-5. Washington, DC, 2013
Aversa A, Francomano D, Bruzziches R, et al: Is there a role for
phosphodiesterase type-5 inhibitors in the treatment of premature
ejaculation? Int J Impot Res 23(1):17–23, 2011 21270821
Baggaley M: Sexual dysfunction in schizophrenia: focus on recent
evidence. Hum Psychopharmacol 23(3):201–209, 2008 18338766
Basson R: Androgen replacement for women. Can Fam Physician 45:2100–
2107, 1999 10509222
Bella AJ, Brant WO, Lue TF, et al: Non-arteritic anterior ischemic optic
neuropathy (NAION) and phosphodiesterase type-5 inhibitors. Can J
Urol 13(5):3233–3238, 2006 17076943
Berigan T: Antidepressant-induced sexual dysfunction treated with
vardenafil. Can J Psychiatry 49(9):643, 2004 15503738
Bitzer J, Platano G, Tschudin S, et al: Sexual counseling in elderly couples.
J Sex Med 5(9):2027–2043, 2008 18637999
Boolell M, Gepi-Attee S, Gingell JC, et al: Sildenafil, a novel effective oral
therapy for male erectile dysfunction. Br J Urol 78(2): 257–261, 1996
8813924
Brotto L, Luria M: Sexual interest/arousal disorder in woman, in Principles
and Practice of Sex Therapy, 5th Edition. Edited by Binik YM, Hall
KSK. New York, Guilford, 2014, pp 17–41
Burns A, Jacoby R, Levy R: Psychiatric phenomena in Alzheimer’s disease,
IV: disorders of behaviour. Br J Psychiatry 157:86–94, 1990 2397368
Buster JE, Kingsberg SA, Aguirre O, et al: Testosterone patch for low
sexual desire in surgically menopausal women: a randomized trial.
Obstet Gynecol 105(5 Pt 1): 944–952, 2005 15863529
Centers for Disease Control and Prevention: HIV Surveillance Report,
2011. Available at:
http://www.cdc.gov/hiv/topics/surveillance/resources/reports/.
Accessed April 2014.
Centers for Disease Control and Prevention: Sexually Transmitted Disease
Surveillance, 2012. Available at:
http://www.cdc.gov/sTD/stats12/Surv2012.pdf. Accessed April 2014.
Clayton AH, Croft HA, Handiwala L: Antidepressants and sexual
dysfunction: mechanisms and clinical implications. Postgrad Med
126(2):91–99, 2014 24685972
Crenshaw TL, Goldberg JP: Sexual Pharmacology: Drugs That Affect
Sexual Function. New York, WW Norton, 1996
Davies HD, Zeiss A, Tinklenberg JR: ’Til death do us part: intimacy and
sexuality in the marriages of Alzheimer’s patients. J Psychosoc Nurs
Ment Health Serv 30(11):5–10, 1992 1494148
Davies HD, Newkirk LA, Pitts CB, et al: The impact of dementia and mild
memory impairment (MMI) on intimacy and sexuality in spousal
relationships. Int Psychogeriatr 22(4):618–628, 2010 20226112
Davis SR, Moreau M, Kroll R, et al: Testosterone for low libido in
postmenopausal women not taking estrogen. N Engl J Med
359(19):2005–2017, 2008 18987368
Dennerstein L, Guthrie JR, Hayes RD, et al: Sexual function, dysfunction,
and sexual distress in a prospective, population-based sample of mid-
aged, Australian-born women. J Sex Med 5(10):2291–2299, 2008
18638008
Dutta TC, Eid JF: Vacuum constriction devices for erectile dysfunction: a
long-term, prospective study of patients with mild, moderate, and
severe dysfunction. Urology 54(5):891–893, 1999 10565753
Eloniemi-Sulkava U, Notkola IL, Hämäläinen K, et al: Spouse caregivers’
perceptions of influence of dementia on marriage. Int Psychogeriatr
14(1):47–58, 2002 12094907
Fava M, Nurnberg HG, Seidman SN, et al: Efficacy and safety of sildenafil
in men with serotonergic antidepressant-associated erectile
dysfunction: results from a randomized, double-blind, placebo-
controlled trial. J Clin Psychiatry 67(2):240–246, 2006 16566619
Feldman HA, Goldstein I, Hatzichristou DG, et al: Impotence and its
medical and psychosocial correlates: results of the Massachusetts Male
Aging Study. J Urol 151(1):54–61, 1994 8254833
Fisher LL: Sex, Romance, and Relationships: AARP Survey of Midlife and
Older Adults. Publication D19234. Washington, DC, AARP, 2010
Goodwin AJ, Agronin ME: A Women’s Guide to Overcoming Sexual Fear
and Pain. Oakland, CA, New Harbinger, 1997
Graham CA: Orgasm disorders in woman, in Principles and Practice of Sex
Therapy, 5th Edition. Edited by Binik YM, Hall KSK. New York,
Guilford, 2014, pp 89–111
Guay DR: Inappropriate sexual behaviors in cognitively impaired older
individuals. Am J Geriatr Pharmacother 6(5):269–288, 2008 19161930
Haider A, Meergans U, Traish A, et al: Progressive improvement of T-
scores in men with osteoporosis and subnormal serum testosterone
levels upon treatment with testosterone over six years. Int J Endocrinol
2014:496948, 2014 24688541
Hajjar RR, Kamel HK: Sexuality in the nursing home, part 1: attitudes and
barriers to sexual expression. J Am Med Dir Assoc 5(2 suppl):S42–
S47, 2004a 14984610
Hajjar RR, Kamel HK: Sexuality in the nursing home, part 2: managing
abnormal behavior—legal and ethical issues. J Am Med Dir Assoc 5 (2
suppl):S48–S52, 2004b 14984611
Hanks N: The effects of Alzheimer’s disease on the sexual attitudes and
behaviors of married caregivers and their spouses. Sex Disabil
10(3):137–151, 1992
Harman SM: Testosterone in older men after the Institute of Medicine
Report: where do we go from here? Climacteric 8(2):124–135, 2005
16096168
Hashmi FH, Krady AI, Qayum F, et al: Sexually disinhibited behavior in
the cognitively impaired elderly. Clin Geriatr 8:61–68, 2000
Heaton JP, Morales A: Andropause: a multi-system disease. Can J Urol
8(2):1213–1222, 2001 11375784
Heiman L, Lopiccolo J: Becoming Orgasmic: A Sexual and Personal
Growth Program for Women, Revised Edition. New York, Prentice-
Hall, 1976
Howell S, Shalet S: Testosterone deficiency and replacement. Horm Res 56
(suppl 1): 86–92, 2001 11786693
Jacob BC: Testosterone replacement therapy in males with erectile
dysfunction. J Pharm Pract 24(3):298–306, 2011 21676853
Jacoby S: Great sex: what’s age got to do with it? Modern Maturity
Sept/Oct:43–48, 1999
Joller P, Gupta N, Seitz DP, et al: Approach to inappropriate sexual
behaviour in people with dementia. Can Fam Physician 59(3):255–260,
2013 23486794
Kaas MJ: Sexual expression of the elderly in nursing homes. Gerontologist
18(4):372–378, 1978 263483
Kaplan HS: The New Sex Therapy. New York, Brunner/Mazel, 1974
Kaplan HS: The Evaluation of Sexual Disorders: Psychological and
Medical Aspects. New York, Brunner/Mazel, 1983
Kaplan HS: Sexual Aversion, Sexual Phobias, and Panic Disorder. New
York, Brunner/Mazel, 1987
Kaplan HS: Overcoming Premature Ejaculation. New York, Brunner/Mazel,
1989
Kavoussi RJ, Segraves RT, Hughes AR, et al: Double-blind comparison of
bupropion sustained release and sertraline in depressed outpatients. J
Clin Psychiatry 58(12):532–537, 1997 9448656
Kimmel DC: Psychotherapy and the older gay man. Psychotherapy: Theory,
Research & Practice 14(4):386–393, 1977
Kingsberg S, Shifren J, Wekselman K, et al: Evaluation of the clinical
relevance of benefits associated with transdermal testosterone
treatment in postmenopausal women with hypoactive sexual desire
disorder. J Sex Med 4(4 Pt 1):1001–1008, 2007 17627745
Kumar A, Koss E, Metzler D, et al: Behavioral symptomatology in
dementia of the Alzheimer type. Alzheimer Dis Assoc Disord
2(4):363–365, 1988 3196470
Kyomen HH, Satlin A, Hennen J, Wei JY: Estrogen therapy and aggressive
behavior in elderly patients with moderate-to-severe dementia: results
from a short-term, randomized, double-blind trial. Am J Geriatr
Psychiatry 7(4):339–348, 1999 10521168
Labbate LA, Croft HA, Oleshansky MA: Antidepressant-related erectile
dysfunction: management via avoidance, switching antidepressants,
antidotes, and adaptation. J Clin Psychiatry 64 (suppl 10):11–19, 2003
12971811
Lacey JVJr, Mink PJ, Lubin JH, et al: Menopausal hormone replacement
therapy and risk of ovarian cancer. JAMA 288(3): 334–341, 2002
12117398
Laumann EO, Waite LJ: Sexual dysfunction among older adults: prevalence
and risk factors from a nationally representative U.S. probability
sample of men and women 57–85 years of age. J Sex Med 5(10):2300–
2311, 2008 18702640
Laumann EO, Paik A, Rosen RC: Sexual dysfunction in the United States:
prevalence and predictors. JAMA 281(6):537–544, 1999 10022110
Laumann EO, Paik A, Glasser DB, et al: A cross-national study of
subjective sexual well-being among older women and men: findings
from the Global Study of Sexual Attitudes and Behaviors. Arch Sex
Behav 35(2):145–161, 2006 16752118
Lewis RW, Fugl-Meyer KS, Bosch R, et al: Epidemiology/risk factors of
sexual dysfunction. J Sex Med 1(1):35–39, 2004 16422981
Lichtenberg PA, Strzepek DM: Assessments of institutionalized dementia
patients’ competencies to participate in intimate relationships.
Gerontologist 30(1):117–120, 1990 2311954
Light SA, Holroyd S: The use of medroxyprogesterone acetate for the
treatment of sexually inappropriate behaviour in patients with
dementia. J Psychiatry Neurosci 31(2):132–134, 2006 16575429
Lindau ST, Schumm LP, Laumann EO, et al: A study of sexuality and
health among older adults in the United States. N Engl J Med
357(8):762–774, 2007 17715410
Ludwig W, Phillips M: Organic causes of erectile dysfunction in men under
40. Urol Int 92(1):1–6, 2014 24281298
Marjoribanks J, Farquhar C, Roberts H, et al: Long term hormone therapy
for perimenopausal and postmenopausal women. Cochrane Database
Syst Rev 7: CD004143, 2012 22786488
Marsiglio W, Donnelly D: Sexual relations in later life: a national study of
married persons. J Gerontol 46(6):S338–S344, 1991 1940100
Masters WH, Johnson VE: Human Sexual Response. Boston, MA, Little,
Brown, 1966
Masters WH, Johnson VE: Human Sexual Inadequacy. Boston, MA, Little,
Brown, 1970
Mayers KS: Sexuality and the patient with dementia. Sex Disabil 12:213–
219, 1994
Mendez MF, Shapira JS: Hypersexual behavior in frontotemporal dementia:
a comparison with early onset Alzheimer’s disease. Arch Sex Behav
42(3):501–509, 2013 23297146
Minkin MJ, Maamari R, Reiter S: Postmenopausal vaginal atrophy:
evaluation of treatment with local estrogen therapy. Int J Womens
Health 2014(6):281–288, 2014 24648772
Montejo AL, Llorca G, Izquierdo JA, et al: Incidence of sexual dysfunction
associated with antidepressant agents: a prospective multicenter study
of 1022 outpatients. J Clin Psychiatry 62 (suppl 3):10–21, 2001
11229449
Morley JE: Testosterone and behavior. Clin Geriatr Med 19(3):605–616,
2003
Morley JE, Perry HM 3rd: Andropause: an old concept in new clothing.
Clin Geriatr Med 19(3):507–528, 2003 14567004
Morley JE, Tariq SH: Sexuality and disease. Clin Geriatr Med 19(3):563–
573, 2003 14567008
Mroczek B, Kurpas D, Gronowska M, et al: Psychosexual needs and sexual
behaviors of nursing care home residents. Arch Gerontol Geriatr
57(1):32–38, 2013 23478162
Mulligan T, Palguta RF Jr: Sexual interest, activity, and satisfaction among
male nursing home residents. Arch Sex Behav 20(2):199–204, 1991
2064543
Mulligan T, Reddy S, Gulur PV, et al: Disorders of male sexual dysfunction.
Clin Geriatr Med 19(3):473–481, 2003 14567002
Nappi RE, Wawra K, Schmitt S: Hypoactive sexual desire disorder in
postmenopausal women. Gynecol Endocrinol 22(6):318–323, 2006
16785156
National Council on the Aging: Healthy Sexuality and Vital Aging.
Executive Summary. Washington, DC, National Council on the Aging,
1998
Nelson HD, Walker M, Zakher B, et al: Menopausal hormone therapy for
the primary prevention of chronic conditions: a systematic review to
update the U.S. Preventive Services Task Force recommendations. Ann
Intern Med 157(2):104–113, 2012 22786830
Nicolai MP, Liem SS, Both S, et al: A review of the positive and negative
effects of cardiovascular drugs on sexual function: a proposed table for
use in clinical practice. Neth Heart J 22(1):11–19, 2014 24155101
Nurnberg HG, Hensley PL, Heiman JR, et al: Sildenafil treatment of
women with antidepressant-associated sexual dysfunction: a
randomized controlled trial. JAMA 300(4):395–404, 2008 18647982
Ozkan B, Wilkins K, Muralee S, et al: Pharmacotherapy for inappropriate
sexual behaviors in dementia: a systematic review of literature. Am J
Alzheimers Dis Other Demen 23(4):344–354, 2008 18509106
Padma-Nathan H, Hellstrom WJG, Kaiser FE, et al: Treatment of men with
erectile dysfunction with transurethral alprostadil. N Engl J Med
336(1):1–7, 1997 8970933
Pines A: Male menopause: is it a real clinical syndrome? Climacteric
14(1):15–17, 2011 20670200
Pope M, Schulz R: Sexual attitudes and behavior in midlife and aging
homosexual males. J Homosex 20(3–4):169–177, 1990 2086646
Porst H, Padma-Nathan H, Giuliano F, et al: Efficacy of tadalafil for the
treatment of erectile dysfunction at 24 and 36 hours after dosing: a
randomized controlled trial. Urology 62(1):121–125, discussion 125–
126, 2003a 12837435
Porst H, Young JM, Schmidt AC, et al: Efficacy and tolerability of
vardenafil for treatment of erectile dysfunction in patient subgroups.
Urology 62(3):519–523, discussion 523–524, 2003b 12946758
Porst H, Montorsi F, Rosen RC, et al: The Premature Ejaculation
Prevalence and Attitudes (PEPA) survey: prevalence, comorbidities,
and professional help-seeking. Eur Urol 51(3):816–823, 2007
16934919
Raji M, Liu D, Wallace D: Case report: sexual aggressiveness in a patient
with dementia: sustained clinical response to citalopram. Annals of
Long-Term Care 8:81–83, 2000
Redinbaugh EM, Zeiss AM, Davies HD, et al: Sexual behavior in men with
dementing illnesses. Clin Geriatr 5:45–50, 1997
Reichenpfader U, Gartlehner G, Morgan LC, et al: Sexual dysfunction
associated with second-generation antidepressants in patients with
major depressive disorder: results from a systematic review with
network meta-analysis. Drug Saf 37(1):19–31, 2014 24338044
Richardson JP, Lazur A: Sexuality in the nursing home patient. Am Fam
Physician 51(1):121–124, 1995 7810464
Roach SM: Sexual behaviour of nursing home residents: staff perceptions
and responses. J Adv Nurs 48(4):371–379, 2004 15500531
Robinson KM, Davis SJ: Influence of cognitive decline on sexuality in
individuals with dementia and their caregivers. J Gerontol Nurs
39(11):30–36, 2013 24066786
Rosen T, Lachs MS, Pillemer K: Sexual aggression between residents in
nursing homes: literature synthesis of an underrecognized problem. J
Am Geriatr Soc 58(10):1970–1979, 2010 20840462
Rossouw JE, Anderson GL, Prentice RL, et al: Risks and benefits of
estrogen plus progestin in healthy postmenopausal women: principal
results from the Women’s Health Initiative randomized controlled trial.
JAMA 288(3):321–333, 2002 12117397
Rothschild AJ: Selective serotonin reuptake inhibitor-induced sexual
dysfunction: efficacy of a drug holiday. Am J Psychiatry
152(10):1514–1516, 1995 7573593
Saretsky K: The right to be human: how one facility cares. Provider
13(10):20, 22–23, 1987 10284712
Schick V, Herbenick D, Reece M, et al: Sexual behaviors, condom use, and
sexual health of Americans over 50: implications for sexual health
promotion for older adults. J Sex Med 7 (suppl 5):315–329, 2010
21029388
Schover LR, Jensen SB: Sexuality and Chronic Illness. New York,
Guilford, 1988
Segraves RT: Antidepressant-induced sexual dysfunction. J Clin Psychiatry
59 (suppl 4):48–54, 1998 9554321
Segraves RT, Balon R: Antidepressant-induced sexual dysfunction in men.
Pharmacol Biochem Behav 121:132–137, 2014 24239785
Segraves RT, Lee J, Stevenson R: Tadalafil for treatment of erectile
dysfunction in men on antidepressants. J Clin Psychopharmacol
27(1):62–66, 2007 17224715
Serretti A, Chiesa A: A meta-analysis of sexual dysfunction in psychiatric
patients taking antipsychotics. Int Clin Psychopharmacol 26(3):130–
140, 2011 21191308
Shifren JL, Davis SR, Moreau M, et al: Testosterone patch for the treatment
of hypoactive sexual desire disorder in naturally menopausal women:
results from the INTIMATE NM1 Study. Menopause 13(5):770–779,
2006 16932240
Sison AS, Godschalk MF, Mulligan T: Erectile dysfunction in the elderly:
treatment recommendations from the recent American Urological
Association guidelines. Clin Geriatr 5:73–76, 1997
Snarch D: Constructing the Sexual Crucible: An Integration of Sexual and
Marital Therapy. New York, WW Norton, 1991
Spector IP, Rosen RC, Leiblum SR: Sexuality, in Psychiatric Care in the
Nursing Home. Edited by Reichman WE, Katz PR. New York, Oxford
University Press, 1996, pp 133–150
Suckling J, Lethaby A, Kennedy R: Local oestrogen for vaginal atrophy in
postmenopausal women. Cochrane Database Syst Rev (4):CD001500,
2006 17054136
Tariq SH, Haleem U, Omran ML, et al: Erectile dysfunction: etiology and
treatment in young and old patients. Clin Geriatr Med 19(3):539–551,
2003 14567006
Taylor MJ, Rudkin L, Bullemor-Day P, et al: Strategies for managing sexual
dysfunction induced by antidepressant medication. Cochrane Database
Syst Rev 5: CD003382, 2013 23728643
Thomas DR: Medications and sexual function. Clin Geriatr Med 19(3):553–
562, 2003 14567007
Tucker I: Management of inappropriate sexual behaviors in dementia: a
literature review. Int Psychogeriatr 22(5):683–692, 2010 20226113
Waldinger MD: Premature ejaculation: state of the art. Urol Clin North Am
34(4):591–599, 2007 17983899
Wallace M, Safer M: Hypersexuality among cognitively impaired older
adults. Geriatr Nurs 30(4):230–237, 2009 19665665
Wang WF, Wang Y, Minhas S, et al: Can sildenafil treat primary premature
ejaculation? A prospective clinical study. Int J Urol 14(4):331–335,
2007 17470165
Wasow M, Loeb MB: Sexuality in nursing homes. J Am Geriatr Soc
27(2):73–79, 1979 762368
Westheimer RK, Lopater S: Human Sexuality: A Psychosocial Perspective.
Philadelphia, PA, Lippincott Williams & Wilkins, 2002
White CB: Sexual interest, attitudes, knowledge, and sexual history in
relation to sexual behavior in the institutionalized aged. Arch Sex
Behav 11(1):11–21, 1982 7073466
White CB, Catania JA: Psychoeducational intervention for sexuality with
the aged, family members of the aged, and people who work with the
aged. Int J Aging Hum Dev 15(2):121–138, 1982 7183559
Wilson MM: Menopause. Clin Geriatr Med 19(3):483–506, 2003 14567003
Wright LK: The impact of Alzheimer’s disease on the marital relationship.
Gerontologist 31(2):224–237, 1991 2044995
Xu F, Schillinger JA, Aubin MR, et al: Sexually transmitted diseases of
older persons in Washington State. Sex Transm Dis 28(5):287–291,
2001 11357895
Zajecka J: Strategies for the treatment of antidepressant-related sexual
dysfunction: management via avoidance, switching antidepressants,
antidotes, and adaptation. J Clin Psychiatry 64 (suppl 10):11–19, 2003
Zeiss AM, Davies HD, Wood M, et al: The incidence and correlates of
erectile problems in patients with Alzheimer’s disease. Arch Sex Behav
19(4):325–331, 1990 2400296
Zilbergeld B, Ellison C: Desire discrepancies and arousal problems in sex
therapy, in Principles and Practice of Sex Therapy. Edited by Leiblum
S, Pervin L. New York, Guilford, 1980, pp 65–101
Suggested Readings
Agronin ME: Sexuality and aging, in Principles and Practice of Sex
Therapy, 5th Edition. Edited by Binik YM, Hall KSK. New York,
Guilford, 2014, pp 525–539
Bitzer J, Platano G, Tschudin S, et al: Sexual counselling in elderly couples.
J Sex Med 5(9):2027–2043, 2008
Eloniemi-Sulkava U, Notkola IL, Hämäläinen K, et al: Spouse caregivers’
perceptions of influence of dementia on marriage. Psychogeriatrics
14(1):47–58, 2002
Fisher LL: Sex, Romance, and Relationships: AARP Survey of Midlife and
Older Adults. Publication D19234. Washington, DC, American
Association of Retired Persons, 2010
Mroczek B, Kurpas D, Gronowska M, et a: Psychosexual needs and sexual
behaviors of nursing care home residents. Arch Gerontol Geriatr
57(1):32–38, 2013
Nelson HD, Walker M, Zakher B, et al: Menopausal hormone therapy for
the primary prevention of chronic conditions: a systematic review to
update the U.S. Preventive Services Task Force recommendations. Ann
Intern Med 157(2):104–113, 2012
Robinson KM, Davis SJ: Influence of cognitive decline on sexuality in
individuals with dementia and their caregivers. J Gerontol Nurs
39(11):30–36, 2013
Rosen T, Lachs MS, Pillemer K: Sexual aggression between residents in
nursing homes: literature synthesis of an underrecognized problem. J
Am Geriatr Soc 58(10):1970–1979, 2010
Tucker I: Management of inappropriate sexual behaviors in dementia: a
literature review. Int Psychogeriatr 22(5):683–692, 2010
Westheimer RK, Lopater S: Human Sexuality: A Psychosocial Perspective.
Philadelphia, PA, Lippincott, Williams & Wilkins, 2002
CHAPTER 15
Bereavement
Moria J. Smoski, Ph.D.
Stephanie T. Jenal, Ph.D.
Larry W. Thompson, Ph.D.
This work was supported in part by grant R01-AG01959 from the National Institute on Aging and
grants R01-MH36834 and R01-MH37196 from the National Institute of Mental Health. In addition,
author M.J.S. was supported by K23-MH087754.
Late-Life Bereavement
The terms bereavement and grief reaction have been used to refer to any
number of losses. These losses include (but are not limited to) the death of a
spouse, an adult child, another family member, or a close personal friend;
divorce (Cain 1988); anticipatory grief associated with caregiving for a
severely impaired loved one (Bass et al. 1991); and a significant decline in
one’s own health, attractiveness, capabilities, opportunities, and so forth
(Kalish 1987). When used in its narrowest sense, however, bereavement
refers to the reaction or process that results after the death of someone
close. Indeed, the death of a spouse is generally accepted as the most
common and traumatic life event in late life (Jacobs and Ostfeld 1977).
In the United States, 40% of women and 13% of men older than 65 years
have lost a spouse (Federal Interagency Forum on Aging Related Statistics
2012). The mean duration of widowhood or widowerhood is approximately
14 years for women versus only 7 years for men (U.S. Census Bureau
2001). These data, plus the fact that widowers are more likely than widows
to remarry after losing their spouses, have often led to the interpretation that
bereavement is a women’s issue. Although elderly women may live without
a spouse longer than their male peers, they also have lower mortality rates.
For example, in the University of Southern California longitudinal study of
spousal bereavement, the first year after bereavement saw a mortality rate
of 12% in men but only about 1% in women (Gallagher-Thompson et al.
1993; Thompson et al. 1991). Thus, the loss of a spouse is an important
issue regardless of gender.
DSM Definitions
The recently revised DSM-5 (American Psychiatric Association 2013)
provides a clear distinction between the feelings of loss and emptiness that
typically characterize bereavement and the persistent, depressed mood and
global inability to experience pleasure indicative of a major depressive
episode. The manual notes that the sadness accompanying grief tends to
come in waves, and when it abates, feelings of joy and the ability for humor
often manifest; in contrast, the “down” state of major depressive episode
offers no respite. According to DSM-IV (American Psychiatric Association
1994), bereavement within the first 2 months following a loss was a specific
rule-out for a major depressive episode (unless the episode was
characterized by “marked functional impairment, morbid preoccupation
with worthlessness, suicidal ideation, psychotic symptoms, or psychomotor
retardation” [p. 355]). In contrast, DSM-5 acknowledges the potential
comorbidity of these conditions by eliminating this “bereavement
exclusion” and noting that bereaved individuals who meet the criteria for
major depressive episode will not be denied this diagnosis because of their
bereaved status (Pies 2013).
References
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Bass DM, Bowman K, Noelker LS: The influence of caregiving and
bereavement support on adjusting to an older relative’s death.
Gerontologist 31(1):32–42, 1991 2007473
Bennett KM, Gibbons K, Mackenzie-Smith S: Loss and restoration in later
life: an examination of the dual process model of coping with
bereavement. Omega (Westport) 61(4):315–332, 2010 21058612
Bierhals AJ, Prigerson HG, Fasiczka A, et al: Gender differences in
complicated grief among the elderly. Omega 32:303–317, 1995–1996
Bonanno GA: The Other Side of Sadness: What the New Science of
Bereavement Tells Us about Life after Loss. New York, Basic Books,
2009
Bonanno GA, Wortman CB, Lehman DR, et al: Resilience to loss and
chronic grief: a prospective study from preloss to 18-months postloss. J
Pers Soc Psychol 83(5):1150–1164, 2002 12416919
Bonanno GA, Wortman CB, Nesse RM: Prospective patterns of resilience
and maladjustment during widowhood. Psychol Aging 19(2):260–271,
2004 15222819
Bowlby J: Processes of mourning. Int J Psychoanal 42:317–340, 1961
13872076
Bowling A: Who dies after widow(er)hood? A discriminant analysis.
Omega 19:135–153, 1988–1989
Bui E, Nadal-Vicens M, Simon NM: Pharmacological approaches to the
treatment of complicated grief: rationale and a brief review of the
literature. Dialogues Clin Neurosci 14(2):149–157, 2012 22754287
Burton AM, Haley WE, Small BJ: Bereavement after caregiving or
unexpected death: effects on elderly spouses. Aging Ment Health
10(3):319–326, 2006 16777661
Cain BS: Divorce among elderly women: a growing social phenomenon.
Soc Casework 69:563–568, 1988
Carlsson ME, Nilsson IM: Bereaved spouses’ adjustment after the patients’
death in palliative care. Palliat Support Care 5(4):397–404, 2007
18044417
Carr D: Gender, preloss marital dependence, and older adults’ adjustment to
widowhood. J Marriage Fam 66:220–235, 2004
Cobb S: Presidential Address 1976. Social support as a moderator of life
stress. Psychosom Med 38(5):300–314, 1976 981490
Coifman KG, Bonanno G, Rafaeli E: Affect dynamics, bereavement and
resilience to loss. J Happiness Stud 8:371–392, 2007a
Coifman KG, Bonanno GA, Ray RD, et al: Does repressive coping promote
resilience? Affective-autonomic response discrepancy during
bereavement. J Pers Soc Psychol 92(4):745–758, 2007b 17469956
Currier JM, Neimeyer RA, Berman JS: The effectiveness of
psychotherapeutic interventions for bereaved persons: a comprehensive
quantitative review. Psychol Bull 134(5):648–661, 2008 18729566
Currier JM, Holland JM, Neimeyer RA: Do CBT-based interventions
alleviate distress following bereavement? A review of the current
evidence. Int J Cogn Ther 3:77–93, 2010
Dimond M, Lund DA, Caserta MS: The role of social support in the first
two years of bereavement in an elderly sample. Gerontologist
27(5):599–604, 1987 3678899
Farberow NL, Gallagher DE, Gilewski MJ, et al: An examination of the
early impact of bereavement on psychological distress in survivors of
suicide. Gerontologist 27(5):592–598, 1987 3678898
Federal Interagency Forum on Aging Related Statistics: Older Americans
2012: Key Indicators of Well-Being. Washington, DC, Federal
Interagency Forum on Aging Related Statistics, 2012
Florsheim M, Gallagher-Thompson D: Cognitive/behavioral treatment of
atypical bereavement: a case study. Clin Gerontol 10:73–76, 1990
Freud S: Mourning and melancholia (1917 [1915]), in The Standard Edition
of the Complete Psychological Works of Sigmund Freud, Vol 14.
Translated and edited by Strachey J. London, Hogarth, 1957, pp 237–
260
Futterman A, Holland JM, Brown PJ, et al: Factorial validity of the Texas
Revised Inventory of Grief-Present scale among bereaved older adults.
Psychol Assess 22(3):675–687, 2010 20822280
Gallagher-Thompson D, Futterman A, Farberow N, et al: The impact of
spousal bereavement on older widows and widowers, in Handbook of
Bereavement. Edited by Stroebe MS, Stroebe W, Hansson R.
Cambridge, United Kingdom, Cambridge University Press, 1993, pp
227–239
García JA, Landa V, Grandes G, et al: Effectiveness of “primary
bereavement care” for widows: a cluster randomized controlled trial
involving family physicians. Death Stud 37(4):287–310, 2013
24520889
Gilewski MJ, Farberow NL, Gallagher DE, Thompson LW: Interaction of
depression and bereavement on mental health in the elderly. Psychol
Aging 6(1):67–75, 1991 2029370
Haley WE, Bergman EJ, Roth DL, et al: Long-term effects of bereavement
and caregiver intervention on dementia caregiver depressive symptoms.
Gerontologist 48(6):732–740, 2008 19139247
Harlow SD, Goldberg EL, Comstock GW: A longitudinal study of the
prevalence of depressive symptomatology in elderly widowed and
married women. Arch Gen Psychiatry 48(12):1065–1068, 1991
1845223
Holland JM, Currier JM, Gallagher-Thompson D: Outcomes from the
Resources for Enhancing Alzheimer’s Caregiver Health (REACH)
program for bereaved caregivers. Psychol Aging 24(1):190–202, 2009
19290751
Holland JM, Futterman A, Thompson LW, et al: Difficulties accepting the
loss of a spouse: a precursor for intensified grieving among widowed
older adults. Death Stud 37(2):126–144, 2013 24520845
Horowitz MJ: Stress Response Syndromes. New York, Jason Aronson,
1976
Horowitz MJ, Krupnick J, Kaltreider N, et al: Initial psychological response
to parental death. Arch Gen Psychiatry 38(3):316–323, 1981 7212963
Horowitz MJ, Weiss DS, Kaltreider N, et al: Reactions to the death of a
parent: results from patients and field subjects. J Nerv Ment Dis
172(7):383–392, 1984 6726208
Horowitz MJ, Siegel B, Holen A, et al: Diagnostic criteria for complicated
grief disorder. Am J Psychiatry 154(7):904–910, 1997 9210739
Itzhar-Nabarro Z, Smoski MJ: A review of theoretical and empirical
perspectives on marital satisfaction and bereavement outcomes:
implications for working with older adults. Clin Gerontol 35:257–269,
2012
Jacobs S: Pathologic Grief: Maladaptation to Loss. Washington, DC,
American Psychiatric Press, 1993
Jacobs S, Ostfeld A: An epidemiological review of the mortality of
bereavement. Psychosom Med 39(5):344–357, 1977 333498
Jordan JR, Neimeyer RA: Does grief counseling work? Death Stud
27(9):765–786, 2003 14577426
Kalish RA: Older people and grief. Generations 11:33–38, 1987
Kitson GC: Adjustment to violent and natural deaths in later and earlier life
for black and white widows. J Gerontol B Psychol Sci Soc Sci
55(6):S341–S351, 2000 11078111
Kübler-Ross E: On Death and Dying. New York, Simon & Schuster, 1969
Lee GR, DeMaris A: Widowhood, gender, and depression: a longitudinal
analysis. Res Aging 29:56–72, 2007
Lichtenstein P, Gatz M, Pedersen NL, et al: A co-twin—control study of
response to widowhood. J Gerontol B Psychol Sci Soc Sci 51(5):P279–
P289, 1996 8809004
Lindemann E: Symptomatology and management of acute grief. Am J
Psychiatry 101:141–148, 1944
Lopata HZ: Current Widowhood: Myths and Realities. Thousand Oaks, CA,
Sage, 1996
Lund DA, Caserta M, Dimond M: The course of spousal bereavement in
later life, in Handbook of Bereavement. Edited by Stroebe MS, Stroebe
W, Hansson R. Cambridge, UK, Cambridge University Press, 1993, pp
240–254
Maciejewski PK, Zhang B, Block SD, Prigerson HG: An empirical
examination of the stage theory of grief. JAMA 297(7): 716–723, 2007
17312291
McCrae RR, Costa PT: Psychological resilience among widowed men and
women: a 10-year follow-up of a national sample, in Handbook of
Bereavement. Edited by Stroebe MS, Stroebe W, Hansson R.
Cambridge, UK, Cambridge University Press, 1993, pp 196–207
Miller MD, Wolfson L, Frank E, et al: Using interpersonal psychotherapy
(IPT) in a combined psychotherapy/medication research protocol with
depressed elders: a descriptive report with case vignettes. J Psychother
Pract Res 7(1):47–55, 1997 9407475
Momtaz YA, Ibrahim R, Hamid TA, Yahaya N: Mediating effects of social
and personal religiosity on the psychological well being of widowed
elderly people. Omega 61(2):145–162, 2010 20712141
National Institutes of Health Consensus Conference: Diagnosis and
treatment of depression in late life. JAMA 268:1018–1024, 1992
1501308
Neimeyer RA: The Lessons of Loss: A Guide to Coping. Raleigh, NC,
McGraw-Hill, 1998
Neimeyer RA: Searching for the meaning of meaning: grief therapy and the
process of reconstruction. Death Stud 24(6):541–558, 2000 11503667
Neimeyer RA, Harris DL, Winokuer HR, et al (eds): Grief and
Bereavement in Contemporary Society: Bridging Research and
Practice. New York, Routledge, 2011
Oakley F, Khin NA, Parks R, et al: Improvement in activities of daily living
in elderly following treatment for post-bereavement depression. Acta
Psychiatr Scand 105(3):231–234, 2002 11939978
Osterweis M, Solomon F, Green M: Bereavement: Reactions,
Consequences, and Care: A Report of the Institute of Medicine,
National Academy of Sciences. Washington, DC, National Academy
Press, 1984
Ott CH, Lueger RJ, Kelber ST, et al: Spousal bereavement in older adults:
common, resilient, and chronic grief with defining characteristics. J
Nerv Ment Dis 195(4):332–341, 2007 17435484
Parkes CM: Bereavement: Studies of Grief in Adult Life. New York,
International Universities Press, 1972
Parkes CM, Weiss RS: Recovery From Bereavement. New York, Basic
Books, 1983
Pasternak RE, Reynolds CF 3rd, Schlernitzauer M, et al: Acute open-trial
nortriptyline therapy of bereavement-related depression in late life. J
Clin Psychiatry 52(7):307–310, 1991 2071562
Pies R: How the DSM-5 got grief, bereavement right. Psych Central, 2013.
Available at: http://psychcentrol.com/blog/archives/2013/05/31/how-
the-dsm-5-got-grief-bereavement-right/. Accessed November 15, 2013.
Prigerson HG, Shear MK, Jacobs SC, et al: Consensus criteria for traumatic
grief: a preliminary empirical test. Br J Psychiatry 174:67–73, 1999
10211154
Raphael B, Minkov C, Dobson M: Psychotherapeutic and pharmacological
intervention for bereaved persons, in Handbook of Bereavement
Research: Consequences, Coping, and Care. Edited by Stroebe MS,
Hansson RO, Stroebe W, et al. Washington, DC, American
Psychological Association, 2001, pp 587–612
Reynolds CF 3rd: Treatment of depression in special populations. J Clin
Psychiatry 53 (suppl):45–53, 1992 1522079
Reynolds CF 3rd, Miller MD, Pasternak RE, et al: Treatment of
bereavement-related major depressive episodes in later life: a
controlled study of acute and continuation treatment with nortriptyline
and interpersonal psychotherapy. Am J Psychiatry 156(2):202–208,
1999 9989555
Rosenblatt PC: Grief that does not end, in Continuing Bonds: New
Understandings of Grief (Series in Death Education, Aging, and Health
Care, 0275–3510). Edited by Klass D, Silverman PR, Nickman SL.
Washington, DC, Taylor and Francis, 1996, pp 45–58
Rozenzweig A, Prigerson H, Miller MD, et al: Bereavement and late-life
depression: grief and its complications in the elderly. Annu Rev Med
48:421–428, 1997 9046973
Sanders CM: Risk factors in bereavement outcome, in Handbook of
Bereavement. Edited by Stroebe MS, Stroebe W, Hansson R.
Cambridge, UK, Cambridge University Press, 1993, pp 255–267
Schut H, Stroebe MS, van den Bout J, et al: The efficacy of bereavement
interventions: determining who benefits, in Handbook of Bereavement
Research: Consequences, Coping, and Care. Edited by Stroebe MS,
Hansson RO, Stroebe W, et al. Washington, DC, American
Psychological Association, 2001, pp 705–737
Shah SM, Carey IM, Harris T, et al: Initiation of psychotropic medication
after partner bereavement: a matched cohort study. PLoS ONE
8(11):e77734, 2013 24223722
Shear K, Frank E, Houck PR, et al: Treatment of complicated grief: a
randomized controlled trial. JAMA 293:2601–2608, 2005 15928281
Simon NM, Shear MK, Fagiolini A, et al: Impact of concurrent naturalistic
pharmacotherapy on psychotherapy of complicated grief. Psychiatry
Res 159(1–2):31–36, 2008 18336918
Steffen E, Coyle A: Sense of presence experiences and meaning-making in
bereavement: a qualitative analysis. Death Stud 35(7):579–609, 2011
24501839
Stroebe M, Schut H: The dual process model of coping with bereavement:
rationale and description. Death Stud 23(3):197–224, 1999 10848151
Stroebe M, Schut H: The dual process model of coping with bereavement: a
decade on. Omega (Westport) 61(4):273–289, 2010 21058610
Stroebe M, Hansson RO, Stroebe W, et al: Introduction: concepts and issues
in contemporary research on bereavement, in Handbook of
Bereavement Research: Consequences, Coping, and Care. Edited by
Stroebe MS, Hansson RO, Stroebe W, et al. Washington, DC,
American Psychological Association, 2001a, pp 3–22
Stroebe MS, Stroebe W, Schut H: Gender differences in adjustment to
bereavement: an empirical and theoretical review. Rev Gen Psychol
5:62–83, 2001b
Stroebe M, Schut H, Boerner K: Continuing bonds in adaptation to
bereavement: toward theoretical integration. Clin Psychol Rev
30(2):259–268, 2010 20034720
Thompson LW, Breckenridge JN, Gallagher D, et al: Effects of
bereavement on self-perceptions of physical health in elderly widows
and widowers. J Gerontol 39(3):309–314, 1984 6715808
Thompson LW, Gallagher-Thompson D, Futterman A, et al: The effects of
late-life spousal bereavement over a 30-month interval. Psychol Aging
6(3):434–441, 1991 1930760
Tol WA, Barbui C, van Ommeren M: Management of acute stress, PTSD,
and bereavement: WHO recommendations. JAMA 310(5):477–478,
2013 23925613
U.S. Census Bureau: Marital Status of People 15 Years and Over, By Age,
Sex, Personal Earnings, Race, and Hispanic Origin, March 2000.
Released June 29, 2001. Available at
http://www.census.gov/population/www/socdemo/hh-
fam/cps2001.html. Accessed August 12, 2008.
van der Houwen K, Stroebe M, Stroebe W, et al: Risk factors for
bereavement outcome: a multivariate approach. Death Stud 34(3): 195–
220, 2010 24479181
van Grootheest DS, Beekman ATF, Broese van Groenou MI, et al: Sex
differences in depression after widowhood. Do men suffer more? Soc
Psychiatry Psychiatr Epidemiol 34(7):391–398, 1999 10477960
Viney L: The construing widow: dislocation and adaptation in bereavement.
Psychother Patient 6:207–222, 1990
Warner J, Metcalfe C, King M: Evaluating the use of benzodiazepines
following recent bereavement. Br J Psychiatry 178(1):36–41, 2001
11136208
Wetherell JL: Complicated grief therapy as a new treatment approach.
Dialogues Clin Neurosci 14(2):159–166, 2012 22754288
Williams K: Has the future of marriage arrived? A contemporary
examination of gender, marriage, and psychological well-being. J
Health Soc Behav 44(4):470–487, 2003 15038144
Wilson J: The assimilation of problematic experiences sequence: an
approach to evidence-based practice in bereavement counseling. J Soc
Work End Life Palliat Care 7(4):350–362, 2011 22150179
Wittouck C, Van Autreve S, De Jaegere E, et al: The prevention and
treatment of complicated grief: a meta-analysis. Clin Psychol Rev
31(1):69–78, 2011 21130937
Worden JW: Grief Counseling and Grief Therapy, 3rd Edition. New York,
Springer, 2002
Wortman C, Silver RC: Coping with irrevocable loss, in Cataclysms, Crises,
and Catastrophes: Psychology in Action (The Master Lectures). Edited
by Van den Bos G, Bryant BK. Washington, DC, American
Psychological Association, 1987, pp 185–235
Wortman CB, Silver RC: The myths of coping with loss. J Consult Clin
Psychol 57(3): 349–357, 1989 2661609
Zisook S, DeVaul R: Measuring acute grief. Psychiatr Med 2(2):169–176,
1984 6571620
Zisook S, Devaul RA, Click MA Jr: Measuring symptoms of grief and
bereavement. Am J Psychiatry 139(12):1590–1593, 1982 7149059
Zisook S, Shuchter SR, Pedrelli P, et al: Bupropion sustained release for
bereavement: results of an open trial. J Clin Psychiatry 62(4):227–230,
2001 11379835
Zygmont M, Prigerson HG, Houck PR, et al: A post hoc comparison of
paroxetine and nortriptyline for symptoms of traumatic grief. J Clin
Psychiatry 59(5):241–245, 1998 9632035
Suggested Readings
Bonanno GA: The Other Side of Sadness: What the New Science of
Bereavement Tells Us About Life After Loss. New York, Basic Books,
2009
Breckenridge J, Gallagher D, Thompson LW, et al: Characteristic
depressive symptoms of bereaved elders. J Gerontol 41:163–168, 1986
Currier JM, Neimeyer RA, Berman JS: The effectiveness of
psychotherapeutic interventions for bereaved persons: a comprehensive
quantitative review. Psychol Bull 134:648–661, 2008
Horowitz MJ, Siegel B, Holen A, et al: Diagnostic criteria for complicated
grief disorder. Am J Psychiatry 154:904–910, 1997
Maciejewski PK, Zhang B, Block SD, et al: An empirical examination of
the stage theory of grief. JAMA 297:716–723, 2007
Neimeyer R: Searching for the meaning of meaning: grief therapy and the
process of reconstruction. Death Stud 24:531–558, 2000
Ott CH, Lueger RJ, Kelber ST, et al: Spousal bereavement in older adults:
common, resilient, and chronic grief with defining characteristics. J
Nerv Ment Dis 195:332–341, 2007
Prigerson HG, Shear MK, Jacobs SC, et al: Consensus criteria for traumatic
grief: a preliminary empirical test. Br J Psychiatry 174:67–73, 1999
Stroebe M, Schut H: The dual process model of coping with bereavement:
rationale and description. Death Stud 23:197–224, 1999
Wortman C, Silver RC: Coping with irrevocable loss, in Cataclysms, Crises,
and Catastrophes: Psychology in Action (The Master Lectures). Edited
by Van den Bos G, Bryant BK. Washington, DC, American
Psychological Association, 1987, pp 185–235
CHAPTER 16
The authors would like to thank Jack D. Edinger, Ph.D., and William K. Wohlgemuth, Ph.D., who
were coauthors on the previous edition of this chapter.
Neuropsychiatric Disorders
Bereavement-Related Depression
Psychological factors that most commonly affect sleep in elderly persons
are reactions to loss, such as loss of health or functional capacity, and
reactions to the death of a friend or loved one. Although grief is a normal
reaction to bereavement, it is often associated with substantial sleep
disturbance (American Psychiatric Association 2013). When bereavement-
related grief is associated with more frequent intrusive thoughts and
avoidance behaviors, there appears to be more sleep disturbance,
predominantly in the form of difficulty in falling asleep (Hall et al. 1997).
Bereavement and depression are closely linked, however. If symptoms are
severe or persistent, depression may be diagnosed in a bereaved person
after taking into consideration his or her psychiatric history and the cultural
norms for what may be normal grieving (American Psychiatric Association
2013). Antidepressant medication may be helpful. A short course of
sedative-hypnotic therapy may provide substantial symptomatic relief. If
this approach is taken, the medication should be tapered off when the other
symptoms of bereavement-related grief diminish. Because the clinician will
not know at the outset how long treatment will be needed, considerations
related to longer-term treatment pertain (see “Pharmacological Treatment”
later in this chapter). Although unlikely, it is possible for rebound insomnia
to occur after a relatively short course (3–4 weeks) of treatment. For this
reason, clinicians should warn patients of this possibility and wait at least
several days after discontinuing the medication to determine whether there
is persistent insomnia. If all symptoms of bereavement depression have
resolved except insomnia, cognitive-behavioral therapy for insomnia (CBT-
I) should be considered (see “Cognitive-Behavioral Treatment” later in this
chapter). Grief counseling should also be considered.
Neurocognitive Disorders
Alzheimer’s Disease
Individuals with Alzheimer’s disease (AD) have been found to experience
an increased number of arousals and awakenings, to take more daytime
naps, and to have a diminished amount of REM sleep and slow-wave sleep
(Prinz et al. 1982). Patients may develop circadian rhythm disturbances
leading to the decreased night-time sleep and increased daytime sleep
(Bliwise 2004). Patients with advanced dementia may have periods of
wakefulness and sleep during each hour of the 24-hour day. Moreover,
cholinesterase inhibitors, the primary treatment for AD, can cause vivid
dreaming and insomnia. Individuals with dementia also often experience
behavioral agitation, including wandering, vocalizations, and general
combativeness, which are thought to worsen in the evening (a phenomenon
called “sundowning”). However not all studies report this kind of temporal
specificity in agitation symptoms (Bliwise 2004). Nocturnal sleep
disruption is among the leading reasons that individuals with dementia
become institutionalized (Pollak and Perlick 1991; Pollak et al. 1990;
Sanford 1975).
Pharmacological and nonpharmacological treatment studies of sleep
disturbance in patients with AD have focused on global impairment of sleep
rather than on the construct of sundowning. Many aspects of good sleep
hygiene that are emphasized in the management of insomnia in the general
population can help patients with dementia who have sleep disturbance.
These include an activity/exercise program, avoidance of daytime naps, and
limiting evening beverages (including elimination of alcohol and coffee).
Morning light therapy and caregiver education may also be helpful (Bliwise
2000; McCurry et al. 2005). For patients who go to bed very early (e.g., 8
P.M.) and awaken in the very early hours of the morning (e.g., 3 A.M.),
exposure to light in the evening hours or exercise in the evening hours can
help postpone bedtime and consequently time of awakening. A recent
Cochrane review found no evidence that melatonin improved any major
sleep outcomes in patients with AD (McCleery et al. 2014). In a small 2-
week study, trazodone 50 mg at bedtime was found to improve nighttime
sleep without worsening daytime sleepiness (Camargos et al. 2014). There
are no placebo-controlled trials of either benzodiazepines or
nonbenzodiazepines in the treatment of sleep disturbance in AD.
Management of sundowning should include identification and treatment of
possible precipitants (e.g., pain, delirium, psychiatric diagnoses, sleep
disorders). If symptoms of agitation persist after addressing causes, then
these should be addressed with nonpharmacological (preferred) and
pharmacological approaches when needed (see Chapter 19, “Agitation in
Older Adults,” for details).
Parkinson’s Disease
Sleep complaints are noted in 60%–90% of individuals with Parkinson’s
disease (PD) (Trenkwalder 1998). The majority of PD patients with affected
sleep experience difficulty in initiating and maintaining sleep, daytime
fatigue and sleepiness, RLS, and an inability to turn over in bed. The last of
these features was rated as the most troublesome symptom of sleep
disturbance in a study by Lees et al. (1988). Another sleep problem seen in
patients with PD is REM sleep behavior disorder (RBD), in which the
patient acts out dreams because the paralysis that usually occurs during
REM sleep is absent (Clarenbach 2000). In fact, patients (typically men)
with the idiopathic form of RBD are at increased risk of developing a
parkinsonian disorder or dementia, usually linked to α-synuclein pathology,
the risk being 20%–40% after an average of 5 years follow-up (Postuma et
al. 2009; Schenck et al. 1996). Dopaminergic medications used to treat PD,
such as carbidopa/levodopa, may contribute to sleep initiation problems and
sleep difficulties in the first half of the night and may cause nightmares
(Trenkwalder 1998). Few studies are available to guide treatment of
insomnia in patients with PD. The preferred treatment for RBD is low-dose
clonazepam (0.5–1 mg at bedtime), in addition to addressing environmental
safety to prevent injury when dreams are acted out. Clonazepam is effective
in 90% of cases, with results seen in as little as 1 week and with only rare
occurrence of tolerance in older adults (Bloom et al. 2009). Results from a
pilot placebo-controlled trial suggest that doxepin (10 mg at bedtime) may
improve insomnia in patients with PD (Rios Romenets et al. 2013). A small
study comparing eszopiclone and placebo did not show improved total
sleep time (which was the primary outcome measure) with eszopiclone, but
eszopiclone did improve quality of sleep and some measures of sleep
maintenance (Menza et al. 2010). A small open-label trial of low-dose
quetiapine (mean dose of 32 mg given at bedtime) demonstrated benefit
with good tolerability (Juri et al. 2005). Finally, although there are no
studies of zolpidem for insomnia in PD, recent preliminary reports suggest
that subhypnotic doses of zolpidem may help reduce dyskinesias in patients
with PD (Chen et al. 2008; Růzicka et al. 2000).
Medical Conditions
Pain
Pain is a central feature of many medical conditions that occur with
increased frequency in elderly individuals; these conditions include
arthritis, neuropathies, angina, reflux esophagitis, and peptic ulcer disease
(Aldrich 2000). Disruption of sleep is frequently noted in persons with
significant pain (Pilowsky et al. 1985). Attempts to ameliorate the condition
causing the pain should be the first step. When these attempts fail, treatment
for the pain should be instituted. Often, combined behavioral and
pharmacological treatment is needed. Some evidence indicates that pain,
like depression, may have a bidirectional relationship with sleep
disturbance. Two studies have suggested that the treatment of insomnia
leads to improvement in pain. These studies involved the treatment of
individuals with fibromyalgia with CBT-I and treatment of individuals with
rheumatoid arthritis with the benzodiazepine triazolam (Edinger et al. 2005;
Walsh et al. 1996). These data speak to the importance of treating insomnia
in patients with chronic pain (see “Treatment of Insomnia” later in this
chapter). In another study, CBT-I was effective for older adults with
insomnia comorbid with osteoarthritis pain; however, pain severity was not
significantly impacted by the treatment (Vitiello et al. 2013).
Cerebrovascular Disease
The sleep pathology associated with cerebrovascular disease depends on
which areas of the brain are affected by the condition. Hypersomnia has
been associated with lesions of the cephalad portions of the ascending
reticular activating system, which includes the midbrain and paramedian
region of the thalamus (the thalamic lesions most commonly occur in the
dorsomedial nucleus, intralaminar nuclei, and centromedian nucleus)
(Bassetti and Chervin 2000). Large lesions of the cerebral hemispheres and
lesions of other regions such as the caudate and striatum have been less
commonly associated with hypersomnia. Insomnia directly related to
damage of specific areas of the brain is much less common than insomnia
caused by multifactorial complications of strokes or other medical or
psychiatric conditions associated with an individual’s cerebrovascular
disease (Bassetti and Chervin 2000). Therefore, treatment should be
directed toward these associated conditions.
Nocturia
The urge to urinate is an often-overlooked cause of awakenings in the
elderly population (Bliwise 2000). Surprisingly, it has been reported that
nocturia (excessive urination at night) is the most common explanation
given by elderly individuals for difficulty in maintaining sleep; 63%–72%
of elderly individuals cite nocturia as a reason for sleep maintenance
problems (Middelkoop et al. 1996). Furthermore, several studies have
documented the sleep disturbance caused by and daytime adverse effects of
nocturia (Bliwise 2000). The most common causes of nocturia are
conditions that increase in frequency with age: benign prostatic hypertrophy
in men and decreased urethral resistance due to decreased estrogen levels in
women (Bliwise 2000). Sleep apnea, which increases in prevalence in the
elderly population, can also lead to nocturia (Bliwise 2000). Thus, when
evaluating elderly individuals with complaints of sleep maintenance, the
clinician should assess for nocturia and the associated conditions that
increase the risk of nocturia. If detected, this disorder can generally be
treated effectively by addressing the underlying condition. Patients should
also be advised to restrict fluids in the evenings, to avoid alcohol, and to
ensure that diuretics are dosed in the mornings.
Menopause-Related Symptoms
Despite the enormous number of individuals with menopause-related sleep
difficulties, there is a striking lack of research in this area (Krystal et al.
1998). Although little is known, there appears to be clear evidence that
many women experience sleep disruption in association with vasomotor
symptoms (night sweats, hot flashes, decreased urethral resistance often
leading to nocturia) that are caused by decreased levels of circulating
estrogen and progesterone (Bliwise 2000; Krystal et al. 1998). Several
factors hinder elucidation of menopausal sleep disturbance. One is that
many disorders that cause insomnia increase in frequency with age and are
highly prevalent during the period in which women experience menopausal
changes. Another factor is that although hormone replacement therapy is
highly effective in ameliorating the vasomotor symptoms of menopause, the
subjective reports of sleep disturbance often do not change (Krystal et al.
1998). Although menopausal sleep disturbance is poorly understood, it has
been suggested that behavioral conditioning occurs, just as is often the case
with individuals who, having experienced insomnia during a period of high
stress, continue to have insomnia after the stress has resolved (Krystal et al.
1998).
Given these considerations, elderly women with insomnia should be
evaluated for underlying causes of sleep disturbance (e.g., medical and
psychiatric conditions, primary sleep disorders), and it should be
determined whether there is an association between changes in menstrual
periods, vasomotor symptoms, and insomnia symptoms. If an association
between insomnia and menopausal changes appears to exist, a trial of
hormone replacement therapy could be considered. If hormone therapy is
contraindicated, the antidepressants venlafaxine, paroxetine, and fluoxetine,
as well as gabapentin, have shown some efficacy in the management of
vasomotor symptoms with good tolerability (North American Menopause
Society 2004). If an insomnia complaint persists after amelioration of
vasomotor symptoms, other treatments such as pharmacological
management of insomnia or CBT-I should be considered (see “Treatment of
Insomnia” later in this chapter). Two studies have been carried out in
women with insomnia occurring in association with menopause (Dorsey et
al. 2004; Soares et al. 2006). These studies document the efficacy of
zolpidem 10 mg at bedtime and eszopiclone 3 mg at bedtime for improving
sleep difficulties that occur in association with hot flashes. Further studies
are needed to determine the efficacy of CBT-I and of other insomnia agents.
Treatment of Insomnia
Although changes in sleep appear to be a part of aging, insomnia is not an
inevitable consequence of aging. No treatment is needed to address the
changes in sleep that normally occur. However, when individuals
experience insomnia as defined in the chapter introduction, treatment
should be instituted, because untreated insomnia is associated with
significant morbidity and decreased quality of life (Breslau et al. 1996;
National Institutes of Health 2005; Ohayon 2002; Ozminkowski et al. 2007;
Weissman et al. 1997; Zammit et al. 1999). Treatment of insomnia in
elderly patients poses some particular challenges. In this section, we discuss
the two major types of treatment for insomnia: cognitive-behavioral
treatment and pharmacological treatment. In this discussion, we focus on
issues relevant to elderly patients.
Cognitive-Behavioral Treatment
Myriad lifestyle changes that accompany aging increase risks of insomnia
among older adults (Morgan 2000). With aging comes the increased
incidence of infirmities that lead to reduced activity levels and a general
flattening of the sleep-wake activity rhythm. Retirement leads to increased
vacant time and a loss of both routine and zeitgebers that regulate and
stabilize the sleep-wake cycle. Retirement coupled with loss of a spouse
may lead to dramatically reduced social contacts and increased boredom.
Many individuals attempt to reduce hours of daytime boredom by daytime
napping and by staying in bed longer during their nighttime sleep period.
Such practices often lead to increased nocturnal wake time. Dysfunctional
beliefs about sleep, such as “Everyone should try to get 8 hours a night” and
“Older adults can do little to improve their sleep,” may actually perpetuate
sleep difficulties over time (Means and Edinger 2002; Morin et al. 1993).
Nonpharmacological interventions that address these misconceptions and
the sleep-disruptive habits they sustain are often useful for combating
insomnia in older patients.
Currently, a range of behavioral interventions are available for treating
these patients, including relaxation therapies, cognitive therapies, and
treatments that target disruptive sleep habits. Among the more effective of
these interventions is stimulus control therapy, developed by Bootzin
(1972). This treatment is particularly useful for older individuals who have
fallen out of a normal sleep-wake routine and for those who compromise
their nighttime sleep by excessive daytime napping. Stimulus control
therapy addresses such problems by curtailing daytime napping and by
enforcing a consistent sleep-wake schedule. In addition, this treatment
enhances sleep-inducing qualities of the bedroom by eliminating sleep-
incompatible behaviors in bed. The patient with insomnia is instructed to go
to bed only when sleepy; establish a standard wake-up time; get out of bed
whenever he or she is awake for more than 15 or 20 minutes; avoid reading,
watching TV, eating, worrying, and engaging in other sleep-incompatible
behaviors in the bed and bedroom; and refrain from daytime napping. This
treatment has appeal because it is easily understood and usually can be
outlined in one visit. However, follow-up visits are usually needed to ensure
compliance and achieve optimal success.
Because older adults appear to have a reduced homeostatic sleep drive
(Dijk et al. 1997) as well as a propensity to spend excessive time in bed
(Carskadon et al. 1982), measures are often needed to reduce the amount of
time older patients with insomnia routinely allot for nocturnal sleep. Such a
reduction is the aim of sleep restriction therapy (Spielman et al. 1987;
Wohlgemuth and Edinger 2000). This treatment typically begins with the
patient maintaining a sleep log. After 2–3 weeks, the average total sleep
time (TST) is calculated. Subsequently, an initial time-in-bed (TIB)
prescription may be set either at the average TST or at a value equal to the
average TST plus an amount of time that is deemed to represent normal
nocturnal wakefulness (e.g., 30 minutes). However, unless evidence
suggests that the individual has an unusually low sleep requirement, the
initial TIB prescription is seldom set at less than 5 hours per night. The TIB
prescription is increased by 15- to 20-minute increments after weeks in
which the person with insomnia sleeps more than 85%–90% of the TIB, on
average, and continues to report daytime sleepiness. Conversely, TIB is
usually reduced by similar increments after weeks in which the individual
sleeps less than 80% of the time spent in bed, on average. Because TIB
adjustments are usually necessary, sleep restriction therapy typically entails
an initial visit, when treatment instructions are given, and follow-up visits,
when TIB prescriptions are altered.
Research suggests that stimulus-control and sleep-restriction therapies
are more effective than most other nonpharmacological interventions
(Morin et al. 1999, 2006; Murtagh and Greenwood 1995). Moreover, a
meta-analytic comparison suggests that behavioral therapies compare
favorably with hypnotic pharmacotherapies in terms of short-term treatment
effects and, unlike hypnotics, have enduring benefits and few side effects
(Smith et al. 2002). In recent years, the term cognitive-behavioral therapy
for insomnia (CBT-I) has been used to refer to a specific combination of
therapies for insomnia that include stimulus control, sleep restriction, and
cognitive strategies to alter dysfunctional sleep-related beliefs. Clinical
trials have generally suggested that this combination of therapies holds
particular promise for treatment of the sleep maintenance difficulties so
common in older age groups (Edinger et al. 2001, 2007; Morin et al. 1999).
Additionally, a study of a brief behavioral intervention focusing on only
behavioral elements of insomnia treatment was found to be robustly
effective (Buysse et al. 2011). Given such findings, behavioral interventions
should be included in treatment plans for older patients with insomnia,
particularly when improper sleep scheduling and other lifestyle factors
contribute to sleep complaints.
Pharmacological Treatment
It is well established that untreated insomnia is associated with significant
adverse effects (Gislason and Almqvist 1987; Krystal 2007; Zammit et al.
1999). Although a number of medications have been demonstrated to have
potent therapeutic effects on insomnia (Table 16–1), the utility of
pharmacotherapy for this purpose has remained controversial primarily
because of concerns about the risks of long-term treatment.
The long-standing view of the medication management of insomnia has
been that tolerance and withdrawal are inevitable with use for longer than
several weeks (National Institutes of Health Consensus Conference 1984).
As a result, treatment for longer periods was discouraged (National
Institutes of Health Consensus Conference 1984). It is important to note
that this view was not empirically based. It is important to note that this
view was not empirically based, given that it was much later that data from
placebo-controlled studies of the treatment of insomnia with these agents
for periods longer than 3 weeks became available. These studies examining
periods up to 1 year of nightly treatment demonstrate that tolerance and
withdrawal are not inevitably associated with longer therapy duration
(Ancoli-Israel et al. 2005; Krystal et al. 2003, 2008; Walsh et al. 2007).
They established the efficacy and safety of the nonbenzodiazepine
eszopiclone 3 mg at bedtime in adults younger than age 65 years in two 6-
month placebo-controlled studies, one of which had a subsequent 6-month
open-label extension phase (Krystal et al. 2003; Walsh et al. 2007). Another
nonbenzodiazepine, zolpidem (10 mg at bedtime), was dosed 3–7 nights per
week for 6 months in younger adults and was found to improve sleep
without evidence of significant adverse effects related to the duration of
therapy (Krystal et al. 2008). The melatonin receptor agonist ramelteon has
also been found to have sustained sleep onset efficacy and safety over 6
months of nightly treatment in a placebo-controlled trial in adults ages 18
years and older (Mayer et al. 2009). Most recently, a yearlong placebo-
controlled study of nightly treatment with the hypocretin/orexin receptor
antagonist suvorexant was carried out in adults at least 18 years of age and
was found to have sustained efficacy and good tolerability (Michelson et al.
2014). This medication has just been approved (August 2014) by the U.S.
Food and Drug Administration (FDA) for prescription treatment of
insomnia in the United States.
Conclusion
Management of sleep disorders in elderly patients is challenging. Although
sleep complaints are not an inevitable consequence of aging, elderly
persons are more prone to primary sleep disorders and medical and
psychiatric conditions that cause sleep difficulties. Therefore, evaluation of
a sleep complaint in an elderly individual should include a thorough workup
to determine whether primary sleep pathology and associated psychiatric
and medical disorders are present. Effective behavioral and medication
treatments exist for treating sleep and circadian rhythm disorders in elderly
patients, but these treatments have significant limitations. More research is
needed to develop and assess nonmedication therapies that are effective in
treating insomnia and normalizing the circadian rhythm. Particularly
promising areas include cognitive-behavioral approaches and exercise
programs.
In addition, research to improve medication treatment is needed. More
medications are needed that can help elderly individuals stay asleep without
causing next-day sedation. Furthermore, medications are needed that do not
cause motor or cognitive impairment or anticholinergic side effects and that
have been evaluated in trials of long-term treatment in older adults. Studies
of the efficacy and safety of antidepressants in the treatment of insomnia in
older adults are also needed.
Key Points
• More than one-half of noninstitutionalized individuals older than age 65
years report chronic sleep difficulties.
• Although disturbed sleep is not an inevitable consequence of aging,
elderly people are at increased risk of experiencing a number of sleep
disorders and are uniquely vulnerable to the consequences of these
disorders, which include insomnia, restless legs syndrome, sleep apnea,
and disorders of circadian rhythm.
• Effective behavioral and medication treatments exist for treating sleep
and circadian rhythm disorders in elderly patients, but more research is
needed to develop improved treatments and to establish the risk-benefit
profiles of some of the most commonly administered therapies.
References
Aldrich MS: Cardinal manifestations of sleep disorders, in Principles and
Practice of Sleep Medicine, 3rd Edition. Edited by Kryger MH, Roth T,
Dement WC. Philadelphia, PA, WB Saunders, 2000, pp 526–534
Allain H, Bentué-Ferrer D, Polard E, et al: Postural instability and
consequent falls and hip fractures associated with use of hypnotics in
the elderly: a comparative review. Drugs Aging 22(9):749–765, 2005
16156679
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric
Association, 2013.
Ancoli-Israel S: Epidemiology of sleep disorders. Clin Geriatr Med
5(2):347–362, 1989 2665916
Ancoli-Israel S, Kripke DF, Mason W: Characteristics of obstructive and
central sleep apnea in the elderly: an interim report. Biol Psychiatry
22(6):741–750, 1987 3593813
Ancoli-Israel S, Kripke DF, Klauber MR, et al: Periodic limb movements in
sleep in community-dwelling elderly. Sleep 14(6):496–500, 1991
1798881
Ancoli-Israel S, Richardson GS, Mangano RM, et al: Long-term use of
sedative hypnotics in older patients with insomnia. Sleep Med
6(2):107–113, 2005 15716214
Asplund R: Sleep and hypnotic use in relation to perceived somatic and
mental health among the elderly. Arch Gerontol Geriatr 31(3):199–205,
2000 11154774
Avidan AY, Fries BE, James ML, et al: Insomnia and hypnotic use, recorded
in the minimum data set, as predictors of falls and hip fractures in
Michigan nursing homes. J Am Geriatr Soc 53(6):955–962, 2005
15935017
Bassetti C, Chervin R: Cerebrovascular diseases, in Principles and Practice
of Sleep Medicine, 3rd Edition. Edited by Kryger MH, Roth T, Dement
WC. Philadelphia, PA, WB Saunders, 2000, pp 1072–1086
Bliwise DL: Sleep in normal aging and dementia. Sleep 16(1):40–81, 1993
8456235
Bliwise DL: Normal aging, in Principles and Practice of Sleep Medicine,
3rd Edition. Edited by Kryger MH, Roth T, Dement WC. Philadelphia,
PA, WB Saunders, 2000, pp 26–42
Bliwise DL: Sleep disorders in Alzheimer’s disease and other dementias.
Clin Cornerstone 6(suppl 1A):S16–S28, 2004 15259536
Bliwise DL, Petta D, Seidel W, et al: Periodic leg movements during sleep
in the elderly. Arch Gerontol Geriatr 4(3):273–281, 1985 4074025
Bliwise DL, Freeman A, Ingram CD, et al: Randomized, double-blind,
placebo-controlled, short-term trial of ropinirole in restless legs
syndrome. Sleep Med 6(2):141–147, 2005 15716217
Bloom HG, Ahmed I, Alessi CA, et al: Evidence-based recommendations
for the assessment and management of sleep disorders in older persons.
J Am Geriatr Soc 57(5):761–789, 2009 19484833
Bootzin RR: A stimulus control treatment for insomnia. Proceedings of the
American Psychological Association 7:395–396, 1972
Brassington GS, King AC, Bliwise DL: Sleep problems as a risk factor for
falls in a sample of community-dwelling adults aged 64–99 years. J
Am Geriatr Soc 48(10): 1234–1240, 2000 11037010
Breslau N, Roth T, Rosenthal L, Andreski P: Sleep disturbance and
psychiatric disorders: a longitudinal epidemiological study of young
adults. Biol Psychiatry 39(6):411–418, 1996 8679786
Buysse DJ, Browman KE, Monk TH, et al: Napping and 24-hour
sleep/wake patterns in healthy elderly and young adults. J Am Geriatr
Soc 40(8):779–786, 1992 1634721
Buysse DJ, Ancoli-Israel S, Edinger JD, et al: Recommendations for a
standard research assessment of insomnia. Sleep 29(9):1155–1173,
2006 17040003
Buysse DJ, Germaine A, Moul DE, et al: Efficacy of brief behavioral
treatment for chronic insomnia in older adults. Arch Int Med
171(10):887–895, 2011
Camargos EF, Louzada LL, Quintas JL, et al: Trazodone improves sleep
parameters in Alzheimer disease patients: a randomized, double-blind,
and placebo-controlled study. Am J Geriatr Psychiatry 22(12):1565–
1574, 2014 24495406
Carskadon MA, Brown ED, Dement WC: Sleep fragmentation in the
elderly: relationship to daytime sleep tendency. Neurobiol Aging
3(4):321–327, 1982 7170049
Chen YY, Sy HN, Wu SL: Zolpidem improves akinesia, dystonia and
dyskinesia in advanced Parkinson’s disease. J Clin Neurosci
15(8):955–956, 2008 18485713
Clarenbach P: Parkinson’s disease and sleep. J Neurol 247 (suppl 4):IV20–
IV23, 2000 1199812
Clark MM: Restless legs syndrome. J Am Board Fam Pract 14(5):368–374,
2001 11572542
Connaughton JJ, Catterall JR, Elton RA, et al: Do sleep studies contribute
to the management of patients with severe chronic obstructive
pulmonary disease? Am Rev Respir Dis 138(2):341–344, 1988
3195833
Czeisler CA, Duffy JF, Shanahan TL, et al: Stability, precision, and near-24-
hour period of the human circadian pacemaker. Science
284(5423):2177–2181, 1999 10381883
Dickel MJ, Mosko SS: Morbidity cut-offs for sleep apnea and periodic leg
movements in predicting subjective complaints in seniors. Sleep
13(2):155–166, 1990 2330474
Dorsey CM, Lee KA, Scharf MB: Effect of zolpidem on sleep in women
with perimenopausal and postmenopausal insomnia: a 4-week,
randomized, multicenter, double-blind, placebo-controlled study. Clin
Ther 26(10):1578–1586, 2004 15598474
Dijk DJ, Duffy JF, Riel E, et al: Altered interaction of circadian and
homeostatic aspects of sleep propensity results in awakening at an
earlier circadian phase in older people. Sleep Research 26:710, 1997
Douglas NJ: Chronic obstructive pulmonary disease, in Principles and
Practice of Sleep Medicine, 3rd Edition. Edited by Kryger MH, Roth T,
Dement WC. Philadelphia, PA, WB Saunders, 2000, pp 965–975
Edinger JD, Wohlgemuth WK, Radtke RA, et al: Cognitive behavioral
therapy for treatment of chronic primary insomnia: a randomized
controlled trial. JAMA 285(14): 1856–1864, 2001 11308399
Edinger JD, Wohlgemuth WK, Krystal AD, et al: Behavioral insomnia
therapy for fibromyalgia patients: a randomized clinical trial. Arch
Intern Med 165(21):2527–2535, 2005 16314551
Edinger JD, Wohlgemuth WK, Radtke RA, et al: Dose-response effects of
cognitive-behavioral insomnia therapy: a randomized clinical trial.
Sleep 30(2):203–212, 2007 17326546
Fava M, McCall WV, Krystal A, et al: Eszopiclone co-administered with
fluoxetine in patients with insomnia coexisting with major depressive
disorder. Biol Psychiatry 59(11):1052–1060, 2006 16581036
Fava M, Asnis GM, Shrivastava RK, et al: Improved insomnia symptoms
and sleep-related next-day functioning in patients with comorbid major
depressive disorder and insomnia following concomitant zolpidem
extended-release 12.5 mg and escitalopram treatment: a randomized
controlled trial. J Clin Psychiatry 72(7):914–928, 2011 21208597
Fawcett J, Scheftner WA, Fogg L, et al: Time-related predictors of suicide
in major affective disorder. Am J Psychiatry 147(9): 1189–1194, 1990
2104515
Foley DJ, Monjan AA, Brown SL, et al: Sleep complaints among elderly
persons: an epidemiologic study of three communities. Sleep
18(6):425–432, 1995 7481413
Gislason T, Almqvist M: Somatic diseases and sleep complaints: an
epidemiological study of 3,201 Swedish men. Acta Med Scand
221(5):475–481, 1987 3496735
Golden RN, Dawkins K, Nicholas L, et al: Trazodone, nefazodone,
bupropion, and mirtazapine, in Textbook of Psychopharmacology, 2nd
Edition. Edited by Schatzberg AF, Nemeroff CB. Washington, DC,
American Psychiatric Press, 1998, pp 251–269
Hall M, Buysse DJ, Dew MA, et al: Intrusive thoughts and avoidance
behaviors are associated with sleep disturbances in bereavement-
related depression. Depress Anxiety 6(3):106–112, 1997 9442984
Hobbs WR, Rall TW, Verdoorn TA: Hypnotics and sedatives: ethanol, in
Goodman and Gilman’s The Pharmacological Basis of Therapeutics,
9th Edition. Edited by Hardman JG, Limbird LE. New York, McGraw-
Hill, 1996, pp 361–398
Hoch CC, Buysse DJ, Reynolds CF 3rd: Sleep and depression in late life.
Clin Geriatr Med 5(2):259–274, 1989 2665910
Juri C, Chaná P, Tapia J, et al: Quetiapine for insomnia in Parkinson
disease: results from an open-label trial. Clin Neuropharmacol
28(4):185–187, 2005 16062098
Koski K, Luukinen H, Laippala P, Kivelä SL: Risk factors for major
injurious falls among the home-dwelling elderly by functional abilities:
a prospective population-based study. Gerontology 44(4):232–238,
1998 9657085
Kryger M, Wang-Weigand S, Zhang J, et al: Effect of ramelteon, a selective
MT(1)/MT (2)-receptor agonist, on respiration during sleep in mild to
moderate COPD. Sleep Breath 12(3):243–250 2008 18060441
Krystal AD: Sleep and psychiatric disorders: future directions. Psychiatr
Clin North Am 29(4):1115–1130, abstract xi, 2006 17118285
Krystal AD: Treating the health, quality of life, and functional impairments
in insomnia. J Clin Sleep Med 3(1):63–72, 2007 17557457
Krystal AD: A compendium of placebo-controlled trials of the
risks/benefits of pharmacologic treatments for insomnia: the empirical
basis for clinical practice. Sleep Med Rev 13(4):265–274, 2009
19153052
Krystal AD, Edinger J, Wohlgemuth W, et al: Sleep in peri-menopausal and
post-menopausal women. Sleep Med Rev 2(4):243–253, 1998
15310495
Krystal AD, Walsh JK, Laska E, et al: Sustained efficacy of eszopiclone
over 6 months of nightly treatment: results of a randomized, double-
blind, placebo-controlled study in adults with chronic insomnia. Sleep
26(7):793–799, 2003 14655910
Krystal AD, Fava M, Rubens R, et al: Evaluation of eszopiclone
discontinuation after cotherapy with fluoxetine for insomnia with
coexisting depression. J Clin Sleep Med 3(1):48–55, 2007 17557453
Krystal AD, Erman M, Zammit GK, et al; ZOLONG Study Group: Long-
term efficacy and safety of zolpidem extended-release 12.5 mg,
administered 3 to 7 nights per week for 24 weeks, in patients with
chronic primary insomnia: a 6-month, randomized, double-blind,
placebo-controlled, parallel-group, multicenter study. Sleep 31(1):79–
90, 2008 18220081
Krystal AD, Durrence HH, Scharf M, et al: Efficacy and safety of doxepin 1
mg and 3 mg in a 12-week sleep laboratory and outpatient trial of
elderly subjects with chronic primary insomnia. Sleep 33(11):1553–
1561, 2010 21102997
Lees AJ, Blackburn NA, Campbell VL: The nighttime problems of
Parkinson’s disease. Clin Neuropharmacol 11(6):512–519, 1988
3233589
Livingston G, Blizard B, Mann A: Does sleep disturbance predict
depression in elderly people? A study in inner London. Br J Gen Pract
43(376):445–448, 1993 8292414
Manber R, Edinger JD, Gress JL, et al: Cognitive behavioral therapy for
insomnia enhances depression outcome in patients with comorbid
major depressive disorder and insomnia. Sleep 31(4):489–495, 2008
18457236
Martin JL, Ancoli-Israel S: Napping in older adults. Sleep Med Clin 1:177–
186, 2006
Mayer G, Wang-Weigand S, Roth-Schechter B, et al: Efficacy and safety of
6-month nightly ramelteon administration in adults with chronic
primary insomnia. Sleep 32(3):351–360, 2009 19294955
McCall WV, Erman M, Krystal AD, et al: A polysomnography study of
eszopiclone in elderly patients with insomnia. Curr Med Res Opin
22(9):1633–1642, 2006 16968566
McCleery J, Cohen DA, Sharpley AL: Pharmacotherapies for sleep
disturbances in Alzheimer’s disease. Cochrane Review. Cochrane
Database Syst Rev (3):CD009178, 2014 24659320
McCurry SM, Gibbons LE, Logsdon RG, et al: Nighttime insomnia
treatment and education for Alzheimer’s disease: a randomized,
controlled trial. J Am Geriatr Soc 53(5):793–802, 2005 15877554
Means MK, Edinger JD: Behavioral treatment of insomnia. Expert Rev
Neurother 2(1): 127–137, 2002 19811022
Menza M, Dopkin RD, Marin H, et al: Treatment of insomnia in
Parkinson’s disease: a controlled trial of eszopiclone and placebo. Mov
Disord 25(11):1708–1714, 2010 20589875
Michelson D, Snyder E, Paradis E, et al: Safety and efficacy of suvorexant
during 1-year treatment of insomnia with subsequent abrupt treatment
discontinuation: a phase 3 randomised, double-blind, placebo-
controlled trial. Lancet Neurol 13(5):461–471, 2014 24680372
Middelkoop HA, Smilde-van den Doel DA, Neven AK, et al: Subjective
sleep characteristics of 1,485 males and females aged 50-93: effects of
sex and age, and factors related to self-evaluated quality of sleep. J
Gerontol A Biol Sci Med Sci 51(3):M108–M115, 1996 8630703
Monk TH: Aging human circadian rhythms: conventional wisdom may not
always be right. J Biol Rhythms 20(4):366–374, 2005 16077155
Montplaisir J, Nicolas A, Denesle R, et al: Restless legs syndrome
improved by pramipexole: a double-blind randomized trial. Neurology
52(5):938–943, 1999 10102409
Morgan K: Sleep and aging, in Treatment of Late-Life Insomnia. Edited by
Lichstein KL, Morin CM. Thousand Oaks, CA, Sage, 2000, pp 3–36
Morgenthaler T, Alessi C, Friedman L, et al; Standards of Practice
Committee; American Academy of Sleep Medicine: Practice
parameters for the use of actigraphy in the assessment of sleep and
sleep disorders: an update for 2007. Sleep 30(4):519–529, 2007
17520797
Morin CM, Stone J, Trinkle D, et al: Dysfunctional beliefs and attitudes
about sleep among older adults with and without insomnia complaints.
Psychol Aging 8(3): 463–467, 1993 8216967
Morin CM, Colecchi C, Stone J, et al: Behavioral and pharmacological
therapies for late-life insomnia: a randomized controlled trial. JAMA
281(11):991–999, 1999 10086433
Morin CM, Bootzin RR, Buysse DJ, et al: Psychological and behavioral
treatment of insomnia: update of the recent evidence (1998–2004).
Sleep 29(11):1398–1414, 2006 17162986
Murciano D, Armengaud MH, Cramer PH, et al: Acute effects of zolpidem,
triazolam and flunitrazepam on arterial blood gases and control of
breathing in severe COPD. Eur Respir J 6(5):625–629, 1993 8519370
Murtagh DR, Greenwood KM: Identifying effective psychological
treatments for insomnia: a meta-analysis. J Consult Clin Psychol
63(1):79–89, 1995 7896994
National Institutes of Health: National Institutes of Health State of the
Science Conference statement on manifestations and management of
chronic insomnia in adults, June 13–15, 2005. Sleep 28(9):1049–1057,
2005 16268373
National Institutes of Health Consensus Conference: Drugs and insomnia:
the use of medications to promote sleep. JAMA 251(18):2410–2414,
1984 6142971
National Institutes of Health Consensus Development Conference
Statement: The treatment of sleep disorders of older people March 26–
28, 1990. Sleep 14(2):169–177, 1991 1866532
Nebes RD, Pollock BG, Halligan EM, et al: Serum anticholinergic activity
and motor performance in elderly persons. J Gerontol A Biol Sci Med
Sci 62(1):83–85, 2007 17301043
Neutel CI, Perry S, Maxwell C: Medication use and risk of falls.
Pharmacoepidemiol Drug Saf 11(2):97–104, 2002 11998544
North American Menopause Society: Treatment of menopause-associated
vasomotor symptoms: position statement of the North American
Menopause Society. Menopause 11(1):11–33, 2004 14716179
Ohayon MM: Epidemiology of insomnia: what we know and what we still
need to learn. Sleep Med Rev 6(2):97–111, 2002 12531146
Ohayon M, Caulet M, Lemoine P: [The elderly, sleep habits and use of
psychotropic drugs by the French population]. Encephale 22(5):337–
350, 1996 9035990
Ohayon MM, Carskadon MA, Guilleminault C, et al: Meta-analysis of
quantitative sleep parameters from childhood to old age in healthy
individuals: developing normative sleep values across the human
lifespan. Sleep 27(7):1255–1273, 2004 15586779
O’Keeffe ST, Gavin K, Lavan JN: Iron status and restless legs syndrome in
the elderly. Age Ageing 23(3):200–203, 1994 8085504
Ozminkowski RJ, Wang S, Walsh JK: The direct and indirect costs of
untreated insomnia in adults in the United States. Sleep 30(3):263–273,
2007 17425222
Physicians’ Desk Reference: 56th Edition. Montvale, NJ, Medical
Economics, 2002
Pilowsky I, Crettenden I, Townley M: Sleep disturbance in pain clinic
patients. Pain 23(1):27–33, 1985 4058926
Pollak CP, Perlick D: Sleep problems and institutionalization of the elderly.
J Geriatr Psychiatry Neurol 4(4):204–210, 1991 1789908
Pollak CP, Perlick D, Linsner JP, et al: Sleep problems in the community
elderly as predictors of death and nursing home placement. J
Community Health 15(2): 123–135, 1990 2355110
Postuma RB, Gagnon JF, Vendette M, et al: Quantifying the risk of
neurodegenerative disease in idiopathic REM sleep behavior disorder.
Neurology 72(15):1296–1300, 2009 19109537
Potter WZ, Manji HK, Rudorfer MV: Tricyclics and tetracyclics, in
Textbook of Psychopharmacology, 2nd Edition. Edited by Schatzberg
AF, Nemeroff CB. Washington, DC, American Psychiatric Press, 1998,
pp 239–250
Prinz PN: Sleep and sleep disorders in older adults. J Clin Neurophysiol
12(2):139–146, 1995 7797628
Prinz PN, Peskind ER, Vitaliano PP, et al: Changes in the sleep and waking
EEGs of nondemented and demented elderly subjects. J Am Geriatr
Soc 30(2):86–93, 1982 7199061
Prinz PN, Vitiello MV, Raskind MA, et al: Geriatrics: sleep disorders and
aging. N Engl J Med 323(8):520–526, 1990 2198467
Reynolds CF 3rd, Frank E, Houck PR, et al: Which elderly patients with
remitted depression remain well with continued interpersonal
psychotherapy after discontinuation of antidepressant medication? Am
J Psychiatry 154(7):958–962, 1997 9210746
Rios Romenets S, Creti L, Fichten C, et al: Doxepin and cognitive
behavioural therapy for insomnia in patients with Parkinson’s disease
—a randomized study. Parkinsonism Relat Disord 19(7):670–675,
2013 23561946
Roehrs T, Zorick F, Sicklesteel J, et al: Age-related sleep-wake disorders at
a sleep disorder center. J Am Geriatr Soc 31(6): 364–370, 1983
6853947
Roth T, Krystal A, Steinberg FJ, et al: Novel sublingual low-dose zolpidem
tablet reduces latency to sleep onset following spontaneous middle-of-
the-night awakening in insomnia in a randomized, double-blind,
placebo-controlled, outpatient study. Sleep 36(2):189–196, 2013
Růzicka E, Roth J, Jech R, et al: Subhypnotic doses of zolpidem oppose
dopaminergic-induced dyskinesia in Parkinson’s disease. Mov Disord
15(4):734–735, 2000 10928588
Sanford JRA: Tolerance of debility in elderly dependants by supporters at
home: its significance for hospital practice. BMJ 3(5981):471–473,
1975 1156826
Schenck CH, Bundlie SR, Mahowald MW: Delayed emergence of a
parkinsonian disorder in 38% of 29 older men initially diagnosed with
idiopathic rapid eye movement sleep behaviour disorder. Neurology
46(2):388–393, 1996 8614500
Smith MT, Perlis ML, Park A, et al: Comparative meta-analysis of
pharmacotherapy and behavior therapy for persistent insomnia. Am J
Psychiatry 159(1):5–11, 2002 11772681
Soares CN, Joffe H, Rubens R, et al: Eszopiclone in patients with insomnia
during perimenopause and early postmenopause: a randomized
controlled trial. Obstet Gynecol 108(6):1402–1410, 2006 17138773
Spielman AJ, Saskin P, Thorpy MJ: Treatment of chronic insomnia by
restriction of time in bed. Sleep 10(1):45–56, 1987 3563247
Steens RD, Pouliot Z, Millar TW, et al: Effects of zolpidem and triazolam
on sleep and respiration in mild to moderate chronic obstructive
pulmonary disease. Sleep 16(4):318–326, 1993 8341892
Stone BM, Turner C, Mills SL, et al: Noise-induced sleep maintenance
insomnia: hypnotic and residual effects of zaleplon. Br J Clin
Pharmacol 53(2):196–202, 2002 11851645
Suzuki M, Okamura T, Shimazu Y, et al: [A study of falls experienced by
institutionalized elderly]. Nippon Koshu Eisei Zasshi 39(12):927–940,
1992 1292738
Trenkwalder C: Sleep dysfunction in Parkinson’s disease. Clin Neurosci
5(2):107–114, 1998 10785836
Vitiello MV, McCurry SM, Shortreed SM, et al: Cognitive-behavioral
treatment for comorbid insomnia and osteoarthritis pain in primary
care: the lifestyles randomized controlled trial. J Am Geriatr Soc
61(6):947–956, 2013 23711168
Walsh JK: Drugs used to treat insomnia in 2002: regulatory-based rather
than evidence-based medicine. Sleep 27(8):1441–1442, 2004 15683131
Walsh JK, Muehlbach MJ, Lauter SA, et al: Effects of triazolam on sleep,
daytime sleepiness, and morning stiffness in patients with rheumatoid
arthritis. J Rheumatol 23(2):245–252, 1996 8882027
Walsh JK, Krystal AD, Amato DA, et al: Nightly treatment of primary
insomnia with eszopiclone for six months: effect on sleep, quality of
life, and work limitations. Sleep 30(8):959–968, 2007 17702264
Weissman MM, Greenwald S, Niño-Murcia G, Dement WC: The morbidity
of insomnia uncomplicated by psychiatric disorders. Gen Hosp
Psychiatry 19(4):245–250, 1997 9327253
White DP: Central sleep apnea, in Principles and Practice of Sleep
Medicine, 3rd Edition. Edited by Kryger MH, Roth T, Dement WC.
Philadelphia, WB Saunders, 2000, pp 827–839
Wohlgemuth WK, Edinger JD: Sleep restriction therapy, in Treatment of
Late-Life Insomnia. Edited by Lichstein KL, Morin CM. Thousand
Oaks, CA, Sage, 2000, pp 147–184
Zammit GK, Weiner J, Damato N, et al: Quality of life in people with
insomnia. Sleep 22(Suppl 2):S379–S385, 1999 10394611
Suggested Readings
Bloom HG, Ahmed I, Alessi CA et al: Evidence-based recommendations
for the assessment and management of sleep disorders in older persons.
J Am Geriatr Soc 57(5):761–789, 2009
Buysee DJ, Germaine A, Moul DE, et al: Efficacy of brief behavioral
treatment for chronic insomnia in older adults. Arch Intern Med
171(10):887–895, 2011
CHAPTER 17
Alcohol and drug misuse are associated with a wide array of negative
physical and mental health outcomes that are exacerbated with advancing
age, such as functional and cognitive decline, compromised immune
function, and depression; however, relatively little work has examined the
correlates and consequences of substance use among older adults.
Accordingly, substance misuse in later life, which encompasses misuse of
alcohol and illicit, prescription, and over-the-counter drugs, has been called
an “invisible epidemic” (Widlitz and Marin 2002). Epidemiological work,
which has focused on younger populations, demonstrates that beginning in
the middle to late 20s, overall rates of alcohol and illicit drug use begin to
decline, with the majority of older adults reporting no substance use.
Nevertheless, changes in demographic and cohort trends suggest that
substance misuse in later life is a pressing public health matter and that
older adults represent a group in growing need of specialized substance
treatment programs and services (Gfroerer et al. 2003).
With the aging of the U.S. population and the resultant increase in the
proportion of adults living to advanced ages, a concomitant increase has
occurred in the number of older adults who misuse alcohol and drugs. Not
only are older adults the fastest growing segment of the U.S. population, but
in the next several decades the aging population will be composed primarily
of “baby boomers,” individuals born between the years 1946 and 1964. The
aging of the baby boom cohort poses unique challenges to providers; in
addition to reporting higher rates of illicit drug and alcohol use and
addiction than earlier cohorts, the baby boom cohort also is significantly
larger than previous cohorts (Han et al. 2009a).
The potential public health impact of these demographic trends is
highlighted by examining changes in rates of substance use and misuse in
the last several decades. For example, it has been estimated that from the
early 1990s until 2002, the prevalence of alcohol abuse or dependence
tripled to 3.1% among adults ages 65 years and older (Grant et al. 2004).
Heavy and binge drinking among adults older than 65 years also has
increased, with recent reports citing rates near 10.2% (Substance Abuse and
Mental Health Services Administration 2013) (Figure 17–1). Reports of
substance use among the baby boomers are notably higher; 29.3% of adults
ages 50–54 years were heavy or binge drinkers in 2012 (see Figure 17–1),
and rates of illicit drug use among those in this age group increased from
3.4% to 7.2% from 2002 to 2012 (Substance Abuse and Mental Health
Services Administration 2013) (Figure 17–2).
FIGURE 17–1. Current, binge, and heavy alcohol use across the life
span: 2012.
Source. Reprinted from Figure 3.1 (Current, binge, and heavy alcohol use among persons aged 12 or
older, by age: 2012) in Substance Abuse and Mental Health Services Administration: Results from
the 2012 National Survey on Drug Use and Health: Summary of National Findings, NSDUH Series
H-46, HHS Publication No. (SMA) 13-4795. Rockville, MD, Substance Abuse and Mental Health
Services Administration, 2013. Available at:
http://www.samhsa.gov/data/NSDUH/2012SummNatFindDetTables/NationalFindings/NSDUHresult
s2012.htm#ch3.1.1. Accessed January 15, 2014.
Alcohol
Even though alcohol misuse is often underreported and thus underestimated
in later life, epidemiological work suggests that alcohol problems are
common among older adults. For instance, the most recent National Survey
on Drug Use and Health found that 53.1% of adults ages 60–64 years
endorsed current alcohol use in the past month, 14.3% endorsed bingeing
behavior, and 4.3% endorsed heavy use (Substance Abuse and Mental
Health Services Administration 2013). Although rates of use were lower for
those age 65 years or older, they were nonetheless significant: 41.2% of
respondents reported past-month alcohol use, 8.2% bingeing, and 2.0%
heavy use (see Figure 17–1). With respect to alcohol use disorder,
epidemiological work has found that 4.1% of adults ages 65–75 years (8.1%
of men and 1.0% of women) reported a lifetime history of alcohol use
disorder, whereas 1.6% of adults ages 75 years and older reported a history
of alcohol use disorder (Gum et al. 2009).
Because substance misuse is more likely to be present in health care
settings, rates of alcohol problems are higher among clinical than among
community-based samples. For example, in their screening program of over
12,000 primary care patients, Barry et al. (1998) found that 5% of patients
screened positive for at-risk or problem drinking, as measured by alcohol
consumption, binge drinking, or the presence of alcohol-related problems.
Similarly, Callahan and Tierney (1995) found that 10.6% of their sample of
3,954 primary care patients ages 60 and older met criteria for problem
drinking. In a more recent study of older adults in primary care settings,
Kirchner et al. (2007) reported that of the 24,863 individuals screened,
21.5% drank within the recommended levels (1–7 drinks per week),
whereas 4.1% were at-risk drinkers (8–14 drinks per week), and 4.5% were
heavy (>14 drinks per week) or binge drinkers. Rates of abuse and
dependence appear to be particularly high among patients in mental health
clinics and nursing homes. In their study of 140 patients enrolled in a
geriatric mental health outpatient clinic, Holroyd and Duryee (1997) found
that 8.6% of patients met DSM-IV criteria for alcohol dependence.
Furthermore, Oslin et al. (1997b) found that 29% of male nursing home
residents had a lifetime diagnosis of alcohol abuse or dependence, with
10% of residents meeting criteria for abuse or dependence within 1 year of
admission to the home. Moreover, a census analysis of Department of
Veterans Affairs (VA) nursing home admissions data found that 29% and
15% of residents ages 57–64 and 65–72, respectively, had a diagnosis of an
alcohol use disorder (Lemke and Schaefer 2010).
Source. DSM-5 criteria reprinted from Diagnostic and Statistical Manual of Mental Disorders, 5th
Edition. Washington, DC, American Psychiatric Association, 2013. Used with permission. Copyright
© 2013 American Psychiatric Association.
Illicit Drugs
Unlike alcohol and prescription or over-the-counter medication use, illicit
drug use among older adults is rare. Based on data from the Epidemiologic
Catchment Area study, analyses in the early 1990s estimated that the
lifetime prevalence rate of drug abuse and dependence was 0.12% for older
men and 0.06% for older women, whereas the lifetime history of illicit drug
use was 2.88% for men and 0.66% for women (Anthony and Helzer 1991).
More recent work, however, suggests that illicit substance use is more
common among older adults than previous estimates suggest. For example,
examination of national trends of drug use among adults ages 50–59 years
revealed that 9.4% of adults used an illicit or non-medical drug in the
previous year (Han et al. 2009a). The rate of illicit substance use (mainly
heroin and cocaine) was reported to be 45% among adults ages 55 years and
older admitted to substance abuse treatment facilities (Lofwall et al. 2008).
Finally, according to the most recent National Survey on Drug Use and
Health, the percentage of adults ages 55–59 years using illicit drugs in the
past month increased from 1.9% in 2002 to 6.6% in 2012 (Substance Abuse
and Mental Health Services Administration 2013). Although 6.6% may not
appear to be a significant proportion of older adults, it is important to keep
in mind that the baby boomers were among the only age groups that showed
notable increases in illicit substance use during the designated time period
(see Figure 17–2). Thus, rates of illicit substance use and abuse among
older adults will likely continue to rise in the next several decades because
of the aging of the baby boom cohort.
As this last point suggests, it important to recognize that the utility of the
questionnaires may vary as a function of the category of drinking (e.g., at-
risk drinking, alcohol use disorder) that is being assessed. In one
comparison of the relative sensitivity and specificity of self-administered
versions of the CAGE and AUDIT, Bradley et al. (1998) demonstrated that
an augmented version of the CAGE (i.e., a measure including the CAGE
items, the first two items of the AUDIT, and the question “Have you ever
had a drinking problem?”) performed better than the traditional CAGE and
AUDIT when screening for active substance use. However, the AUDIT was
superior to both versions of the CAGE in identifying heavy drinkers.
Finally, although both the augmented CAGE and the AUDIT were effective
in identifying both heavy drinkers and those actively abusing or dependent
on alcohol, the AUDIT performed better. In sum, these results suggest that
the selection of screening instruments used in clinical practice should be
driven by the goals of screening. For example, when screening for both
heavy drinking and alcohol use disorder, clinicians might consider using the
AUDIT or AUDIT-C as opposed to the CAGE (Aertgeerts et al. 2001;
Rubinsky et al. 2013).
Following administration of a screening instrument such as the AUDIT
or CAGE, clinicians can ask follow-up questions about consequences,
health risks, and social/family issues related to substance use. In accordance
with DSM-5 criteria, to assess severity of alcohol use disorder, the clinician
should ask questions about alcohol-related problems, a history of failed
attempts to stop or to cut back, and withdrawal symptoms (e.g., anxiety,
tremors, sleep disturbance). The use of a substance abuse assessment
instrument such as DSM-5 criteria can assist clinicians and researchers by
providing a structured approach to assessment as well as a checklist of
items that can be evaluated across older adults. Furthermore, such
assessments can inform clinicians’ decision making and help determine
whether specialized alcohol treatment might be needed.
Although the screening assessments described in this section focus on
alcohol use, there are also screening instruments for drug consumption,
including a modified version of the CAGE (Hinkin et al. 2001) and the
Drug and Alcohol Problem Assessment for Primary Care (DAPA-PC;
Nemes et al. 2004). Although further work is needed to determine the
validity of these instruments, initial findings are positive.
Psychosocial Treatments
The literature regarding the efficacy of psychological therapies specifically
for the treatment of substance use disorder in older adulthood is sparse. In
one study of older veterans with substance abuse problems, Schonfeld et al.
(2000) found that individuals who completed 16 weeks of a group
intervention for relapse prevention were more likely than noncompleters to
abstain at 6-month follow-up. Using cognitive-behavioral and self-
management approaches, the group sessions included modules on coping
with factors such as social problems, loneliness, depression, and anxiety
and on dealing with high-risk situations for relapse. In a more recent
treatment study, participants ages 50 years and older who completed an 18-
session program that also incorporated both cognitive-behavioral and self-
management approaches evidenced significant improvement in their social
functioning, decreased use of nonmedical prescription drugs and alcohol,
and reduced binge drinking at 6-month follow-up (Outlaw et al. 2012).
Twelve-Step Programs
A large proportion of community-based and residential treatment programs
incorporate the traditional 12-step peer support model of recovery and
rehabilitation. Originally developed by Alcoholics Anonymous (AA) and
later adapted by groups such as Narcotics Anonymous, the 12-step model
involves group support and encouragement to help members achieve and
maintain sobriety. Participants share their experiences with one another and
follow the 12 steps, which include admitting to one’s addiction, recognizing
the influence of a greater power as a source of strength, and acknowledging
and atoning for past mistakes (Alcoholics Anonymous 2004).
Although self-help groups have been associated with positive outcomes
for many individuals, findings regarding rates of group engagement and
outcomes among older individuals remain mixed. In their matched
comparison of older versus younger and middle-aged adults who
participated in age-integrated residential treatment, Lemke and Moos
(2003a) found that older patients engaged in 12-step programs as frequently
as their younger and middle-aged counterparts when assessed at follow-up.
Results also indicated that more involvement in self-help groups
posttreatment was associated with better outcomes across all three age
groups.
Similarly, an investigation of patients who had completed an outpatient
treatment program for chemical dependency yielded no age group
differences in AA affiliation 5 years posttreatment (Satre et al. 2004a).
Upon examination of a subset of participants in the sample who reported
attending 12-step meetings in the prior year, no age group differences in the
actual number of meetings attended emerged as significant. However,
despite the fact that rates of attendance appeared to be comparable across
age groups, the depth of involvement differed; older adults were less likely
than middle-aged adults to self-identify as being a 12-step group member
and were less likely than younger and middle-aged adults to report calling a
fellow group member for help. Comparable results were observed in
examining 1-month postdischarge outcomes among alcohol-dependent
patients admitted to a 12-step residential rehabilitation program (Oslin et al.
2005). Although rates of postdischarge abstinence and AA attendance did
not differ across middle-aged and older adults, older adults were
significantly less likely to contact a sponsor. Furthermore, older adults were
less likely than middle-aged adults to engage in formal aftercare (31.2% vs.
56.4%).
Taken together, these findings highlight the importance of more careful
examination of factors that may be related to 12-step program attendance,
degree of engagement, and outcomes among older adults. These factors
include, but are not limited to, perceived stigma, level of comfort regarding
disclosure of personal information in group settings, degree to which age-
relevant issues are addressed during group meetings, and logistic barriers
such as lack of transportation and health problems that may preclude older
adults from attending group sessions and engaging with sponsors (Oslin et
al. 2005; Satre et al. 2004a).
Pharmacotherapy
Until relatively recently, the long-term treatment of older alcohol-dependent
adults did not involve the use of pharmacological agents. Although
disulfiram was originally the only medication approved for the treatment of
alcohol dependence, it was seldom used in older patients because of the
potential for adverse effects. In 1995, the opioid antagonist naltrexone
became the first pharmacological agent approved by the U.S. Food and
Drug Administration in over 50 years for use in the treatment of alcohol
dependence. Approval of the drug was based on findings from clinical trials
that showed that naltrexone was safe and effective in preventing relapse and
reducing alcohol cravings (O’Malley et al. 1992; Volpicelli et al. 1992).
Although these original clinical trials used samples of middle-aged adults,
naltrexone also has been shown to be effective among older adults. For
example, results from a double-blind, placebo-controlled, randomized trial
demonstrated that among older veterans ages 50–70 years, half as many
naltrexone-treated subjects relapsed to significant drinking when compared
with those treated with placebo (Oslin et al. 1997a).
Acamprosate has emerged as another promising agent in the treatment of
alcohol dependence (Maisel et al. 2013; Rösner et al. 2008). Although the
exact action of acamprosate is still unclear, it is believed to reduce
glutamate response (Kennedy et al. 2010). The clinical evidence favoring
acamprosate is impressive. Sass et al. (1996), for example, found that 43%
of alcohol-dependent subjects treated with acamprosate were abstinent at
the conclusion of their 48-week randomized, placebo-controlled trial
compared with 21% in the placebo group. Similar findings for the favorable
association between acamprosate treatment and abstinence have been
observed in other randomized trials (Baltieri and De Andrade 2004).
Moreover, meta-analytic work supports the notion that although
acamprosate might be considered more effective in promoting and
sustaining abstinence, naltrexone may be more effective in reducing heavy
drinking and craving (Maisel et al. 2013). Unfortunately, no studies of the
efficacy or safety of acamprosate among older patients have been conducted
to date.
Future Directions
Substance misuse among older adults represents a pressing public health
issue, both now and for years to come. In light of changes in demographic
and cohort trends, recent years have seen an increase in the number of older
individuals who misuse or abuse alcohol and drugs. Moreover, there is a
growing awareness that older adults often engage in at-risk or problem
substance use. Nevertheless, individuals in need of treatment or at risk for
future problems often go unidentified and untreated. Therefore, research
and clinical efforts aimed at improving screening efforts and identifying
system-, provider-, and patient-level factors that may interfere with
screening and referral processes for older adults at risk are warranted. In
this vein, a better understanding among clinicians and patients of
recommended drinking levels and the risks associated with moderate to
heavy alcohol consumption is needed, particularly in light of the high
prevalence of co-occurring medical and psychiatric problems in this age
group. Clinicians also should ensure that screening becomes a part of
routine practice when caring for their older patients.
Furthermore, because both provider recommendations and patient
engagement are influenced, in part, by the availability of effective
treatment, better dissemination of information regarding currently available
and efficacious treatments for at-risk use and substance use disorder is
needed. It is important to note, however, that treatment studies in addiction
have traditionally excluded patients older than age 65, resulting in a gap in
knowledge regarding treatment outcomes and an understanding of the
neurobiology of addiction in older individuals. Thus, research endeavors
should continue to focus on developing more effective treatments for
substance misuse in later life, taking into consideration and empirically
assessing the various factors (e.g., patient-, treatment-, and system-related
factors) that may moderate treatment engagement and outcomes. Along
these lines, more formal research that focuses on the relative efficacy of
various treatment modalities, specifically among older adults, is needed.
Finally, given that these issues are particularly relevant to older adults,
future work may benefit from examining nutrition, vitamin
supplementation, and comorbid medical and psychiatric illness, both as foci
for treatment and as aspects of health that may be complicated by substance
use.
Key Points
• Although demographic and cohort trends suggest that rates of substance
misuse among older adults are increasing, the misuse of alcohol and/or
drugs among this group remains largely underrecognized and
undertreated. Accordingly, substance misuse in later life has been
referred to as an “invisible epidemic.”
• Proper screening, diagnosis, and treatment of older adults with drug
and/or alcohol problems require an understanding of both age-specific
guidelines and the full range of substance use behavior seen among older
adults.
• In light of physiological changes that accompany aging, it is
recommended that adults ages 65 and older should consume no more
than an average of 1 standard drink per day.
• The use of multiple pharmaceutical drugs is prevalent in older
adulthood, and thus the risk of misusing prescription and over-the-
counter medications increases with age. Psychotherapeutic medications,
in particular, should be closely monitored, because they are subject to
improper use and can lead to negative health outcomes and interactions
when used alone or in combination with other drugs and alcohol.
• Factors such as medical comorbidity, history of past use, gender, and
social and family environment are related to increased late-life
vulnerability to substance misuse and the maintenance of problematic
substance use patterns.
• Diagnostic criteria and symptoms for alcohol and drug misuse are not
easily applied to older adults, because they are often confounded with
symptoms of comorbid medical illnesses. Thus, when diagnosing and
treating older adults, it is important that providers be able to distinguish
between symptoms of substance misuse and those stemming from
comorbid conditions.
• Routine screening allows for the identification not only of older adults
with substance use disorder, but also of those who are at risk of misusing
drugs and alcohol. Proper screening also helps determine needs for
additional assessment and/or intervention. Standardized brief, low-cost
assessments exist that can be used to assess the quantity and frequency
of alcohol and drug use in clinical practice.
• A variety of treatments for substance misuse, such as brief interventions,
psychotherapy, 12-step programs, and pharmacotherapy, have been
shown to be effective among older adults.
• Special attention should be paid to co-occurrence of problematic
substance use and other medical and psychiatric conditions (e.g.,
depression, dementia, sleep disturbance) because such comorbidity may
affect the course, treatment, and prognosis of both conditions.
References
Abramson JL, Williams SA, Krumholz HM, et al: Moderate alcohol
consumption and risk of heart failure among older persons. JAMA
285(15):1971–1977, 2001 11308433
Aertgeerts B, Buntinx F, Ansoms S, et al: Screening properties of
questionnaires and laboratory tests for the detection of alcohol abuse or
dependence in a general practice population. Br J Gen Pract
51(464):206–217, 2001 11255902
Alcoholics Anonymous: Twelve Steps and Twelve Traditions. New York,
Alcoholics Anonymous, 2004
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Publishing, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Publishing, 2013
Anthony JC, Helzer JE: Syndromes of drug abuse and dependence, in
Psychiatric Disorders in America: The Epidemiologic Catchment Area
Study. Edited by Robins LN, Regier DA. New York, Free Press, 1991,
pp 116–154
Baltieri DA, De Andrade AG: Acamprosate in alcohol dependence: a
randomized controlled efficacy study in a standard clinical setting. J
Stud Alcohol 65(1):136–139, 2004 15000513
Barry KL, Fleming MF: Computerized administration of alcoholism
screening tests in a primary care setting. J Am Board Fam Pract
3(2):93–98, 1990 2185611
Barry KL, Blow FC, Walton MA, et al: Elder-specific brief alcohol
intervention: 3-month outcomes. Alcohol Clin Exp Res 22:30A, 1998
Bartels SJ, Coakley EH, Zubritsky C, et al: PRISM-E Investigators:
Improving access to geriatric mental health services: a randomized trial
comparing treatment engagement with integrated versus enhanced
referral care for depression, anxiety, and at-risk alcohol use. Am J
Psychiatry 161(8):1455–1462, 2004 15285973
Berks J, McCormick R: Screening for alcohol misuse in elderly primary
care patients: a systematic literature review. Int Psychogeriatr
20(6):1090–1103, 2008 18538045
Blazer DG, Wu LT: The epidemiology of substance use and disorders
among middle aged and elderly community adults: national survey on
drug use and health. Am J Geriatr Psychiatry 17(3):237–245, 2009a
19454850
Blazer DG, Wu LT: Nonprescription use of pain relievers by middle-aged
and elderly community-living adults: National Survey on Drug Use and
Health. J Am Geriatr Soc 57(7):1252–1257, 2009b 19486199
Blow FC: (Consensus Panel Chair): Substance Abuse Among Older Adults.
Treatment Improvement Series Protocol (TIP) Series No. 26. Center for
Substance Abuse Treatment. Rockville, MD, U.S. Department of
Health and Human Services, 1998
Bradley KA, Bush KR, McDonell MB, et al: Screening for problem
drinking: comparison of CAGE and AUDIT. J Gen Intern Med 13:379–
388, 1998
Brower KJ, Mudd S, Blow FC, et al: Severity and treatment of alcohol
withdrawal in elderly versus younger patients. Alcohol Clin Exp Res
18(1):196–201, 1994 8198220
Bryson CL, Mukamal KJ, Mittleman MA, et al: The association of alcohol
consumption and incident heart failure: the Cardiovascular Health
Study. J Am Coll Cardiol 48(2):305–311, 2006 16843180
Buchsbaum DG, Buchanan RG, Welsh J, et al: Screening for drinking
disorders in the elderly using the CAGE questionnaire. J Am Geriatr
Soc 40(7):662–665, 1992 1607581
Bush K, Kivlahan DR, McDonell MB, et al: The AUDIT alcohol
consumption questions (AUDIT-C): an effective brief screening test for
problem drinking. Ambulatory Care Quality Improvement Project
(ACQUIP). Alcohol Use Disorders Identification Test. Arch Intern
Med 158(16):1789–1795, 1998 9738608
Bushardt RL, Massey EB, Simpson TW, et al: Polypharmacy: misleading,
but manageable. Clin Interv Aging 3(2):383–389, 2008 18686760
Callahan CM, Tierney WM: Health services use and mortality among older
primary care patients with alcoholism. J Am Geriatr Soc 43(12):1378–
1383, 1995 7490389
Cawthon PM, Fink HA, Barrett-Connor E, et al; Osteoporotic Fractures in
Men Research Group: Alcohol use, physical performance, and
functional limitations in older men. J Am Geriatr Soc 55(2):212–220,
2007 17302657
Charlson F, Degenhardt L, McLaren J, et al: A systematic review of
research examining benzodiazepine-related mortality.
Pharmacoepidemiol Drug Saf 18(2):93–103, 2009 19125401
Chou KL, Mackenzie CS, Liang K, et al: Three-year incidence and
predictors of first-onset of DSM-IV mood, anxiety, and substance use
disorders in older adults: results from Wave 2 of the National
Epidemiologic Survey on Alcohol and Related Conditions. J Clin
Psychiatry 72(2):144–155, 2011 21382305
Closser MH, Blow FC: Recent advances in addictive disorders. Special
populations. Women, ethnic minorities, and the elderly. Psychiatr Clin
North Am 16(1): 199–209, 1993 8456045
Conigliaro J, Kraemer K, McNeil M: Screening and identification of older
adults with alcohol problems in primary care. J Geriatr Psychiatry
Neurol 13(3):106–114, 2000 11001132
Conwell Y: Suicide in elderly patients, in Diagnosis and Treatment of
Depression in Late Life. Edited by Schneider LS, Reynolds CF,
Lebowitz BD, et al. Washington, DC, American Psychiatric Press,
1991, pp 397–418
Dawson DA, Grant BF, Ruan WJ: The association between stress and
drinking: modifying effects of gender and vulnerability. Alcohol
Alcohol 40(5):453–460, 2005a 15972275
Dawson DA, Grant BF, Stinson FS, et al: Effectiveness of the derived
Alcohol Use Disorders Identification Test (AUDIT-C) in screening for
alcohol use disorders and risk drinking in the U.S. general population.
Alcohol Clin Exp Res 29(5):844–854, 2005b 15897730
Djoussé L, Biggs ML, Mukamal KJ, et al: Alcohol consumption and type 2
diabetes among older adults: the Cardiovascular Health Study. Obesity
(Silver Spring) 15(7):1758–1765, 2007 17636094
Dowling GJ, Weiss SR, Condon TP: Drugs of abuse and the aging brain.
Neuropsychopharmacology 33(2):209–218, 2008 17406645
Fink A, Morton SC, Beck JC, et al: The alcohol-related problems survey:
identifying hazardous and harmful drinking in older primary care
patients. J Am Geriatr Soc 50(10):1717–1722, 2002 12366628
Finlayson RE, Hurt RD, Davis LJ Jr, et al: Alcoholism in elderly persons: a
study of the psychiatric and psychosocial features of 216 inpatients.
Mayo Clin Proc 63(8):761–768, 1988 3398595
George LK, Landerman R, Blazer DG, et al: Cognitive impairment, in
Psychiatric Disorders in America: The Epidemiologic Catchment Area
Study. Edited by Robins LN, Regier DA. New York, Free Press, 1991,
pp 291–327
Gfroerer J, Penne M, Pemberton M, et al: Substance abuse treatment need
among older adults in 2020: the impact of the aging baby-boom cohort.
Drug Alcohol Depend 69(2):127–135, 2003 12609694
Golden AG, Preston RA, Barnett SD, et al: Inappropriate medication
prescribing in homebound older adults. J Am Geriatr Soc 47(8):948–
953, 1999 10443855
Gomberg ES: Treatment for alcohol-related problems: special populations:
research opportunities. Recent Dev Alcohol 16: 313–333, 2003
12638644
Grant BF, Dawson DA, Stinson FS, et al: The 12-month prevalence and
trends in DSM-IV alcohol abuse and dependence: United States, 1991–
1992 and 2001–2002. Drug Alcohol Depend 74(3):223–234, 2004
15194200
Gum AM, King-Kallimanis B, Kohn R: Prevalence of mood, anxiety, and
substance-abuse disorders for older Americans in the National
Comorbidity Survey-Replication. Am J Geriatr Psychiatry 17(9):769–
781, 2009 19700949
Hajjar ER, Cafiero AC, Hanlon JT: Polypharmacy in elderly patients. Am J
Geriatr Pharmacother 5(4):345–351, 2007 18179993
Han B, Gfroerer JC, Colliver JD: An Examination of Trends in Illicit Drug
Use Among Adults Aged 50 to 59 in the United States. Rockville, MD,
Substance Abuse and Mental Health Services Administration, Office of
Applied Studies, 2009a
Han B, Gfroerer JC, Colliver JD, Penne MA: Substance use disorder among
older adults in the United States in 2020. Addiction 104(1):88–96,
2009b 19133892
Hinkin CH, Castellon SA, Dickson-Fuhrman E, et al: Screening for drug
and alcohol abuse among older adults using a modified version of the
CAGE. Am J Addict 10(4):319–326, 2001 11783746
Hock B, Schwarz M, Domke I, et al: Validity of carbohydrate-deficient
transferrin (%CDT), gamma-glutamyltransferase (gamma-GT) and
mean corpuscular erythrocyte volume (MCV) as biomarkers for
chronic alcohol abuse: a study in patients with alcohol dependence and
liver disorders of non-alcoholic and alcoholic origin. Addiction
100(10):1477–1486, 2005 16185209
Holroyd S, Duryee JJ: Substance use disorders in a geriatric psychiatry
outpatient clinic: prevalence and epidemiologic characteristics. J Nerv
Ment Dis 185(10):627–632, 1997 9345253
Jonas DE, Garbutt JC, Amick HR, et al: Behavioral counseling after
screening for alcohol misuse in primary care: a systematic review and
meta-analysis for the U.S. Preventive Services Task Force. Ann Intern
Med 157(9):645–654, 2012 23007881
Kashner TM, Rodell DE, Ogden SR, et al: Outcomes and costs of two VA
inpatient treatment programs for older alcoholic patients. Hosp
Community Psychiatry 43(10):985–989, 1992 1328022
Katz IR, Sands LP, Bilker W, et al: Identification of medications that cause
cognitive impairment in older people: the case of oxybutynin chloride.
J Am Geriatr Soc 46(1):8–13, 1998 9434659
Kennedy WK, Leloux M, Kutscher EC, et al: Acamprosate. Expert Opin
Drug Metab Toxicol 6(3):363–380, 2010 20163323
Kirchner JE, Zubritsky C, Cody M, et al: Alcohol consumption among
older adults in primary care. J Gen Intern Med 22(1): 92–97, 2007
17351846
Klatsky AL: Alcohol and cardiovascular health. Physiol Behav 100(1):76–
81, 2010 20045009
Lader M: Benzodiazepine harm: how can it be reduced? Br J Clin
Pharmacol 77(2):295–301, 2014 22882333
Lang I, Wallace RB, Huppert FA, et al: Moderate alcohol consumption in
older adults is associated with better cognition and well-being than
abstinence. Age Ageing 36(3):256–261, 2007 17353234
Leigh BC: Using daily reports to measure drinking and drinking patterns. J
Subst Abuse 12(1-2):51–65, 2000 11288474
Lemke S, Moos RH: Outcomes at 1 and 5 years for older patients with
alcohol use disorders. J Subst Abuse Treat 24(1):43–50, 2003a
12646329
Lemke S, Moos RH: Treatment and outcomes of older patients with alcohol
use disorders in community residential programs. J Stud Alcohol
64(2):219–226, 2003b 12713195
Lemke SP, Schaefer JA: Recent changes in the prevalence of psychiatric
disorders among VA nursing home residents. Psychiatr Serv 61(4):356–
363, 2010 20360274
Lemke S, Schutte KK, Brennan PL, et al: Gender differences in social
influences and stressors linked to increased drinking. J Stud Alcohol
Drugs 69(5):695–702, 2008 18781244
Lemmens P, Tan ES, Knibbe RA: Measuring quantity and frequency of
drinking in a general population survey: a comparison of five indices. J
Stud Alcohol 53(5):476–486, 1992 1405641
Lofwall MR, Schuster A, Strain EC: Changing profile of abused substances
by older persons entering treatment. J Nerv Ment Dis 196(12):898–
905, 2008 19077857
Madhusoodanan S, Bogunovic OJ: Safety of benzodiazepines in the
geriatric population. Expert Opin Drug Saf 3(5):485–493, 2004
15335303
Maisel NC, Blodgett JC, Wilbourne PL, et al: Meta-analysis of naltrexone
and acamprosate for treating alcohol use disorders: when are these
medications most helpful? Addiction 108(2):275–293, 2013 23075288
Mallet L, Spinewine A, Huang A: The challenge of managing drug
interactions in elderly people. Lancet 370(9582):185–191, 2007
17630042
Maust DT, Oslin DW, Marcus SC: Effect of age on the profile of
psychotropic users: results from the 2010 National Ambulatory
Medical Care Survey. J Am Geriatr Soc 62(2):358–364, 2014
Mayfield D, McLeod G, Hall P: The CAGE questionnaire: validation of a
new alcoholism screening instrument. Am J Psychiatry 131(10):1121–
1123, 1974 4416585
Miller W, Rollnick S: Motivational Interviewing: Preparing People to
Change Addictive Behavior. New York, Guilford, 1991
Moeller FG, Gillin JC, Irwin M, et al: A comparison of sleep EEGs in
patients with primary major depression and major depression
secondary to alcoholism. J Affect Disord 27(1):39–42, 1993 8432959
Moore AA, Beck JC, Babor TF, et al: Beyond alcoholism: identifying older,
at-risk drinkers in primary care. J Stud Alcohol 63(3):316–324, 2002
12086132
Moore AA, Gould R, Reuben DB, et al: Longitudinal patterns and
predictors of alcohol consumption in the United States. Am J Public
Health 95(3):458–465, 2005 15727977
Moore AA, Whiteman EJ, Ward KT: Risks of combined alcohol/medication
use in older adults. Am J Geriatr Pharmacother 5(1):64–74, 2007
17608249
Moore AA, Blow FC, Hoffing M, et al: Primary care-based intervention to
reduce at-risk drinking in older adults: a randomized controlled trial.
Addiction 106(1):111–120, 2011 21143686
Moos RH, Schutte K, Brennan P, et al: Ten-year patterns of alcohol
consumption and drinking problems among older women and men.
Addiction 99(7):829–838, 2004 15200578
Moos RH, Schutte KK, Brennan PL, Moos BS: Older adults’ alcohol
consumption and late-life drinking problems: a 20-year perspective.
Addiction 104(8):1293–1302, 2009 19438836
Mukamal KJ, Mackey RH, Kuller LH, et al: Alcohol consumption and
lipoprotein subclasses in older adults. J Clin Endocrinol Metab
92(7):2559–2566, 2007a 17440017
Mukamal KJ, Robbins JA, Cauley JA, et al: Alcohol consumption, bone
density, and hip fracture among older adults: the cardiovascular health
study. Osteoporos Int 18(5):593–602, 2007b 17318666
National Institute on Alcohol Abuse and Alcoholism: Alcohol and sleep.
Alcohol Alert No. 41 (July). Bethesda, MD, U.S. Department of Health
and Human Services, Public Health Services, National Institutes of
Health, 1998
National Institute on Alcohol Abuse and Alcoholism: Module 10C: Older
adults and alcohol problems. Bethesda, MD, U.S. Department of
Health and Human Services, Public Health Services, National Institutes
of Health, 2005
Nemes S, Rao PA, Zeiler C, et al: Computerized screening of substance
abuse problems in a primary care setting: older vs. younger adults. Am
J Drug Alcohol Abuse 30(3):627–642, 2004 15540497
O’Donnell A, Anderson P, Newbury-Birch D, et al: The impact of brief
alcohol interventions in primary healthcare: a systematic review of
reviews. Alcohol Alcohol 49(1):66–78, 2014 24232177
O’Malley SS, Jaffe AJ, Chang G, et al: Naltrexone and coping skills
therapy for alcohol dependence: a controlled study. Arch Gen
Psychiatry 49(11):881–887, 1992 1444726
Onder G, Pedone C, Landi F, et al: Adverse drug reactions as cause of
hospital admissions: results from the Italian Group of
Pharmacoepidemiology in the Elderly (GIFA). J Am Geriatr Soc
50(12):1962–1968, 2002 12473007
Oslin DW: Alcohol use in late life: disability and comorbidity. J Geriatr
Psychiatry Neurol 13(3):134–140, 2000 11001136
Oslin DW, Cary MS: Alcohol-related dementia: validation of diagnostic
criteria. Am J Geriatr Psychiatry 11(4):441–447, 2003 12837673
Oslin D, Liberto JG, O’Brien J, et al: Naltrexone as an adjunctive treatment
for older patients with alcohol dependence. Am J Geriatr Psychiatry
5(4):324–332, 1997a 9363289
Oslin DW, Streim JE, Parmelee P, et al: Alcohol abuse: a source of
reversible functional disability among residents of a VA nursing home.
Int J Geriatr Psychiatry 12(8):825–832, 1997b 9283927
Oslin D, Atkinson RM, Smith DM, et al: Alcohol related dementia:
proposed clinical criteria. Int J Geriatr Psychiatry 13(4):203–212,
1998a 9646147
Oslin DW, Pettinati HM, Luck G, et al: Clinical correlations with
carbohydrate-deficient transferrin levels in women with alcoholism.
Alcohol Clin Exp Res 22(9):1981–1985, 1998b 9884141
Oslin DW, Katz IR, Edell WS, et al: Effects of alcohol consumption on the
treatment of depression among elderly patients. Am J Geriatr
Psychiatry 8(3):215–220, 2000 10910419
Oslin DW, Pettinati H, Volpicelli JR: Alcoholism treatment adherence:
older age predicts better adherence and drinking outcomes. Am J
Geriatr Psychiatry 10(6):740–747, 2002 12427583
Oslin DW, Slaymaker VJ, Blow FC, et al: Treatment outcomes for alcohol
dependence among middle-aged and older adults. Addict Behav
30(7):1431–1436, 2005 16022937
Outlaw FH, Marquart JM, Roy A, et al: Treatment outcomes for older
adults who abuse substances. J Appl Gerontol 31:78–100, 2012
Peele S, Brodsky A: Exploring psychological benefits associated with
moderate alcohol use: a necessary corrective to assessments of drinking
outcomes? Drug Alcohol Depend 60(3):221–247, 2000 11053757
Plunk AD, Syed-Mohammed H, Cavazos-Rehg P, et al: Alcohol
consumption, heavy drinking, and mortality: rethinking the J-shaped
curve. Alcohol Clin Exp Res 38(2):471–478, 2014 24033586
Rehm J, Mathers C, Popova S, et al: Global burden of disease and injury
and economic cost attributable to alcohol use and alcohol-use
disorders. Lancet 373(9682): 2223–2233, 2009 19560604
Rehm J, Room R, Graham K, et al: The relationship of average volume of
alcohol consumption and patterns of drinking to burden of disease: an
overview. Addiction 98(9):1209–1228, 2003 12930209
Ridley NJ, Draper B, Withall A: Alcohol-related dementia: an update of the
evidence. Alzheimers Res Ther 5(1):3, 2013 23347747
Rösner S, Leucht S, Lehert P, et al: Acamprosate supports abstinence,
naltrexone prevents excessive drinking: evidence from a meta-analysis
with unreported outcomes. J Psychopharmacol 22(1):11–23, 2008
18187529
Rubinsky AD, Dawson DA, Williams EC, et al: AUDIT-C scores as a
scaled marker of mean daily drinking, alcohol use disorder severity,
and probability of alcohol dependence in a U.S. general population
sample of drinkers. Alcohol Clin Exp Res 37(8):1380–1390, 2013
Sacco P, Bucholz KK, Spitznagel EL: Alcohol use among older adults in
the National Epidemiologic Survey on Alcohol and Related
Conditions: a latent class analysis. J Stud Alcohol Drugs 70(6):829–
838, 2009 19895759
Sass H, Soyka M, Mann K, et al: Relapse prevention by acamprosate:
results from a placebo-controlled study on alcohol dependence. Arch
Gen Psychiatry 53(8): 673–680, 1996 8694680
Satre DD, Mertens J, Areán PA, et al: Contrasting outcomes of older versus
middle-aged and younger adult chemical dependency patients in a
managed care program. J Stud Alcohol 64(4):520–530, 2003 12921194
Satre DD, Mertens JR, Areán PA, et al: Five-year alcohol and drug
treatment outcomes of older adults versus middle-aged and younger
adults in a managed care program. Addiction 99(10):1286–1297, 2004a
15369567
Satre DD, Mertens JR, Weisner C: Gender differences in treatment
outcomes for alcohol dependence among older adults. J Stud Alcohol
65(5):638–642, 2004b 15536774
Schonfeld L, Dupree LW, Dickson-Euhrmann E, et al: Cognitive-behavioral
treatment of older veterans with substance abuse problems. J Geriatr
Psychiatry Neurol 13(3):124–129, 2000 11001134
Satre DD, Blow FC, Chi FW, et al: Gender differences in seven-year
alcohol and drug treatment outcomes among older adults. Am J Addict
16(3):216–221, 2007 17612826
Simoni-Wastila L, Yang HK: Psychoactive drug abuse in older adults. Am J
Geriatr Pharmacother 4(4):380–394, 2006 17296542
Sorock GS, Chen LH, Gonzalgo SR, et al: Alcohol-drinking history and
fatal injury in older adults. Alcohol 40(3):193–199, 2006 17418699
Stoehr GP, Ganguli M, Seaberg EC, et al: Over-the-counter medication use
in an older rural community: the MoVIES Project. J Am Geriatr Soc
45(2):158–165, 1997 9033513
Substance Abuse and Mental Health Services Administration: Results from
the 2012 National Survey on Drug Use and Health: Summary of
National Findings, NSDUH Series H-46 (HHS Publ No SMA-13-
4795). Rockville, MD, Substance Abuse and Mental Health Services
Administration, 2013
Tannenbaum C, Paquette A, Hilmer S, et al: A systematic review of
amnestic and nonamnestic mild cognitive impairment induced by
anticholinergic, antihistamine, GABAergic and opioid drugs. Drugs
Aging 29(8):639–658, 2012 22812538
Topic A, Djukic M: Diagnostic characteristics and application of alcohol
biomarkers. Clin Lab 59(3-4):233–245, 2013 23724610
Tucker JA, Foushee HR, Black BC, et al: Agreement between prospective
interactive voice response self-monitoring and structured retrospective
reports of drinking and contextual variables during natural resolution
attempts. J Stud Alcohol Drugs 68(4):538–542, 2007 17568958
Volpicelli JR, Alterman AI, Hayashida M, et al: Naltrexone in the treatment
of alcohol dependence. Arch Gen Psychiatry 49(11):876–880, 1992
1345133
Waern M: Alcohol dependence and misuse in elderly suicides. Alcohol
Alcohol 38(3): 249–254, 2003 12711660
Wagman AM, Allen RP, Upright D: Effects of alcohol consumption upon
parameters of ultradian sleep rhythms in alcoholics. Adv Exp Med Biol
85A:601–616, 1977 920494
Weathermon R, Crabb DW: Alcohol and medication interactions. Alcohol
Res Health 23(1):40–54, 1999 10890797
Werch CE: Quantity-frequency and diary measures of alcohol consumption
for elderly drinkers. Int J Addict 24(9):859–865, 1989 2621007
Widlitz M, Marin DB: Substance abuse in older adults: an overview.
Geriatrics 57(12):29–34, quiz 37, 2002 12494731
Wilsnack SC, Wilsnack RW: Drinking and problem drinking in U.S.
women: patterns and recent trends. Recent Dev Alcohol 12:29–60,
1995 7624549
Wu L, Blazer DG: Substance use disorders and psychiatric comorbidity in
mid and later life: a review. Int J Epidemiol 43(2):304–317, 2014
Suggested Readings
Blow FC (Consensus Panel Chair): Substance Abuse Among Older Adults.
Treatment Improvement Series Protocol (TIP) Series No. 26. Center for
Substance Abuse Treatment. Rockville, MD, U.S. Department of
Health and Human Services, 1998
Han B, Gfroerer JC, Colliver JD, et al: Substance use disorder among older
adults in the United States in 2020. Addiction 104(1):88–96, 2009
19133892
Kirchner JE, Zubritsky C, Cody M, et al: Alcohol consumption among
older adults in primary care. J Gen Intern Med 22(1):92–97, 2007
17351846
Oslin D, Liberto JG, O’Brien J, et al: Naltrexone as an adjunctive treatment
for older patients with alcohol dependence. Am J Geriatr Psychiatry
5(4):324–332, 1997 9363289
Plunk AD, Syed-Mohammed H, Cavazos-Rehg P, et al: Alcohol
consumption, heavy drinking, and mortality: rethinking the J-shaped
curve. Alcohol Clin Exp Res 38(2):471–478, 2014 24033586
Satre DD, Mertens JR, Areán PA, et al: Five-year alcohol and drug
treatment outcomes of older adults versus middle-aged and younger
adults in a managed care program. Addiction 99(10):1286–1297, 2004
15369567
Schonfeld L, Dupree LW, Dickson-Euhrmann E, et al: Cognitive-behavioral
treatment of older veterans with substance abuse problems. J Geriatr
Psychiatry Neurol 13(3):124–129, 2000 11001134
Simoni-Wastila L, Yang HK: Psychoactive drug abuse in older adults. Am J
Geriatr Pharmacother 4(4):380–394, 2006 17296542
Substance Abuse and Mental Health Services Administration: Results from
the 2012 National Survey on Drug Use and Health: Summary of
National Findings, NSDUH Series H-46 (HHS Publ No SMA-13-
4795). Rockville, MD, Substance Abuse and Mental Health Services
Administration, 2013
Wu L, Blazer DG: Substance use disorders and psychiatric comorbidity in
mid and later life: a review. Int J Epidemiol 43(2):304–317, 2014
CHAPTER 18
Personality Disorders
Thomas E. Oxman, M.D.
Definitions
Personality refers to an individual’s “enduring patterns of perceiving,
relating to, and thinking about the environment and oneself” (American
Psychiatric Association 2013, p. 826). Personality consists of both
temperament (i.e., genetic dispositions) and character (i.e., qualities
developed through interaction with the environment) (Robinson 2005). For
a patient to be diagnosed with a personality disorder, DSM-5 requires that
the individual have an enduring pattern of experience and behavior that is
noticeably different from cultural expectations, as manifested in two or
more of the following domains: cognition, affectivity, interpersonal
functioning, and impulse control. The pattern must have developed by early
adulthood and must be intractable and relatively stable across the lifespan.
A personality disorder influences a wide range of both social and personal
situations. As is the case with all DSM-5 disorders, a personality disorder
results in significant distress or functional impairment. Finally, the enduring
pattern must not be a manifestation of another mental disorder, a medical
disorder, or use of a substance.
In treating elderly persons, ruling out manifestations of other disorders is
particularly important because of the high prevalence of chronic medical
disorders. When a change in personality occurs because of the comorbidity
of another mental disorder, a medical disorder, or use of a substance, the
DSM-5 diagnosis of personality change due to another medical condition is
the appropriate diagnosis. However, dementias are also increasingly
prevalent with age and are often associated with changes in personality. A
change in personality is often the chief complaint when an older individual
is brought by caregivers for a specialist evaluation (Duchek et al. 2007).
These changes may be totally unrelated to premorbid personality, as occurs
in frontal lobe dementias (Rabinovici and Miller 2010), or may be marked
exaggerations of preexisting traits, as occurs in Alzheimer’s disease (Archer
et al. 2007). Because of the progressive pervasiveness of neurocognitive
disorders, the DSM-5 diagnosis of personality change due to another
medical condition does not apply. Although these issues are important in
differential diagnosis, for the purposes of this chapter the discussion of
personality disorder is focused more closely on conditions that meet DSM-5
personality disorder criteria.
After making a global assessment of the presence or absence of a general
personality disorder based on the key features listed previously, the next
step is to identify a specific personality disorder or a personality cluster.
Since 1980, DSM has categorized three different personality disorder
clusters (Table 18–1): Cluster A—odd, eccentric (including paranoid,
schizoid, and schizotypal personality disorders); Cluster B—dramatic,
emotional, erratic (including antisocial, borderline, histrionic, and
narcissistic personality disorders); and Cluster C—anxious, fearful
(including avoidant, dependent, and obsessive-compulsive personality
disorders). The number of specific disorders has varied slightly over the
past 20 years. Likewise, over the length of a lifespan often reaching 75–85
years, both personality and a personality disorder are likely to change
somewhat; therefore, attention to identification of a cluster is particularly
helpful. Because older patients with personality disorders commonly show
symptoms of more than one personality disorder, diagnosis of a specific
disorder is likely to be more difficult than in younger adults; in these cases,
patients should be diagnosed with other specified personality disorder or
unspecified personality disorder.
A. Odd, eccentric
Paranoid Perception that people Trust, acceptance Forced intimate contact
are dangerous; from physical
vigilance, dependence highlights
suspiciousness the disorder.
Schizoid Isolation, autonomy Reciprocity, intimacy The disorder is
relatively persistent.
Schizotypal Bizarre behavior Trust, social skills, Individuals without the
abstract thinking disorder who
experience theft and
ageism, leading to
appropriate
suspiciousness, may
be misdiagnosed.
B. Dramatic, erratic
Antisocial Exploitiveness, taking Empathy Elderly individuals are
advantage of others more law abiding, and
antisocial personality
is less common in
older prisoners.
Borderline Sensitivity to rejection, Reflection, Prevalence and/or
feelings of systematization severity of the
abandonment disorder declines.
Histrionic Expressiveness, Self-esteem beyond With older age there is
exhibitionism attractiveness, less energy and less
reflection opportunity for
promiscuity,
shoplifting, and
impulse expression.
Narcissistic Self-aggrandizement, Group identification, Increased losses, such as
perceived specialness, ability to share retirement and
competitiveness bereavement,
aggravate regulation
of self-esteem.
C. Anxious, fearful
Avoidant Vulnerability, inhibition Self-assertion, This disorder is
expressiveness associated with poor
outcome in major
depression.
Dependent Helplessness, attachment Mobility, self-reliance Individuals without the
disorder who have
fewer social
opportunities and
more medical illnesses
may be subject to
overdiagnosis.
Obsessive- Perfectionism, Playfulness, spontaneity The disorder is
responsibility, relatively persistent.
compulsive systematization
Adult Development
Erikson’s concept of epigenesis is important in understanding how
personality disorders might improve with age. Erikson was a strong
proponent of the interaction of the psychosocial environment with
development across the lifespan. Erikson’s stage theory of late-life
development (Erikson et al. 1986) proposed that the major developmental
task of older age is to look back and seek meaning across the lifespan,
rather than looking forward as in previous developmental modes that are
now in decline. The goal of this task, as discussed by Erikson, is to maintain
more integrity than despair about one’s life. In this process, as at previous
life stages, each earlier life stage conflict must be reconciled and integrated
with the current stage, allowing resolution of earlier conflicts. Individuals
with personality disorders might be expected to have greater difficulty in
accomplishing this resolution than other individuals. However, this
resolution is not an all-or-nothing phenomenon. The achievement of even
some resolution may contribute to the mellowing of a personality disorder.
As Vaillant (2012) has pointed out through his extended involvement with
longitudinal research, “If you follow lives long enough, they change as do
the factors that affect healthy adjustment” (p. 52).
Vaillant and others (e.g., Diehl et al. 1996) have provided empirical
verification for Erikson’s life-stage concepts through longitudinal study of
the maturation of defenses across the lifespan (Vaillant 2012). Defenses are
involuntary mental mechanisms for regulating the realities that persons are
powerless to change. Vaillant and others (Haan 1977) have described a
hierarchy of defenses from immature and maladaptive to mature and
adaptive. Mature defenses include humor, altruism, sublimation,
anticipation, and suppression. Mature and adaptive defenses synthesize and
attenuate conflicts rather than distorting or denying them. Across several
longitudinal studies that included privileged persons (Vaillant 2012), gifted
persons (Terman 1925), and persons from the core inner cities (Glueck and
Glueck 1968), Vaillant established that mature defenses were more
consistently identified primarily in Erikson’s later developmental stages
(Vaillant 1993) and that the development of these defenses was independent
of education and social privilege (Vaillant 1993, 2012). Similarly, others
have identified a socioemotional selectiveness for optimizing positive
emotional meaningfulness with age (Charles and Carstensen 2010) and
improved coping skills with aging (Ryff 1999).
Events with psychological and social impact can reveal previously
submerged difficulty. For example, the death of a spouse may unmask
dependency problems. Retirement or bereavement may lead to narcissistic
problems such as poor regulation of self-esteem and faulty adaptation to
loss. However, experience—something that the elderly have more of than
any other age group—may attenuate the impact of such later-life events,
even for older persons with personality disorders (Birditt and Fingerman
2005; Ryff 1999). For example, to cope effectively with stress, individuals
must learn to recognize the difference between situations that can and
cannot be changed and then must match the right coping skill with the right
situation. Emotion-focused coping is used when a situation cannot be
changed; problem-focused coping is used when a situation can be changed.
It is only through repeated experience that these skills develop (Ryff 1999).
Although stressful situations certainly exacerbate the symptoms of
personality disorders, Neugarten (1970) pointed out how the meaning of
stressful situations changes over time. People expect to experience the death
of loved ones and anticipate their own declining health as they age and have
time to mentally rehearse how they will respond to these “on-time” losses.
When these losses occur “off time”—at earlier stages of life—the stress is
usually experienced as much greater. The improvement in coping skills with
aging is also accompanied by socioemotional selectivity—that is, the
tendency of elderly persons to remember more pleasant events and positive
emotions in favor of unpleasant ones (Charles and Carstensen 2010;
Erikson et al. 1986; Nashiro et al. 2012; Vaillant 2012).
Consideration of this line of evidence should not minimize the impact of
personality disorders or the need for some management of them in later life.
Even if the prevalence of personality disorders truly does decline, there are
still some elderly persons with continuing disorders that cause impairment
to themselves and their environment. Consideration of adult development
helps us understand how some persons with personality disorders may
improve and to understand that this improvement is consistent with the
epidemiological findings of reduced psychopathology in later life. Equally
important, adult developmental theory helps the clinician consider the
positive aspects of development, even in individuals with severe
psychopathology (Erikson et al. 1986; Ryff 1999). Some people who
experienced a terrible childhood and young adulthood can still find
enjoyment at the end of life. Waiting 50 or more years for relative happiness
is a long time, so assisting in positive adaptation can certainly be
worthwhile (Vaillant 2012). Until the debate on the validity of the
epidemiological findings versus the criteria used to make those findings is
more definitively settled, perhaps designations of personality disorder “in
remission” or “in partial remission” would be a more appropriate and
parsimonious way of addressing diagnoses of personality disorder in the
elderly population.
Evaluation
Because personality disorders typically have a lifelong pattern, diagnosis
generally requires greater historical and collateral information than for
many other disorders. Complete understanding of the causes of signs and
symptoms in geriatric psychiatry is a difficult accomplishment. The
interplay of multiple etiological factors is the rule rather than the exception.
The Structured Clinical Interview for DSM-IV Personality Disorders
(SCID-II; First et al. 1997) and the Personality Disorders Examination
(PDE; Loranger 1988) are semistructured interviews for personality
disorders that can be used to guide a diagnostic interview and increase
reliability. Historical information, from medical records and from persons
who have known a patient over a long period, is still an essential
component of an accurate and valid diagnosis. The interview requires a
longitudinal inquiry about various life stages to establish the historical
presence of a personality disorder, even if not all current criteria are met.
Several ancillary self-report instruments are available for initial screening
purposes. However, the results of these self-reports have a low concordance
with the results of interview methods (Perry 1992), and their use is best
established and tolerated in younger populations, not among elderly
persons, in whom acquired brain disease is an increasing issue.
General personality inventories, as opposed to personality disorder
instruments, are more valid and reliable dimensional assessment tools that
have also been used with elderly patients. Originally derived through
empirical lexical research into natural language trait descriptions, a
significant body of research has repeatedly identified five broad domains of
personality (Digman 1990). The NEO Personality Inventory—Revised
(NEO PI-R; Costa and McCrae 1992), which uses a neuroticism (e.g.,
worrying vs. calm), extraversion (e.g., joiner vs. loner), openness (e.g.,
preference for variety vs. routine), agreeableness (e.g., trusting vs.
suspicious), and conscientiousness (e.g., hardworking vs. lazy) five-factor
model with 30 facets, is the most commonly used and researched
personality inventory (Widiger and Mullins-Sweatt 2009). The NEO PI-R
has also proved useful as a screening tool for personality disorders (Miller
et al. 2005). The NEO PI-R has been validated in a geriatric population and
appears valid for screening of paranoid, borderline, histrionic, avoidant, and
dependent personality disorders (Van den Broeck et al. 2013). However, the
NEO PI-R has more than 200 items, and copyright fee issues make it
unwieldy for routine clinical use. Although DSM-5 did not make the
recommended paradigm shift to a dimensional model of personality
disorders (Oldham and Skodol 2013; Widiger and Mullins-Sweatt 2009),
Section III of DSM-5 provided an “Alternative DSM-5 Model for
Personality Disorders,” including a five-factor personality trait model with
personality disorders understood as maladaptive variants of five domains
and 25 facets. The five broad trait domains in DSM-5 are negative
affectivity versus emotional stability, detachment versus extraversion,
antagonism versus agreeableness, disinhibition versus conscientiousness,
and psychoticism versus lucidity. The Personality Inventory for DSM-5
(PID-5; Krueger et al. 2012), an instrument derived from this model, is an
“emerging measure” available for free at the American Psychiatric
Association Web site (www.psychiatry.org/practice/dsm/dsm5/online-
assessment-measures). For most clinicians, patients, and families, the full-
length 220-item instrument is probably too cumbersome for routine use;
therefore, brief forms are also available. Notably for geriatric psychiatry,
the PID-5 does not sufficiently focus on the positive dimensions of adult
development, which should be accorded equal importance with negative
dimensions when personality is being evaluated.
Geriatric psychiatrists are familiar with the phenomenon that acquired
brain disease in later life appears to strengthen undesirable personality traits
that were present, although less intense and conspicuous, in earlier adult
life. However, if signs and symptoms of personality disorder were not
present before the onset of a neurocognitive illness or brain injury, it is
rational to assume that such illnesses play a causative role in the personality
change. A reasonable approach to a diagnosis in geriatrics, as always,
includes these elements: 1) careful and detailed review of the medical and
psychosocial history; 2) mental status and physical examinations, with
special attention to the neurological examination; and 3) a screening
laboratory examination—including, in some cases, brain imaging, an
electroencephalogram, and neuropsychological tests.
In many instances, family members are better able than older patients to
provide the historical information that provides the best clues about the
patient. Whenever possible, their help should be sought concerning the
older patient in whom personality disorder is suspected. Viewing the patient
within the family context usually gives added depth of understanding to the
clinical perspective.
Syndromes based on frontal lobe pathology that result in loss of normal
executive function present some of the most difficult diagnostic challenges,
especially if the onset of symptoms is subtle, the rate of progression is slow,
and the main attributes of the premorbid personality are obscure.
Individuals with frontal or frontotemporal lobe disease may show good
preservation of memory function. They are, however, prone to trouble with
“mechanistic planning, verbal reasoning, or problem solving” and “obeying
the rules of interpersonal social behaviour, the experience of reward and
punishment, and the interpretation of complex emotions” (Grafman and
Litvan 1999, p. 1921; Passant et al. 2005).
These difficulties are similar to some of the problems experienced by
many people with borderline, narcissistic, histrionic, paranoid, and
antisocial personality disorders. A question of interest is whether a person
can develop a later-life personality disorder de novo, without the presence
of underlying brain disease or substance abuse. The answer is not
definitively known, but the occurrence is probably rare and such a diagnosis
is not allowed in DSM-5, which requires onset of personality disorder by
early adulthood. Also of relevance, signs and symptoms of personality
disorders can be quite evident in early adult life, then be diminished or
quiescent in mid-adult life, and then reemerge under the stress of social
losses or physical illness in later life (Rosowsky and Gurian 1992).
Additionally, the manifestations of personality disorders may vary in
different parts of the life cycle. For example, in persons with borderline
personality disorder, phenomena such as splitting, intense and unstable
interpersonal relationships, impaired affective regulation, and extreme
difficulty with control and regulation of anger often persist throughout the
life cycle. Problems such as severe impulsivity, risky behavior, and self-
mutilation tend to diminish with advancing age (Zanarini et al. 2007).
However, other self-injurious behaviors may take their place. These include
self-starvation, abuse of medications, and noncompliance with medical
treatment (Rosowsky and Gurian 1992). These behaviors may occur for
other reasons, but their presence should at least alert the clinician to the
possibility of borderline personality disorder. Notably, late-onset obsessive-
compulsive symptoms or traits are particularly likely to have a basis in
brain disease.
Treatment Issues
The broad focus of personality disorder treatment is on reducing symptoms,
improving social functioning, and changing responses to the environment
(Robinson 2005). Off-label use of psychotropic agents is sometimes
valuable for symptom reduction, whereas psychotherapy and caregiver
education are helpful for addressing environmental response. All of these
possibilities should be considered for their contribution to improved
function of older persons.
Psychotherapy
In contrast to the unconscious maturation of defenses, the conscious
alteration of personality is unusual. Although individuals are sometimes
able to change patterns of behavior, attitudes, ways of thinking, and ways of
feeling, changing the fundamental personality structure is extremely
difficult. At the same time, it is possible and important to try to help
patients avoid behavior that significantly harms themselves or others.
Validating feelings and empathizing with distress, without necessarily
agreeing to the proportionality of the response, is a first step in the
therapeutic relationship (Hunt 2007). Helping patients recognize and alter
erroneous or distorted thinking is thus also important and possible.
Cognitive-behavioral therapy, and its variant Dialectical Behavior Therapy
(Lynch et al. 2007), or insight-oriented psychotherapy may significantly
help older individuals who are functioning at higher levels and who are not
otherwise seriously ill or incapacitated (Beck et al. 2004; Clarkin et al.
2007; Hunt 2007). For patients in psychiatric hospitals and for residents of
group homes or nursing homes, intensive psychotherapy with a goal of
changing lifelong maladaptive personality features is neither an available
nor an indicated treatment modality. However, for such individuals,
supportive and consistent psychotherapeutic contact can be of great benefit
(Hunt 2007).
As noted, psychotherapy of any type, either by itself or in combination
with pharmacotherapy, with a goal of a global revision of maladaptive
aspects of personality in later life, is unlikely to succeed. Individualized
treatment targeting specific symptoms that discomfort, threaten, or
endanger patients or their family or caregivers—for example, behavioral
management to minimize harm from impaired social judgment—is far more
realistic and more likely to realize success. However, it is still important not
to be rigid or negative about the psychotherapeutic potential of older
individuals. It is illness, not merely age, that limits or impairs the plasticity
of the personality and the potential for self-change. Older patients are often
impressive in their resilience, courage, open-mindedness, and willingness to
try new ways of thinking and behaving—and those showing these
characteristics include some individuals whose adaptation to life in earlier
years was far from optimal (Vaillant 2012). This observation is in keeping
with Erikson’s view of progressive development throughout life.
Psychoanalytically oriented psychotherapy, cognitive-behavioral therapy,
interpersonal therapy, dialectical behavior therapy, and other forms of
psychotherapy all have their adherents and proponents (De Leo et al. 1999).
All are probably helpful to certain individuals. The principal features of
successful psychotherapy for geriatric patients are a structure with
consistency, availability, empathic and respectful listening, flexibility, and
open-mindedness on the part of the psychotherapist. These features are
probably more important than a particular theoretical orientation (Clarkin et
al. 2007).
Therapists who are not yet old themselves have a difficult challenge.
They have no direct experience with or memory of being old (Rosowsky
1999). Some geriatric psychiatrists may be two full generations younger
than their patients. Extraordinary empathy is required of these clinicians,
but also of most practicing geriatric psychiatrists, who are usually caring for
persons older than themselves.
Pharmacotherapy
A diagnosis of personality disorder should not preclude pharmacological
treatment of concomitant psychiatric disorders, such as affective illness or
psychosis, as well as specific symptoms that may respond to psychotropic
medications. Successful treatment of affective or psychotic symptoms may
show that the symptoms were the result of these eminently treatable
diseases rather than entrenched maladaptive personality traits. Personality
disorder, depressive illness, and acquired brain disease share overlapping
symptom constellations: symptoms such as irritability, hostility, and
uncooperativeness can derive from all three. Even the most perspicacious
diagnostician may not be able to tease out a clear etiological diagnosis for
these difficult behavioral symptoms. In such cases, an empirical treatment
trial with antidepressant medicine is indicated. Preliminary evidence
suggests that selective serotonin reuptake inhibitors (SSRIs) (Knutson et al.
1998; Tang et al. 2009) and second-generation antipsychotics (Black et al.
2014) may alter aspects of personality separate from specific symptoms.
Individuals with other symptoms—such as anger outbursts, apathy, and
impaired social judgment—may also benefit from pharmacotherapy
(Hollander et al. 2003). Pharmacological treatment should be a systematic
trial guided by three principles. First, a medication should be selected for an
identified target symptom area (e.g., affect, impulsivity, aggression,
anxiety) (Table 18–2). Second, such trials should include a repeatable
assessment strategy (e.g., global rating, self-report, or caregiver report
targeted to the symptom area). Third, trials should have a specified duration
at the end of which a decision is made whether or not to continue the
medication. SSRIs and other newer antidepressant drugs, anticonvulsants,
and atypical antipsychotic drugs, used alone or in combinations, may be
useful in systematic trials for specified symptoms. The evidence is limited
or mixed for geriatric patients, and caution is indicated, especially because
of the side effects of antipsychotics (Maglione et al. 2011).
Caregiver Education
Education of caregivers is an important function of geriatric psychiatrists.
Family members may be having great difficulty in dealing with unfamiliar
negative, disinhibited, or inappropriate behavior. If an underlying medical
or neurological etiology can be discerned, caregivers can be reassured about
the cause of the otherwise inexplicable changes in their relationship with
their loved one. This reassurance helps reduce guilt, anxiety, and
uncertainty in the family of the afflicted person.
Anger Benzodiazepines
Anxiety
Behavior
Impulsivity Mood stabilizers
Reward/dependence
Note. MAOI=monoamine oxidase inhibitor; SSRI=selective serotonin reuptake inhibitor; TCA =
tricyclic antidepressant.
Primary caregivers in nursing homes, who are usually overworked and
underpaid, may not realize that much of the unpleasant behavior of patients
that they encounter in their work is not under full volitional control.
Uncooperativeness or angry outbursts may have the appearance of simple
willfulness or intentionally oppositional behavior. Patients with long-
standing personality disorders (as well as those with dementia, stroke, or
other types of brain injury) typically have significant deficits in volitional
capacity. Understanding this point does not necessarily make the care of
these patients easier, but it does provide a perspective on behavior that is
otherwise difficult to comprehend and tolerate. However, at times it may be
necessary for the psychiatrist to evaluate the competence of a patient with
personality disorder and consider proposing guardianship (Little and Little
2010).
Conclusion
Understanding of the causes of disordered personality development is far
from adequate. Diagnosis, and especially treatment, of personality disorders
in elderly patients is difficult. Understanding the differentiation between
lifelong development and acquired neuropsychiatric illness is an important
diagnostic challenge. Geriatric psychiatrists usually try to manage
personality disorder symptoms and ameliorate their harmful effects rather
than attempt to cure the underlying disorder. Behavioral management,
psychotherapy, pharmacotherapy, and caregiver support are the tools that
must be judiciously used.
Key Points
• The natural history of personality in later life is influenced by the
combination of increasing medical-neuropsychiatric comorbidity and
ongoing normal psychosocial maturation.
• Longitudinal history is necessary for determining whether presumed
personality problems in later life are a manifestation of a personality
disorder, a neurocognitive disorder, or both.
• The prevalence of personality disorders in older persons is generally
lower than that in younger persons in the general population.
• Positive adaptation in late-life development can mediate the severity and
presentation of personality disorders.
• Substantial evidence shows the negative effect of personality disorders
on the outcome of depressive disorders in elderly persons.
• It is possible and important to use psychotherapeutic and caregiver
interventions to try to help older patients with personality disorders
avoid behavior that significantly harms themselves or others.
• Systematic pharmacological treatment should include a target symptom
area, an assessment strategy for that symptom area, and a specified
duration of treatment.
References
Abrams RC, Alexopoulos GS, Spielman LA, et al: Personality disorder
symptoms predict declines in global functioning and quality of life in
elderly depressed patients. Am J Geriatr Psychiatry 9(1):67–71, 2001
11156754
Agronin ME, Maletta G: Personality disorders in late life: understanding
and overcoming the gap in research. Am J Geriatr Psychiatry 8(1):4–
18, 2000 10648290
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition. Washington, DC, American Psychiatric
Association, 1994
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 4th Edition, Text Revision. Washington, DC,
American Psychiatric Association, 2000
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Ames A, Molinari V: Prevalence of personality disorders in community-
living elderly. J Geriatr Psychiatry Neurol 7(3):189–194, 1994 7916944
Archer N, Brown RG, Reeves SJ, et al: Premorbid personality and
behavioral and psychological symptoms in probable Alzheimer disease.
Am J Geriatr Psychiatry 15(3):202–213, 2007 17322133
Balsis S, Woods CM, Gleason MEJ, et al: Overdiagnosis and
underdiagnosis of personality disorders in older adults. Am J Geriatr
Psychiatry 15(9):742–753, 2007 17804828
Beck AT, Freeman A, Davis DD: Cognitive Therapy of Personality
Disorders, 2nd Edition. New York, Guilford, 2004
Birditt KS, Fingerman KL: Do we get better at picking our battles? Age
group differences in descriptions of behavioral reactions to
interpersonal tensions. J Gerontol B Psychol Sci Soc Sci 60(3):121–
128, 2005 15860781
Black DW, Zanarini MC, Romine A, et al: Comparison of low and
moderate dosages of extended-release quetiapine in borderline
personality disorder: a randomized, double-blind, placebo-controlled
trial. Am J Psychiatry. 2014 Jun 27. [Epub ahead of print] 24968985
Charles ST, Carstensen LL: Social and emotional aging. Annu Rev Psychol
61:383–409, 2010 19575618
Clarkin JF, Levy KN, Lenzenweger MF, et al: Evaluating three treatments
for borderline personality disorder: a multiwave study. Am J Psychiatry
164(6):922–928, 2007 17541052
Costa PT, McCrae RR: Revised NEO Personality Inventory (NEO PI-R)
and NEO Five Factor Inventory (NEO-FFI) Professional Manual.
Odessa, FL, Psychological Assessment Resources, 1992
Dar-Nimrod I, Chapman BP, Franks P, et al: Personality factors moderate
the associations between apolipoprotein genotype and cognitive
function as well as late onset Alzheimer disease. Am J Geriatr
Psychiatry 20(12):1026–1035, 2012 23079898
De Leo D, Scocco P, Meneghel G: Pharmacological and psychotherapeutic
treatment of personality disorders in the elderly. Int Psychogeriatr
11(2):191–206, 1999 11475433
Diehl M, Coyle N, Labouvie-Vief G: Age and sex differences in strategies
of coping and defense across the life span. Psychol Aging 11(1):127–
139, 1996 8726378
Digman JM: Personality structure: emergence of the five-factor model.
Annu Rev Psychol 41:417–440, 1990
Duchek JM, Balota DA, Storandt M, et al: The power of personality in
discriminating between healthy aging and early-stage Alzheimer’s
disease. J Gerontol B Psychol Sci Soc Sci 62(6):353–361, 2007
18079420
Erikson EH, Erikson JM, Kivnick HQ: Vital Involvement in Old Age. New
York, WW Norton, 1986
First MB, Gibbon M, Spitzer RL, et al: Structured Clinical Interview for
DSM-IV Axis II Personality Disorders (SCID-II). Washington, DC,
American Psychiatric Press, 1997
Galione JN, Oltmanns TF: The relationship between borderline personality
disorder and major depression in later life: acute versus temperamental
symptoms. Am J Geriatr Psychiatry 21(8):747–756, 2013 23567384
Glueck S, Glueck E: Delinquents and Non-Delinquents in Perspective.
Cambridge, MA, Harvard University Press, 1968
Grafman J, Litvan I: Importance of deficits in executive functions. Lancet
354(9194): 1921–1923, 1999 10622291
Grant BF, Hasin DS, Stinson FS, et al: Prevalence, correlates, and disability
of personality disorders in the United States: results from the national
epidemiologic survey on alcohol and related conditions. J Clin
Psychiatry 65(7):948–958, 2004 15291684
Haan NA: Coping and Defending. San Francisco, CA, Jossey-Bass, 1977
Hanlon TE, Nurco DN, Kinlock TW, Duszynski KR: Trends in criminal
activity and drug use over an addiction career. Am J Drug Alcohol
Abuse 16(3–4):223–238, 1990 2288322
Harlow CW: Special Report: Profile of Jail Inmates 1996 (Publ No NCJ
164620). Washington, DC, U.S. Department of Justice, 1998
Hayward RD, Taylor WD, Smoski MJ, et al: Association of five-factor
model personality domains and facets with presence, onset, and
treatment outcomes of major depression in older adults. Am J Geriatr
Psychiatry 21(1):88–96, 2013 23290206
Hillman J, Stricker G, Zweig R: Clinical psychologists’ judgment of older
adult patients with character pathology: implications for practice. Prof
Psychol Res Pr 28:179–183, 1997
Hollander E, Tracy KA, Swann AC, et al: Divalproex in the treatment of
impulsive aggression: efficacy in cluster B personality disorders.
Neuropsychopharmacology 28(6):1186–1197, 2003 12700713
Hunt M: Borderline personality disorder across the lifespan. J Women
Aging 19(1–2):173–191, 2007 17588886
Iwasa H, Masui Y, Gondo Y, et al: Personality and all-cause mortality
among older adults dwelling in a Japanese community: a five-year
population-based prospective cohort study. Am J Geriatr Psychiatry
16(5):399–405, 2008 18403571
Knutson B, Wolkowitz OM, Cole SW, et al: Selective alteration of
personality and social behavior by serotonergic intervention. Am J
Psychiatry 155(3):373–379, 1998 9501748
Krueger RF, Derringer J, Markon KE, et al: Initial construction of a
maladaptive personality trait model and inventory for DSM-5. Psychol
Med 42(9):1879–1890, 2012 22153017
Kuzma E, Sattler C, Toro P, et al: Premorbid personality traits and their
course in mild cognitive impairment: results from a prospective
population-based study in Germany. Dement Geriatr Cogn Disord
32(3):171–177, 2011 22005607
Lenzenweger MF, Lane MC, Loranger AW, et al: DSM-IV personality
disorders in the National Comorbidity Survey Replication. Biol
Psychiatry 62(6):553–564, 2007 17217923
Little J, Little B: Borderline personality disorder: exceptions to the concept
of responsible and competent. Australas Psychiatry 18(5):445–450,
2010 20863185
Loranger AW: Personality Disorders Examination (PDE) Manual. Yonkers,
NY, DV Communications, 1988
Low LF, Harrison F, Lackersteen SM: Does personality affect risk for
dementia? A systematic review and meta-analysis. Am J Geriatr
Psychiatry 21(8):713–728, 2013 23567438
Lynch TR, Cheavens JS, Cukrowicz KC, et al: Treatment of older adults
with co-morbid personality disorder and depression: a dialectical
behavior therapy approach. Int J Geriatr Psychiatry 22(2):131–143,
2007 17096462
Maglione M, Maher AR, Hu J, et al: Off-Label Use of Atypical
Antipsychotics: An Update. AHRQ Comparative Effectiveness
Reviews, Agency for Healthcare Research and Quality. 2011. Available
at: http://www.ncbi.nlm.nih.gov/books/NBK66081/. Accessed June 7,
2014.
Marengoni A, Angleman S, Melis R, et al: Aging with multimorbidity: a
systematic review of the literature. Ageing Res Rev 10(4):430–439,
2011 21402176
Miller JD, Bagby RM, Pilkonis PA, et al: A simplified technique for scoring
DSM-IV personality disorders with the Five-Factor Model. Assessment
12(4):404–415, 2005 16244121
Morse JQ, Lynch TR: A preliminary investigation of self-reported
personality disorders in late life: prevalence, predictors of depressive
severity, and clinical correlates. Aging Ment Health 8(4):307–315,
2004 15370047
Nashiro K, Sakaki M, Mather M: Age differences in brain activity during
emotion processing: reflections of age-related decline or increased
emotion regulation? Gerontology 58(2):156–163, 2012 21691052
Neugarten BL: Dynamics of transition of middle age to old age: adaptation
and the life cycle. J Geriatr Psychiatry 4(1):71–100, 1970 5527462
Oldham JM, Skodol AE: Personality and personality disorders, and the
passage of time. Am J Geriatr Psychiatry 21(8):709–712, 2013
23711737
Passant U, Elfgren C, Englund E, et al: Psychiatric symptoms and their
psychosocial consequences in frontotemporal dementia. Alzheimer Dis
Assoc Disord 19(Suppl 1):S15–S18, 2005 16317252
Perry JC: Problems and considerations in the valid assessment of
personality disorders. Am J Psychiatry 149(12):1645–1653, 1992
1443240
Putkonen H, Weizmann-Henelius G, Repo-Tiihonen E, et al: Homicide,
psychopathy, and aging—a nationwide register-based case-comparison
study of homicide offenders aged 60 years or older. J Forensic Sci
55(6):1552–1556, 2010 20629908
Rabinovici GD, Miller BL: Frontotemporal lobar degeneration:
epidemiology, pathophysiology, diagnosis and management. CNS
Drugs 24(5):375–398, 2010 20369906
Roberts BW, Walton KE, Viechtbauer W: Patterns of mean-level change in
personality traits across the life course: a meta-analysis of longitudinal
studies. Psychol Bull 132(1):1–25, 2006 16435954
Roberts BW, Smith J, Jackson JJ, et al: Compensatory conscientiousness
and health in older couples. Psychol Sci 20(5):553–559, 2009
19476589
Robinson DJ: Field Guide to Personality Disorders, 2nd Edition. Port
Huron, MI, Rapid Psychler Press, 2005
Rosowsky E: Personality disorders and the difficult nursing home resident,
in Personality Disorders in Older Adults: Emerging Issues in Diagnosis
and Treatment. Edited by Rosowsky E, Abrams RC. Mahwah, NJ,
Erlbaum, 1999, pp 257–274
Rosowsky E, Gurian B: Impact of borderline personality disorder in late life
on systems of care. Hosp Community Psychiatry 43(4):386–389, 1992
1577432
Ryff CD: Psychology and aging, in Principles of Geriatric Medicine and
Gerontology, 4th Edition. Edited by Hazzard WR, Blass JP, Ettinger
WH, et al. New York, McGraw-Hill, 1999, pp 159–169
Schuster JP, Hoertel N, Le Strat Y, et al: Personality disorders in older
adults: findings from the National Epidemiologic Survey on Alcohol
and Related Conditions. Am J Geriatr Psychiatry 21(8):757–768, 2013
23567365
Tang TZ, DeRubeis RJ, Hollon SD, et al: Personality change during
depression treatment: a placebo-controlled trial. Arch Gen Psychiatry
66(12):1322–1330, 2009 19996037
Terman LM: Genetic Studies of Genius, Vol 1: Mental and Physical Traits
of a Thousand Gifted Children. Palo Alto, CA, Stanford University
Press, 1925
Tracie Shea M, Edelen MO, Pinto A, et al: Improvement in borderline
personality disorder in relationship to age. Acta Psychiatr Scand
119(2):143–148, 2009 18851719
Tyrer P, Seiverwright H: Studies of outcome, in Personality Disorders:
Diagnosis, Management and Course. Edited by Tyrer P. London,
Wright, 1988, pp 119–136
Vaillant GE: The Wisdom of the Ego. Cambridge, MA, Harvard University
Press, 1993
Vaillant GE: Ego mechanisms of defense and personality psychopathology.
J Abnorm Psychol 103(1):44–50, 1994 8040479
Vaillant GE: Triumphs of Experience: The Men of the Harvard Grant Study.
Cambridge, MA, London, England, 2012
Van den Broeck J, Rossi G, De Clercq B, et al: Validation of the FFM PD
count technique for screening personality pathology in later middle-
aged and older adults. Aging Ment Health 17(2):180–188, 2013
22913535
van Ockenburg SL, de Jonge P, van der Harst P, et al: Does neuroticism
make you old? Prospective associations between neuroticism and
leukocyte telomere length. Psychol Med 44(4):723–729, 2014
Widiger TA, Mullins-Sweatt SN: Five-factor model of personality disorder:
a proposal for DSM-V. Annu Rev Clin Psychol 5:197–220, 2009
19046124
Zanarini MC, Frankenburg FR, Reich DB, et al: The subsyndromal
phenomenology of borderline personality disorder: a 10-year follow-up
study. Am J Psychiatry 164(6):929–935, 2007 17541053
Zanarini MC, Frankenburg FR, Fitzmaurice G: Defense mechanisms
reported by patients with borderline personality disorder and axis II
comparison subjects over 16 years of prospective follow-up:
description and prediction of recovery. Am J Psychiatry 170(1):111–
120, 2013 23223866
Suggested Readings
Erikson EH, Erikson JM, Kivnick HQ: Vital Involvement in Old Age. New
York, WW Norton, 1986
Oldham JM, Skodol AE: Personality and personality disorders, and the
passage of time. Am J Geriatr Psychiatry 21:709–712, 2013
Oltmanns TF, Balsis S: Personality disorders in later life: questions about
the measurement, course, and impact of disorders. Annu Rev Clin
Psychol 7:321–349, 2011
Terracciano A, Iacono D, O’Brien RJ, et al: Personality and resilience to
Alzheimer’s disease neuropathology: a prospective autopsy study.
Neurobiol Aging 34:1045–1050, 2013
Vaillant GE: Triumphs of Experience: The Men of the Harvard Grant Study.
Cambridge, MA, Harvard University Press, 2012
van Ockenburg SL, de Jonge P, van der Harst P, et al: Does neuroticism
make you old? Prospective associations between neuroticism and
leukocyte telomere length. Psychol Med 44(4):723–729, 2014
CHAPTER 19
We would like to thank W. Goforth, M.D., who was a co-author on the previous edition of this
chapter.
Nonpharmacological Approaches
Current person-centered dementia care assumes that most resistive
behaviors in dementia are responses to unmet needs or to environmental or
interpersonal triggers. Nonpharmacological strategies are recommended as
first-line approaches for agitation in dementia. These approaches can be
taught effectively to family and nonprofessional caregivers (Belle et al.
2006; Cohen-Mansfield et al. 2007; Doody et al. 2001; Hepburn et al. 2007;
Logsdon et al. 2007; Teri et al. 2005). These strategies focus on changing
the patient’s activities, routines, and/or human, physical, and social
environments to provide reassurance, appropriate stimulation and cues, and
security. As the individual with dementia becomes less adaptable to change,
the human and physical environment must adapt to him or her. Behavioral
approaches generally include person-specific problem solving, enriched
cues, adapted work or expressive activities, exercise, communication
strategies, and caregiver skills training.
Please don’t correct me. Remember, my feelings are intact and I get hurt easily. Try to ignore
offhand remarks that I wouldn’t have made in the past. If you focus on my mistakes, it just
makes me feel worse. I may say something that is real to me but not factual to you. It is not a
lie. Don’t argue—it won’t solve anything. (Snyder 2001, p. 2)
How dare you question me? I have always taken care of myself.
Pharmacological/Medical Approaches
There are times when agitation warrants pharmacological intervention, and
the risks of persistent agitation versus the risks and benefits of treating the
agitation must be weighed carefully. Most clinicians view agitation as a
condition manifested by excessive verbal and/or motor behavior. It is
distinguished from aggression, which can also be verbal (e.g., cursing,
threats) or physical (e.g., hitting, kicking, shoving objects or people, biting,
scratching). Agitation can escalate to aggression, so it is vital for the
clinician to intervene early in approaching agitated patients. However, it is
imperative to determine the cause of agitation so that interventions can then
be directed at both treating the underlying cause and managing the agitation
itself, given that uncontrolled agitation has been demonstrated to have
multiple deleterious effects on patient and family safety and welfare.
Agitation most commonly occurs in the context of delirium or dementia,
and often these conditions coexist in frail elderly patients. These conditions
are discussed in depth in the next two subsections of the chapter. Agitation
can also be a feature of late-life depression, and although treatment of the
underlying depression should also treat the agitation, the full effect of
antidepressant medications may not be apparent for several weeks.
However, acute, severe, or escalating agitation may require medication such
as the newer atypical neuroleptics for adequate control. Benzodiazepines
should generally be avoided in an agitated patient because of their high
potential for worsening delirium as well as potentially disinhibiting the
patient further; however, they may be appropriately used in cases of alcohol
or benzodiazepine withdrawal, or when the agitation can be assumed to be
the result of severe anxiety rather than delirium or the nonspecific agitation
that can accompany dementia.
Pharmacological Treatment
Currently, no treatments have been approved by the U.S. Food and Drug
Administration (FDA) for the management of agitation in dementia.
Moreover, nonpharmacological treatments are considered first line in the
management of these patients. Given the potential risks and limited efficacy
associated with most medications that have been studied for this indication,
pharmacological treatments are reserved for the following situations: 1) to
alleviate persistent distress, to maintain function, or to allow delivery of
needed care when nonpharmacological treatments have been tried and have
failed and 2) if the situation is serious enough to warrant urgent
pharmacological intervention (i.e., the patient is a danger to self or others).
Several medication classes have been studied for management of agitation
in dementia and are reviewed here.
Conclusion
Geriatric psychiatrists are frequently called upon to guide the management
of agitation in elderly persons in different settings. It is important to
understand and address underlying contributors to agitation, whether they
be medical, psychological, or environmental. Instituting measures to
prevent agitation is critical, as is educating caregivers about
nonpharmacological approaches for managing agitation. Unfortunately,
most pharmacological options for treating agitation are associated with
risks. More research is needed to address what nonpharmacological
interventions are best suited to which patients and in what situations. Safer
pharmacological options are also needed to manage agitation so as to
improve the quality of life of our older patients.
Key Points
• Agitation is a common and disabling condition in elderly people.
• The causes of agitation include a wide differential that must be evaluated
and corrected prior to consideration of treatment.
• Nonpharmacological approaches are the preferred treatment for agitation
in older adults.
• When the patient does not respond to nonpharmacological approaches or
if symptoms are severe, pharmacological approaches may be warranted.
• If pharmacological means are used, ongoing risk-benefit analyses should
guide treatment; the lowest possible dosages should be used for the
shortest possible times.
References
Alao AO, Soderberg M, Pohl EL, et al: Aripiprazole in the treatment of
delirium. Int J Psychiatry Med 35(4):429–433, 2005 16673842
Alzheimer’s Association: Aggression and anger. 2014a. Available at:
http://www.alz.org/care/alzheimers-dementia-aggression-anger.asp.
Accessed March 2014.
Alzheimer’s Association: Anxiety and agitation. 2014b. Available at:
https://www.alz.org/care/alzheimers-dementia-agitation-anxiety.asp.
Accessed March 2014.
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Ballard C, Lana MM, Theodoulou M, et al; Investigators DART AD: A
randomised, blinded, placebo-controlled trial in dementia patients
continuing or stopping neuroleptics (the DART-AD trial). PLoS Med
5(4):e76, 2008 18384230
Ballard C, Hanney ML, Theodoulou M, et al; DART-AD investigators: The
dementia antipsychotic withdrawal trial (DART-AD): long-term
follow-up of a randomised placebo-controlled trial. Lancet Neurol
8(2):151–157, 2009 19138567
Belle SH, Burgio L, Burns R, et al; Resources for Enhancing Alzheimer’s
Caregiver Health (REACH) II Investigators: Enhancing the quality of
life of dementia caregivers from different ethnic or racial groups: a
randomized, controlled trial. Ann Intern Med 145(10):727–738, 2006
17116917
Boettger S, Friedlander M, Breitbart W, et al: Aripiprazole and haloperidol
in the treatment of delirium. Aust N Z J Psychiatry 45(6):477–482,
2011 21563866
Chen JC, Borson S, Scanlan JM: Stage-specific prevalence of behavioral
symptoms in Alzheimer’s disease in a multi-ethnic community sample.
Am J Geriatr Psychiatry 8(2):123–133, 2000 10804073
Chow TW, MacLean CH: Quality indicators for dementia in vulnerable
community-dwelling and hospitalized elders. Ann Intern Med 135(8 Pt
2):668–676, 2001 11601949
Cohen-Mansfield J, Libin A, Marx MS: Nonpharmacological treatment of
agitation: a controlled trial of systematic individualized intervention. J
Gerontol A Biol Sci Med Sci 62(8):908–916, 2007 17702884
Devanand DP, Mintzer J, Schultz SK, et al: Relapse risk after
discontinuation of risperidone in Alzheimer’s disease. N Engl J Med
367(16):1497–1507, 2012 23075176
Doody RS, Stevens JC, Beck C, et al; Report of the Quality Standards
Subcommittee of the American Academy of Neurology: Practice
parameter: management of dementia (an evidence-based review).
Neurology 56(9):1154–1166, 2001 11342679
Flaherty JH, Gonzales JP, Dong B: Antipsychotics in the treatment of
delirium in older hospitalized adults: a systematic review. J Am Geriatr
Soc 59 (suppl 2): S269-S276, 2011 22091572
Fox C, Crugel M, Maidment I, et al: Efficacy of memantine for agitation in
Alzheimer’s dementia: a randomised double-blind placebo controlled
trial. PLoS One 7(5):e35185, 2012 22567095
Gallicchio L, Siddiqi N, Langenberg P, et al: Gender differences in burden
and depression among informal caregivers of demented elders in the
community. Int J Geriatr Psychiatry 17(2):154–163, 2002 11813279
Gitlin LN, Kales HC, Lyketsos CG: Nonpharmacologic management of
behavioral symptoms in dementia. JAMA 308(19): 2020–2029, 2012
23168825
Gwyther LP: Family issues in dementia: finding a new normal. Neurol Clin
18(4):993–1010, 2000 11072271
Gwyther L: Caring for People With Alzheimer’s Disease: A Manual for
Facility Staff. Washington, DC, American Health Care Association and
Alzheimer’s Association, 2001
Hall KA, Keks NA, O’Connor DW: Transdermal estrogen patches for
aggressive behavior in male patients with dementia: a randomized,
controlled trial. Int Psychogeriatr 17(2):165–178, 2005 16050428
Hepburn K, Lewis M, Tomatore J, et al: The Savvy Caregiver Program: the
demonstrated effectiveness of a transportable dementia caregiver
psychoeducation program. J Gerontol Nurs 33:30–36, 2007 17378189
Howard RJ, Juszczak E, Ballard CG, et al; CALM-AD Trial Group:
Donepezil for the treatment of agitation in Alzheimer’s disease. N Engl
J Med 357(14):1382–1392, 2007 17914039
Husebo BS, Ballard C, Sandvik R, et al: Efficacy of treating pain to reduce
behavioural disturbances in residents of nursing homes with dementia:
cluster randomised clinical trial. BMJ 343:d4065, 2011 21765198
Inouye SK, Baker DI, Fugal P, et al; HELP Dissemination Project:
Dissemination of the hospital elder life program: implementation,
adaptation, and successes. J Am Geriatr Soc 54(10):1492–1499, 2006
17038065
Inouye SK, Zhang Y, Jones RN, et al: Risk factors for delirium at discharge:
development and validation of a predictive model. Arch Intern Med
167(13):1406–1413, 2007 17620535
Jeste DV, Blazer D, Casey D, et al: ACNP White Paper: update on use of
antipsychotic drugs in elderly persons with dementia.
Neuropsychopharmacology 33(5):957–970, 2008 17637610
Kales HC, Kim HM, Zivin K, et al: Risk of mortality among individual
antipsychotics in patients with dementia. Am J Psychiatry 169(1):71–
79, 2012 22193526
Kales HC, Gitlin LN, Lyketsos CG: Management of neuropsychiatric
symptoms of dementia in clinical settings: recommendations from a
multidisciplinary expert panel. J Am Geriatr Soc 62(4):762–769, 2014
24635665
Kalisvaart KJ, de Jonghe JF, Bogaards MJ, et al: Haloperidol prophylaxis
for elderly hip-surgery patients at risk for delirium: a randomized
placebo-controlled study. J Am Geriatr Soc 53(10):1658–1666, 2005
16181163
Lebert F, Stekke W, Hasenbroekx C, et al: Frontotemporal dementia: a
randomised, controlled trial with trazodone. Dement Geriatr Cogn
Disord 17(4):355–359, 2004 15178953
Leso L, Schwartz TL: Ziprasidone treatment of delirium. Psychosomatics
43(1):61–62, 2002 11927760
Levkoff S, Cleary P, Liptzin B, et al: Epidemiology of delirium: an
overview of research issues and findings. Int Psychogeriatr 3(2):149–
167, 1991 1811770
Little JT, Satlin A, Sunderland T, et al: Sundown syndrome in severely
demented patients with probable Alzheimer’s disease. J Geriatr
Psychiatry Neurol 8(2):103–106, 1995 7794472
Logsdon RG, McCurry SM, Teri L: Evidence-based psychological
treatments for disruptive behaviors in individuals with dementia.
Psychol Aging 22(1):28–36, 2007 17385980
Lonergan E, Luxenberg J: Valproate preparations for agitation in dementia.
Cochrane Database Syst Rev July 8;(3): CD003945, 2009 19588348
Lonergan E, Britton AM, Luxenberg J, et al: Antipsychotics for delirium.
Cochrane Database Syst Rev Apr 18;(2):CD005594, 2007 17443602
Lyketsos CG, Steinberg M, Tschanz JT, et al: Mental and behavioral
disturbances in dementia: findings from the Cache County Study on
Memory in Aging. Am J Psychiatry 157(5):708–714, 2000 10784462
Lyketsos CG, DelCampo L, Steinberg M, et al: Treating depression in
Alzheimer disease: efficacy and safety of sertraline therapy, and the
benefits of depression reduction: the DIADS. Arch Gen Psychiatry
60(7):737–746, 2003 12860778
Maglione M, Ruelaz Maher A, Hu J, et al: Off-Label Use of Atypical
Antipsychotics: An Update. Comparative Effectiveness Review No 43
(Prepared by the Southern California/RAND Evidence-based Practice
Center under Contract No HHSA290-2007-10062-1; AHRQ Publ No
11-EHC087-EF). Rockville, MD, Agency for Healthcare Research and
Quality, 2011
Maneeton B, Maneeton N, Srisurapanont M, et al: Quetiapine versus
haloperidol in the treatment of delirium: a double-blind, randomized,
controlled trial. Drug Des Devel Ther 7:657–667, 2013 23926422
McKeith I, Del Ser T, Spano P, et al: Efficacy of rivastigmine in dementia
with Lewy bodies: a randomised, double-blind, placebo-controlled
international study. Lancet 356(9247):2031–2036, 2000 11145488
Olin JT, Fox LS, Pawluczyk S, et al: A pilot randomized trial of
carbamazepine for behavioral symptoms in treatment-resistant
outpatients with Alzheimer disease. Am J Geriatr Psychiatry 9(4):400–
405, 2001 11739066
Peskind ER, Tsuang DW, Bonner LT, et al: Propranolol for disruptive
behaviors in nursing home residents with probable or possible
Alzheimer disease: a placebo-controlled study. Alzheimer Dis Assoc
Disord 19(1):23–28, 2005 15764868
Pollock BG, Mulsant BH, Rosen J, et al: Comparison of citalopram,
perphenazine, and placebo for the acute treatment of psychosis and
behavioral disturbances in hospitalized, demented patients. Am J
Psychiatry 159(3):460–465, 2002 11870012
Porsteinsson AP, Drye LT, Pollock BG, et al: Effect of citalopram on
agitation in Alzheimer disease: the CitAD randomized clinical trial.
JAMA 311(7):682–691, 2014 24549548
Rodda J, Morgan S, Walker Z: Are cholinesterase inhibitors effective in the
management of the behavioral and psychological symptoms of
dementia in Alzheimer’s disease? A systematic review of randomized,
placebo-controlled trials of donepezil, rivastigmine and galantamine.
Int Psychogeriatr 21(5):813–824, 2009 19538824
Schneider LS, Dagerman K, Insel PS: Efficacy and adverse effects of
atypical antipsychotics for dementia: meta-analysis of randomized,
placebo-controlled trials. Am J Geriatr Psychiatry 14(3):191–210, 2006
16505124
Shankar KN, Hirschman KB, Hanlon AL, et al: Burden in caregivers of
cognitively impaired elderly adults at time of hospitalization: a cross-
sectional analysis. J Am Geriatr Soc 62(2):276–284, 2014 24502827
Snyder L: Perspectives: A Newsletter for Individuals Diagnosed With
Alzheimer’s Disease. San Diego, CA, Alzheimer’s Disease Research
Center, University of California, San Diego, 2001, p 2
Tariot PN, Erb R, Podgorski CA, et al: Efficacy and tolerability of
carbamazepine for agitation and aggression in dementia. Am J
Psychiatry 155(1):54–61, 1998 9433339
Tariot PN, Schneider LS, Mintzer JE et al: Safety and tolerability of
divalproex sodium in the treatment of signs and symptoms of mania in
elderly patients with dementia: results of a double-blind, placebo-
controlled trial. Current Therapeutic Research 62(1):51–67, 2001
Tariot PN, Raman R, Jakimovich L, et al; Alzheimer’s Disease Cooperative
Study; Valproate Nursing Home Study Group: Divalproex sodium in
nursing home residents with possible or probable Alzheimer Disease
complicated by agitation: a randomized, controlled trial. Am J Geriatr
Psychiatry 13(11):942–949, 2005 16286437
Teri L: Behavior and caregiver burden: behavioral problems in patients with
Alzheimer disease and its association with caregiver distress.
Alzheimer Dis Assoc Disord 11(Suppl 4):S35-S38, 1997 9339271
Teri L, Logsdon RG, Peskind E, et al; Alzheimer’s Disease Cooperative
Study: Treatment of agitation in AD: a randomized, placebo-controlled
clinical trial. Neurology 55(9):1271–1278, 2000 11087767
Teri L, Huda P, Gibbons L, et al: STAR: a dementia-specific training
program for staff in assisted living residences. Gerontologist
45(5):686–693, 2005 16199404
Tractenberg RE, Gamst A, Weiner MF, et al: Frequency of behavioral
symptoms characterizes agitation in Alzheimer’s disease. Int J Geriatr
Psychiatry 16(9):886–891, 2001 11571769
Wang LY, Shofer JB, Rohde K, et al: Prazosin for the treatment of
behavioral symptoms in patients with Alzheimer disease with agitation
and aggression. Am J Geriatr Psychiatry 17(9):744–751, 2009
19700947
Wilcock GK, Ballard CG, Cooper JA, et al: Memantine for
agitation/aggression and psychosis in moderately severe to severe
Alzheimer’s disease: a pooled analysis of 3 studies. J Clin Psychiatry
69(3):341–348, 2008 18294023
Suggested Readings
Jeste DV, Blazer D, Casey D, et al: ACNP White Paper: update on use of
antipsychotic drugs in elderly persons with dementia.
Neuropsychopharmacology 33(5):957–970, 2008
Nassisi D, Korc B, Hahn S, et al: The evaluation and management of the
acutely agitated elderly patient. Mt Sinai J Med 73:976–984, 2006
Roger KS: A literature review of palliative care, end of life, and dementia.
Palliat Support Care 4:295–303, 2006
Spira AP, Edelstein BA: Behavioral interventions for agitation in older
adults with dementia: an evaluative review. Int Psychogeriatr 18:195–
225, 2006
Zuidema S, Koopmans R, Verhey F: Prevalence and predictors of
neuropsychiatric symptoms in cognitively impaired nursing home
patients. J Geriatr Psychiatry Neurol 20:41–49, 2007
Psychopharmacology
Benoit H. Mulsant, M.D.
Bruce G. Pollock, M.D., Ph.D.
Antidepressant Medications
Bupropion
Published data supporting the safety and efficacy of bupropion in geriatric
depression are limited to small controlled trials (see Table 20–5) and one
small open study (Steffens et al. 2001). Expert consensus favors the use of
bupropion—alone or as an augmentation agent—in older depressed patients
whose symptoms have not responded to SSRIs or who cannot tolerate them
(Alexopoulos et al. 2001; Buchanan et al. 2006; Mulsant et al. 2001a, 2014).
In particular, bupropion can be helpful for patients who complain of nausea,
diarrhea, unbearable fatigue, or sexual dysfunction during SSRI treatment
(Nieuwstraten and Dolovich 2001; Thase et al. 2005b). Although
augmentation with bupropion has been reported to be helpful in patients who
were partial responders to SSRIs or venlafaxine (Bodkin et al. 1997; Spier
1998), the safety of this combination in older patients has not been
established (Joo et al. 2002). Controlled data on the use of bupropion in
individuals with heart disease (Kiev et al. 1994; Roose et al. 1991), in
smokers (Tashkin et al. 2001), and in persons with neuropathic pain
(Semenchuk et al. 2001) confirm clinical experience that bupropion is
relatively well tolerated by medically ill patients. Bupropion is
contraindicated in patients who have or are at risk for seizure disorders (e.g.,
poststroke patients). However, the sustained-release preparation of
bupropion appears to be associated with a very low incidence of seizure,
comparable to that of other antidepressants (Dunner et al. 1998). Bupropion
also has been associated with the onset of psychosis in case reports (Howard
and Warnock 1999), and the prudent action is to avoid this medication in
psychotic patients or in agitated patients at risk for the development of
psychotic symptoms. The propensity of bupropion to induce psychosis in at-
risk patients has been attributed to its action on dopaminergic
neurotransmission (Howard and Warnock 1999). The same mechanism has
been hypothesized to underlie the association of bupropion with gait
disturbance and falls in some patients (Joo et al. 2002; Szuba and Leuchter
1992).
Bupropion is a moderate inhibitor of CYP2D6 (Kotlyar et al. 2005) (see
Table 20–4). It appears to be metabolized by CYP2B6 (Hesse et al. 2004),
and adverse effects of bupropion such as seizures or gait disturbance may be
more likely in individuals who take drugs that inhibit CYP2B6, such as
fluoxetine or paroxetine (Joo et al. 2002).
Mirtazapine
The antidepressant activity of mirtazapine has been attributed to its blockade
of α2 autoreceptors, resulting in a direct enhancement of noradrenergic
neurotransmission and an increase in the synaptic levels of serotonin (5-
hydroxytryptamine [5-HT]), indirectly enhancing neurotransmission
mediated by serotonin type 1A (5-HT1A) receptors. In addition, like the
antinausea drugs granisetron and ondansetron, mirtazapine inhibits 5-HT2
and 5-HT3 receptors. Thus, mirtazapine could be particularly helpful for
individuals who do not tolerate SSRIs because of sexual dysfunction
(Gelenberg et al. 2000; Montejo et al. 2001), tremor (Pact and Giduz 1999),
or severe nausea (Pedersen and Klysner 1997). In one case series,
mirtazapine was used successfully to treat depression in 19 mixed-age
oncology patients who were receiving chemotherapy (Thompson 2000). It
also has been combined with SSRIs (Pedersen and Klysner 1997); however,
these combinations should be used cautiously because they have been
associated with a serotonin syndrome in an older patient (Benazzi 1998).
The STAR*D study found that a combination of mirtazapine and venlafaxine
XR had modest efficacy in patients with treatment-resistant depression,
comparable to the efficacy of the MAOI tranylcypromine (Rush et al.
2006a); however, only a few STAR*D participants were elderly, and the
safety of this combination has not been established in older patients.
No published placebo-controlled trials and only two comparator-
controlled trials of mirtazapine in geriatric depression have been done (see
Table 20–5). Consistent with this paucity of controlled data, experts favor
the use of mirtazapine as a third-line drug in older depressed patients who
cannot tolerate or whose symptoms have not responded to SSRIs or
venlafaxine (Alexopoulos et al. 2001). Mirtazapine also has been used to
treat depression in frail nursing home patients (Roose et al. 2003) and in
older patients with dementia (Raji and Brady 2001), but there are concerns
about its effect on cognition. It has been shown to impair driving
performance in two placebo- and active comparator–controlled trials in
healthy volunteers (Ridout et al. 2003; Wingen et al. 2005) and to cause
delirium in older patients with organic brain syndromes (Bailer et al. 2000).
This deleterious effect on cognition is possibly a result of mirtazapine’s
antihistaminergic and sedative effects. Other adverse effects of mirtazapine
include weight gain with lipid increase (Nicholas et al. 2003), hyponatremia
(Cheah et al. 2008), and, very rarely, neutropenia or even agranulocytosis
(Hutchison 2001). In a large observational study in older primary care
patients treated for depression, mirtazapine was associated with a higher risk
of stroke and mortality than other commonly prescribed antidepressants
(Coupland et al. 2011); however, in the absence of randomization, one
cannot rule out that this association was due to confounding factors that
could not be controlled for. In March 2014, Merck Canada issued a warning
endorsed by Health Canada that mirtazapine can cause QT prolongation and
torsades de pointes in association with overdose or when other risk factors
for QT prolongation are present (e.g., in patients with cardiovascular disease
or when combined with other medications that can cause QT prolongation)
(Health Canada Advisory 2014).
Trazodone
Trazodone is indicated for the treatment of major depressive disorder, but it
is now almost exclusively used off-label as a hypnotic or a sedative agent
(Bossini et al. 2012) due to its sedative effect associated with antagonism of
the 5-HT2A receptor and, to a lesser extent, the 5-HT2B, 5HT1A, and α1
receptors. To minimize adverse effects, doses should be kept low (e.g., 50–
150 mg at bedtime) when trazodone is used as a hypnotic agent. Some
evidence going back to the early 1990s indicates that trazodone at low to
moderate doses (50–300 mg/day) has an efficacy comparable to that of
haloperidol in the treatment of agitation or aggression in patients with
dementia (Henry et al. 2011; Houlihan et al. 1994; Sultzer et al. 1997; Teri et
al. 2000). In a unique small (N=30) RCT, trazodone (50 mg given at 10
P.M.) was well tolerated, and it was more efficacious than placebo in the
treatment of sleep disturbances of patients with Alzheimer’s disease
(Camargos et al. 2014).
At doses typically used to treat depression (300–600 mg/day), trazodone
antagonism of α1-adrenergic receptors may cause dry mouth, orthostatic
hypotension (with syncope), QT prolongation or arrhythmias, and priapism
(which is rare in older adults). Like other psychotropic medications,
trazodone has been associated with hyponatremia and it can be involved in a
serotonergic syndrome, in particular when combined with SSRIs or SNRIs
or other medications that also affect the serotonergic system. In a large
observational study in older primary care patients treated for depression,
trazodone was associated with the highest risk of all-cause mortality among
11 commonly prescribed antidepressants (Coupland et al. 2011); however, in
the absence of randomization, one cannot rule out that this association was
due to confounding factors that could not be controlled for. Trazodone is
mostly metabolized by CYP3A4. Therefore, its dose should be reduced
when it is coprescribed with medications that inhibit this liver enzyme (see
Table 20–4), and drinking a large quantity of grapefruit juice should be
discouraged in patients taking trazodone.
Nefazodone
Given the absence of any controlled trials of nefazodone in treating geriatric
depression, mediocre outcomes in an open study (Saiz-Ruiz et al. 2002),
potentially problematic drug-drug interactions caused by its strong inhibition
of CYP3A4 (see Table 20–4), and reports that the incidence of hepatic
toxicity or even liver failure is 10- to 30-fold higher with nefazodone than
with other antidepressants (Carvajal García-Pando et al. 2002), nefazodone
should not be used in older patients.
Psychostimulants
Even though some clinicians prescribe psychostimulants for the treatment of
late-life mood disorders, this practice has minimal empirical support. A few
small double-blind trials suggested that methylphenidate is generally well
tolerated and modestly efficacious for medically burdened depressed elders
(Satel and Nelson 1989; Wallace et al. 1995). Methylphenidate also has been
used for the treatment of apathy and anergia associated with late-life
depression or dementia (Herrmann et al. 2008). A small study suggested that
methylphenidate can be used in older depressed patients to augment SSRIs,
which inhibit dopamine release and may contribute to apathy and fatigue
(Lavretsky et al. 2006). The wakefulness-promoting agent modafinil and its
R-enantiomer armodafinil appear to induce a calm alertness through
nondopaminergic mechanisms. These agents have been used to target apathy
and fatigue in patients taking SSRIs (Dunlop et al. 2007; Fava et al. 2007;
Goss et al. 2013) and as adjunctive treatment for negative symptoms of
schizophrenia (Lindenmayer et al. 2013), but there are almost no published
geriatric data for these drugs (Darwish et al. 2011; Varanese et al. 2013).
Caution is advised regarding the possible exacerbation by methylphenidate
and other psychostimulants of anxiety, psychosis, anorexia, or hypertension
and potential interactions with warfarin. Experience with other dopaminergic
medications, such as pergolide, piribedil, pramipexole, and ropinirole, in
elderly patients has been limited, but there have been several encouraging
controlled trials in patients with Parkinson’s disease and depression (Aiken
2007; Barone et al. 2006, 2010; Rektorová et al. 2003) and in elderly
patients with cognitive impairment (Nagaraja and Jayashree 2001).
Antipsychotic Medications
In older adults, as in other age groups, atypical antipsychotics are now being
prescribed as first-line drugs for the treatment of psychotic symptoms of any
etiology. Studies support the efficacy of these agents in the treatment of late-
life schizophrenia and late-onset psychoses (Scott et al. 2011; Suzuki et al.
2011) and in the treatment of behavioral and psychological symptoms of
dementia (Maher et al. 2011; Maher and Theodore 2012). However, use of
these agents in patients with dementia is being questioned (Ballard and
Corbett 2010; Mulsant 2014; Salzman et al. 2008). In 2005, two highly
publicized reports and an FDA warning indicated a nearly twofold increase
in the rate of deaths in older patients with behavioral and psychological
symptoms of dementia treated with atypical antipsychotics when compared
with placebo (Kuehn 2005; Schneider et al. 2005). These reports have led to
a reexamination of the safety of both conventional and atypical
antipsychotics in older patients. Over the past decade, a series of studies
have emphasized their association with mortality (Ballard et al. 2009;
Langballe et al. 2014; Ray et al. 2009; Wang et al. 2005), stroke (Gill et al.
2005; Herrmann et al. 2004), severe hyperglycemia in patients with diabetes
(Lipscombe et al. 2009), fractures (Liperoti et al. 2007), and venous
thromboembolism (Kleijer et al. 2010). The relative safety of atypical
compared with conventional antipsychotics remains unclear: atypical
antipsychotics appears to be associated with lower mortality than typical
antipsychotics (Langballe et al. 2014; Schneider et al. 2005) and they may
cause fewer falls (Hien et al. 2005; Landi et al. 2005) and fewer
extrapyramidal symptoms (Lee et al. 2004; Meagher et al. 2013; Rochon et
al. 2005; van Iersel et al. 2005); however, they may cause more
cerebrovascular events (Percudani et al. 2005), venous thromboembolism
(Liperoti et al. 2005), and pancreatitis (Koller et al. 2003). Given the
increased recognition of the risks associated with the use of antipsychotics in
older patients, clinicians need to consider their potential risks and benefits
for each individual patient (Gauthier et al. 2010; Rabins and Lyketsos 2005).
Antipsychotics should be prescribed only to patients who have failed to
respond to nonpharmacological interventions or alternative medications
(Ballard and Corbett 2010; Mulsant 2014; Sink et al. 2005). When
antipsychotics are prescribed to older patients, the minimal effective dose
should be used for the shortest possible duration (Tsuboi et al. 2011). Their
long-term use is justified when they are used to treat schizophrenia, bipolar
disorder, and possibly major depressive disorder with psychotic features;
discontinuation should be attempted in stable patients with other disorders.
Although antipsychotics can be discontinued safely in most older patients
with dementia, their discontinuation can be associated with poor outcomes,
in particular in older patients who had presented with more severe agitation
or psychosis (Declercq et al. 2013).
Risperidone
Of the atypical antipsychotics currently available in the United States,
risperidone has the most published geriatric data for a variety of conditions
(Schneider et al. 2005, 2006a; Sink et al. 2005; Suzuki et al. 2011). The
efficacy and tolerability of risperidone in the treatment of behavioral and
psychological symptoms of dementia have been reported in several
randomized placebo-controlled trials (e.g., Brodaty et al. 2003; De Deyn et
al. 1999, 2005b; Katz et al. 1999; Schneider et al. 2006a, 2006b; Sink et al.
2005); in randomized comparisons with haloperidol (Chan et al. 2001; De
Deyn et al. 1999; Suh et al. 2004), promazine and olanzapine (Gareri et al.
2004), and olanzapine (Fontaine et al. 2003; Mulsant et al. 2004); and in
many uncontrolled studies or large case series. The efficacy of risperidone in
the treatment of agitation or psychosis is further supported by a placebo-
controlled trial showing that individuals with Alzheimer’s disease whose
agitation or psychosis had responded acutely to risperidone experienced an
increased risk of relapse when they were switched to placebo after 4 months
(hazard ratio 1:9) or 8 months (hazard ratio 4:9) compared with those who
remained on risperidone (Devanand et al. 2012). Taken together, these data
support risperidone as a first choice among antipsychotics for the treatment
of patients with dementia and distressing psychosis or severe agitation.
However, the substantial risks associated with the use of risperidone and
other atypical antipsychotics in these patients—including increased mortality
(number needed to harm=87) and stroke (number needed to harm=53)
(Maher and Theodore 2012; Maher et al. 2011)—should lead to caution.
The efficacy and tolerability of risperidone in the treatment of late-life
schizophrenia are supported by one randomized comparison with olanzapine
(Harvey et al. 2003; Jeste et al. 2003) and one randomized open-label study
of crossover from conventional antipsychotics to risperidone or olanzapine
(Ritchie et al. 2003, 2006). The parallel study showed similar efficacy
between olanzapine and risperidone but more weight gain and less cognitive
improvement with olanzapine. In the crossover study, patients switched to
olanzapine were more likely to complete the switching process and to show
an improvement in psychological quality of life. The results from these two
controlled trials are supported by a large body of uncontrolled data in older
patients with schizophrenia and other psychotic disorders (e.g., Davidson et
al. 2000; Madhusoodanan et al. 1999). In addition, an analysis of 57 patients
with schizophrenia ages 65 years and older who participated in randomized
studies of the long-acting injectable (“depot”) risperidone (Risperdal Consta)
found that it was well tolerated and produced significant symptomatic
improvements (Lasser et al. 2004).
One randomized comparison with haloperidol (Han and Kim 2004) and
some uncontrolled data (e.g., Mittal et al. 2004; Parellada et al. 2004)
support the efficacy and tolerability of risperidone in the treatment of
delirium (Wang et al. 2013). In another RCT, a significantly lower incidence
of delirium was observed in patients given a single 1-mg dose of risperidone
just after cardiac surgery than in those given placebo (Prakanrattana and
Prapaitrakool 2007; Zhang et al. 2013). However, there have been several
case reports of delirium induced by risperidone. One small randomized
comparison with clozapine (N=10) (Ellis et al. 2000) and several open trials
of low-dose risperidone in the treatment of Parkinson’s disease and drug-
induced psychosis or Lewy body dementia have had inconsistent results,
with clear worsening of parkinsonian symptoms in some studies (e.g., Culo
et al. 2010; Ellis et al. 2000; Leopold 2000). Thus, risperidone should be
used with great caution in the treatment of these disorders (Parkinson Study
Group 1999).
As with other atypical antipsychotics, the efficacy and tolerability of
risperidone in younger patients with bipolar disorder (and possibly other
mood disorders) (Andreescu et al. 2006) are well established. However,
given the risks associated with antipsychotics, experts continue to favor the
use of mood stabilizers as first-line agents for older patients with bipolar
disorder, except in the presence of severe mania or mania with psychosis, in
which case they favor combining risperidone, olanzapine, or quetiapine with
a mood stabilizer (Sajatovic et al. 2005b, 2013; Young et al. 2004).
Commonly reported side effects of risperidone include orthostatic
hypotension (on initiation of treatment) and extrapyramidal symptoms that
are dose dependent (Katz et al. 1999). At a given dosage, concentrations of
risperidone (and possibly its active metabolite paliperidone or 9-
hydroxyrisperidone) seem to increase with age (Aichhorn et al. 2005).
Therefore, typical dosages should be between 0.5 and 2 mg/day for older
patients with dementia and lower than 4 mg/day for older patients without
dementia. Of all the atypical antipsychotics, risperidone appears to be the
most likely to be associated with hyperprolactinemia (Kinon et al. 2003).
Risperidone causes only moderate electroencephalographic abnormalities
(Centorrino et al. 2002), and it is rarely associated with cognitive
impairment, probably because of its low affinity for muscarinic receptors
(Chew et al. 2006; Harvey et al. 2003; Mulsant et al. 2004). Like other
antipsychotics, risperidone can cause weight gain, diabetes, or dyslipidemia.
It is more likely to do so than are aripiprazole and ziprasidone but less likely
than are clozapine, olanzapine, and quetiapine (American Diabetes
Association et al. 2004; Feldman et al. 2004; Zheng et al. 2009).
Paliperidone
Paliperidone is the active 9-hydroxy metabolite of risperidone, and therefore
some of its pharmacological action, efficacy, and side effects are similar to
those of risperidone. Its once-daily extended-release formulation takes 24
hours to reach a maximum concentration, and its clearance is not affected by
hepatic impairment or CYP2D6 metabolism but is affected by renal
function. It is the only medication indicated for the treatment of
schizoaffective disorder in the United States. Its efficacy and tolerability in
the treatment of older patients with psychosis is supported by data from 125
subjects ages 65 years and older who participated in three 6-week
registration trials that led to the medication’s approval by the FDA for the
treatment of schizophrenia (e.g., Davidson et al. 2007; Kane et al. 2007).
Otherwise, limited available data support the efficacy and safety of
paliperidone in the treatment of older patients with schizophrenia
(Madhusoodanan and Zaveri 2010): in a 6-week randomized placebo-
controlled trial followed by a 24-week open-label extension, 114 patients
ages 65 years and older (mean age of 70) received paliperidone 3–12 mg/day
or placebo. Discontinuation due to adverse events and weight gain were
similar in the two groups. Half of the patients treated with paliperidone
experienced prolactin elevation, but it was not related to any adverse event.
Changes in efficacy measures were similar in the two groups (Tzimos et al.
2008). Paliperidone has not yet been systematically studied in older patients
with bipolar disorder or dementia, and doses remain speculative for these
populations. The availability of a long-acting injectable form of paliperidone
that requires only monthly injections is an attractive option for patients who
require a long-acting injectable antipsychotic (González-Rodríguez et al.
2014; Rado and Janicak 2012), but its more widespread use is impeded by a
paucity of geriatric data.
Olanzapine
Next to risperidone, olanzapine has the most published geriatric data. Its
efficacy and tolerability in the treatment of behavioral and psychological
symptoms of dementia have been reported in several randomized placebo-
controlled trials (e.g., Clark et al. 2001; De Deyn et al. 2004; Schneider et al.
2006b; Street et al. 2000) and in randomized comparisons with haloperidol
(Verhey et al. 2006), promazine or risperidone (Gareri et al. 2004), and
risperidone (Fontaine et al. 2003; Mulsant et al. 2004). However, a meta-
analysis of published and nonpublished placebo-controlled trials of
olanzapine in the treatment of behavioral and psychological symptoms of
dementia concluded that “olanzapine was not associated with efficacy
overall” (Schneider et al. 2006a, p. 205). More recently, a 2011 review
published by the U.S. Agency for Healthcare Research and Quality focusing
on data available since 2006 concluded that—like aripiprazole and
risperidone—olanzapine is associated with statistically significant small
benefits for older patients with behavioral disturbances associated with
dementia (Maher and Theodore 2012; Maher et al. 2011).
The efficacy and tolerability of olanzapine in the treatment of late-life
schizophrenia have been confirmed in two randomized comparisons with
haloperidol (Barak et al. 2002; Kennedy et al. 2003) and two randomized
comparisons with risperidone (Harvey et al. 2003; Jeste et al. 2003; Ritchie
et al. 2003, 2006). In one RCT in patients with delirium, olanzapine and
haloperidol were found to have comparable efficacy (Skrobik et al. 2004). In
another RCT, a significantly lower incidence of delirium was observed in
patients given 5 mg of olanzapine just before and just after joint replacement
surgery compared with patients given placebo; however, when delirium
occurred, it was longer and more severe in patients who had received
olanzapine (Larsen et al. 2010; Zhang et al. 2013). Caution is needed when
using olanzapine in patients with delirium because some controlled trials
have reported some cognitive worsening in patients with dementia while
taking olanzapine (Kennedy et al. 2005; Mulsant et al. 2004), and several
case reports of delirium induced by olanzapine have been published.
Similarly, the need for caution when olanzapine is used to treat psychosis in
patients with Parkinson’s disease or Lewy body dementia is reinforced by
two comparative trials (Breier et al. 2002; Goetz et al. 2000) and several
open trials or case series (e.g., Marsh et al. 2001; Molho and Factor 1999;
Parkinson Study Group 1999; Walker et al. 1999) that have reported a
significant worsening of motor symptoms in these patients.
The evidence supporting the efficacy and safety of olanzapine in younger
patients with bipolar disorder and other mood disorders (Andreescu et al.
2006; Shelton et al. 2001; Thase 2002) is strong. One large published trial in
which more than half of the randomized patients were age 65 years and older
supports the efficacy and tolerability of olanzapine in the treatment of major
depressive disorder with psychotic features (Meyers et al. 2009). Otherwise,
there is a paucity of data relevant to older individuals with mood disorders
(Sajatovic et al. 2005a, 2005b; Young et al. 2004). Similarly, very few
geriatric data are available on the rapidly dissolving or the intramuscular
preparations of olanzapine (Belgamwar and Fenton 2005).
On review of all evidence available in 2004, a consensus conference
concluded that among the atypical antipsychotics, clozapine and olanzapine
were associated with the highest risk for diabetes and caused the greatest
weight gain and dyslipidemia (American Diabetes Association et al. 2004).
Limited geriatric data show a similar higher risk of metabolic problems in
older patients (Feldman et al. 2004; Micca et al. 2006; Zheng et al. 2009).
Other common side effects include sedation and gait disturbance.
Extrapyramidal symptoms appear to be dose dependent and are rare at the
lower dosages typically used in older patients (5–10 mg/day). Olanzapine
also has been associated with electroencephalographic abnormalities
(Centorrino et al. 2002), and its strong blocking of the muscarinic receptor
(Chew et al. 2005, 2006; Mulsant et al. 2003) (Table 20–6) may explain why
it has been associated with the following: constipation in a large series of
long-term-care patients (Martin et al. 2003); an inverted dose-response
relationship, with lower efficacy in older agitated or psychotic patients with
dementia randomized to 15 mg/day than in those randomized to 5 mg/day,
suggesting that higher doses may be toxic in these patients (Street et al.
2000); a differential cognitive effect from risperidone in randomized trials
involving older patients with schizophrenia (Harvey et al. 2003) or dementia
(Mulsant et al. 2004); worsening of cognition in a large placebo-controlled
trial in older nonagitated, nonpsychotic patients with Alzheimer’s disease
(Kennedy et al. 2005); and frank delirium in some clinical cases. Individuals
who are older, female, or nonsmokers or who are taking a drug that inhibits
CYP1A2 (e.g., fluvoxamine or ciprofloxacin) have higher concentrations of
olanzapine and may be at higher risk for adverse effects (Gex-Fabry et al.
2003). Because of olanzapine’s adverse-effect profile, experts do not
recommend it as a first-line antipsychotic in older patients at special risk for
anticholinergic or metabolic adverse effects (Bell et al. 2010).
Quetiapine
Results of several randomized placebo-controlled trials of quetiapine in
older patients with behavioral and psychological symptoms of dementia are
inconclusive (Cheung and Stapelberg 2011; Schneider et al. 2006a). For
instance, in a large trial of 333 institutionalized participants, quetiapine 200
mg/day (but not 100 mg/day) differed from placebo on global impressions
and positive symptom ratings but not on the important primary outcome
measures of agitation and psychosis (Zhong et al. 2007).
Clozapine
Clozapine is still considered the drug of choice for younger patients with
treatment-refractory schizophrenia, and one small case series suggested that
it can be similarly helpful in older patients for the treatment of primary
psychotic disorders refractory to other treatments (Sajatovic et al. 1997). An
RCT comparing clozapine and chlorpromazine in older patients with
schizophrenia (Howanitz et al. 1999) and one large case series (Barak et al.
1999) also supported the use of clozapine in moderate dosages (i.e.,
approximately 50–200 mg/day) in older patients with primary psychotic
disorders. The strongest published geriatric studies of clozapine are focused
on the treatment of drug-induced psychosis in patients with Parkinson’s
disease (Ellis et al. 2000; Goetz et al. 2000; Parkinson Study Group 1999).
The results of these studies suggest that clozapine at low dosages (12.5–50
mg/day) could be the preferred treatment for this condition (Parkinson Study
Group 1999). However, the use of clozapine in older individuals is severely
limited because of its significant hematological, anticholinergic (including
severe constipation and ileus associated with fatalities), neurological (e.g.,
seizures), cognitive, metabolic, and cardiac adverse effects (Alvir et al.
1993; Bishara and Taylor 2014; Centorrino et al. 2002; Chew et al. 2006;
Hibbard et al. 2009; O’Connor et al. 2010; Rajji et al. 2010).
Aripiprazole
Aripiprazole has partial dopamine type 2 (D2) receptor agonist properties
(i.e., in high dopaminergic states it acts as an antagonist, and in low
dopaminergic states it acts as an agonist). This may explain why it is
unlikely to cause extrapyramidal side effects or prolactin elevation
(associated with osteoporosis), even at high D2 receptor occupancy (Mamo
et al. 2007). It has only moderate affinity to the adrenergic α1 receptor and
histamine H1 receptor and negligible affinity to the muscarinic receptor
(Chew et al. 2006). As a result, orthostatic hypotension and
antihistaminergic and anticholinergic adverse effects are less likely to occur
than with other atypical agents. However, akathisia may be a common side
effect in older patients (Coley et al. 2009; Sheffrin et al. 2009). Several
randomized placebo-controlled trials of aripiprazole in older patients with
behavioral and psychological symptoms of dementia have been published
(De Deyn et al. 2005a; Mintzer et al. 2007; Streim et al. 2008). Recent
expert opinions (De Deyn et al. 2013; Herrmann et al. 2013b) are congruent
with a meta-analysis of these trials that concluded that “efficacy on rating
scales was observed by meta-analysis for aripiprazole” (Schneider et al.
2006a, p. 191) and with a 2011 review published by the U.S. Agency for
Healthcare Research and Quality that reached a similar conclusion (Maher
and Theodore 2012; Maher et al. 2011).
Aripiprazole is approved by the FDA for the treatment of manic or mixed
episodes associated with bipolar disorder and as an adjunctive treatment for
major depressive disorder. Lenze, Mulsant, Reynolds, and their collaborators
have completed a large federally funded randomized placebo-controlled trial
of aripiprazole augmentation of venlafaxine XR in older patients with major
depression who did not respond to venlafaxine XR monotherapy (300
mg/day). As of November 2014, the results of this study had not yet been
published. However, two small prospective open studies (Sajatovic et al.
2008; Sheffrin et al. 2009) and analyses of pooled geriatric data (Steffens et
al. 2011; Suppes et al. 2008) support the efficacy and tolerability of
aripiprazole augmentation in older patients with major depression that does
not respond fully to an antidepressant.
Ziprasidone
On the basis of ziprasidone’s lower effect on glucose, lipids, and weight
(American Diabetes Association et al. 2004) and its lack of affinity for the
muscarinic receptor (Chew et al. 2006) (see Table 20–6) and thus its low
potential to cause cognitive impairment, ziprasidone is an attractive
medication for older patients with psychosis. However, geriatric data on oral
ziprasidone remain limited (Berkowitz 2003; Wilner et al. 2000). Three
published studies on the use of intramuscular ziprasidone found no adverse
cardiovascular or electrocardiographic changes in a small number of older
patients (Greco et al. 2005; Kohen et al. 2005; Rais et al. 2010). However,
after thioridazine, ziprasidone remains the antipsychotic that is most likely to
be associated with QT prolongation (Wenzel-Seifert et al. 2011). Thus, in the
absence of systematic geriatric studies, ziprasidone should be used with
caution in older patients and should be avoided in patients with cardiac
disease or in those who take other drugs associated with QT prolongation.
Mood Stabilizers
As a class, mood stabilizers are high-risk medications for older patients.
There is a paucity of controlled studies and an abundance of concerns
regarding the drugs’ potential toxicity, problematic side effects, and drug
interactions. Beyond their approved indications, anticonvulsants are also
used in the management of agitation accompanying dementia. Currently, no
consensus exists as to which drug should be preferred as a first-line mood
stabilizer in older individuals with bipolar disorder or secondary mania
(Sajatovic et al. 2005b; Shulman 2010; Young et al. 2004). However, the
neuroprotective properties of lithium (Foland et al. 2008; Germaná et al.
2010; Hajek et al. 2012a, 2012b; Macritchie et al. 2010) and the favorable
cognitive effects of lamotrigine (Gualtieri and Johnson 2006) make them
attractive agents for older patients with bipolar disorder (D’Souza et al.
2011; Sajatovic et al. 2013).
Lithium
Lithium continues to be used in older patients for the treatment of bipolar
disorder (D’Souza et al. 2011; Shulman 2010) or, less commonly, as an
augmentation agent in treatment-resistant depression (Cooper et al. 2011;
Flint and Rifat 2001; Ross 2008) and for the prevention of depressive relapse
following electroconvulsive therapy (Sackeim et al. 2001). Several
publications have described the design and the characteristics of the
participants in the first RCT of the pharmacotherapy of manic or mixed
episodes in older patients with bipolar disorder (Al Jurdi et al. 2012; Beyer
et al. 2014; Young et al. 2010). Although the results of this randomized
comparison of the efficacy and tolerability of divalproex and lithium have
been reported at several scientific meetings, they have not yet been
published. Available data from open and controlled trials suggest that
lithium is efficacious in the acute treatment and prophylaxis of mania in
older patients (D’Souza et al. 2011; Sajatovic et al. 2005a; Shulman 2010);
however, age-related reductions in renal clearance and decreased total body
water significantly affect the pharmacokinetics of lithium in older patients,
increasing the risk of toxicity (D’Souza et al. 2011). Medical comorbidities
common in late life—such as impaired renal function, hyponatremia,
dehydration, and heart failure—further exacerbate the risk of toxicity
(D’Souza et al. 2011; Sajatovic et al. 2006, 2013). Thiazide diuretics,
angiotensin-converting enzyme inhibitors, and NSAIDs may precipitate
toxicity by further diminishing the renal clearance of lithium. Lithium
toxicity can produce persistent central nervous system impairment or be
fatal: it is a medical emergency that requires careful correction of fluid and
electrolyte imbalances and that may require administration of mannitol (or
even hemodialysis) to increase lithium excretion.
Older patients require lower lithium dosages than do younger patients to
produce similar serum lithium levels, and their lithium levels, electrolytes,
and thyroid-stimulating hormone should be monitored regularly (D’Souza et
al. 2011; Rej et al. 2014a). Also, older persons are more sensitive to
neurological side effects of lithium and experience them at lower lithium
levels. This sensitivity may be a consequence of increased permeability of
the blood-brain barrier and subtle changes in sodium-lithium
countertransport, resulting in a higher ratio of brain-to-serum concentration
in older patients than in younger patients (Forester et al. 2009).
Neurotoxicity may manifest as coarse tremor, slurred speech, ataxia,
hyperreflexia, and muscle fasciculations. In vitro, lithium has moderate
anticholinergic activity (Chew et al. 2008). This may explain why cognitive
impairment has been observed with levels well below 1 mEq/L and why
frank delirium has been reported with levels as low as 1.5 mEq/L (Sproule et
al. 2000). Consequently, treatment in older patients may require lithium
levels to be kept as low as 0.4–0.8 mEq/L. In addition, despite the absence of
definite evidence, concerns about the association between long-term use of
lithium and renal disease and the possible causal role of lithium in this
association remain (Rej et al. 2014b). Despite its potential toxicity, lithium
remains an important drug in the treatment of bipolar disorder and treatment-
resistant depression in late life because of its potential effect on suicidality
(Müller-Oerlinghausen and Lewitzka 2010) and its potential neuroprotective
properties (Foland et al. 2008; Germaná et al. 2010; Hajek et al. 2012a,
2012b; Macritchie et al. 2010).
Anticonvulsants
Anticonvulsants are used as alternatives to lithium in the treatment of bipolar
disorder (Young et al. 2004) and as third-line alternatives to antipsychotics
and SSRIs for the management of agitation associated with dementia
(Herrmann et al. 2013b). There may be a subgroup of patients with bipolar
disorder with dysphoria or rapid cycling who respond poorly to lithium but
do well with anticonvulsants (Post et al. 1998).
Divalproex
Divalproex, a compound of sodium valproate and valproic acid in an enteric-
coated form, is a broad-spectrum anticonvulsant approved by the FDA for
the treatment of acute manic or mixed episodes associated with bipolar
disorder, with or without psychotic features. It also may be efficacious in the
treatment of bipolar depression (Bond et al. 2010). Small case series have
suggested that divalproex is relatively well tolerated by older patients with
bipolar disorder (Kando et al. 1996; Noaghiul et al. 1998). Nonetheless, it
should not be used in patients with dementia because of six negative
placebo-controlled trials showing that compared with placebo, divalproex is
not more effective but is more toxic—including potential neurotoxicity and
cognitive toxicity—in older patients with dementia and agitation (Herrmann
et al. 2013b; Sink et al. 2005; Tariot et al. 2005, 2011). Sedation, nausea,
weight gain, and hand tremors are common dose-related side effects.
Reversible thrombocytopenia can occur in up to half of the elderly patients
taking divalproex and may ensue at lower total drug levels than in younger
patients (Fenn et al. 2006). Other dose-related adverse effects include
reversible elevations in liver enzymes and transient elevations in blood
ammonia levels. However, liver failure and pancreatitis are rare. Divalproex
has other metabolic effects of concern to aging patients, such as increases in
bone turnover and reductions of serum folates, with concomitant elevations
in plasma homocysteine concentrations (Sato et al. 2001; Schwaninger et al.
1999).
The pharmacokinetics of valproate vary according to formulation, and
valproic acid, divalproex sodium, and its extended-release (ER) preparation
are not interchangeable. Valproate is metabolized principally by
mitochondrial β-oxidation and secondarily by the cytochrome P450 system;
typical half-lives are in the range of 5–16 hours and are not affected by aging
alone. Concomitant administration of valproate will increase concentrations
of carbamazepine, diazepam, lamotrigine, phenobarbital, and primidone.
Conversely, concurrent administration of carbamazepine, lamotrigine,
phenytoin, and topiramate may decrease levels of valproate. Fluoxetine and
erythromycin may potentiate the effects of valproate. Changes in protein
binding as a result of drug interactions are no longer considered clinically
important beyond causing the misinterpretation of total (i.e., free and bound)
drug levels (Benet and Hoener 2002). Because valproate binding to plasma
proteins is generally reduced in the elderly, use of free drug levels correlates
better with adverse effects (Fenn et al. 2006).
Lamotrigine
Lamotrigine is approved by the FDA for the maintenance treatment of
bipolar I disorder to prevent mood episodes (depressive, manic, or mixed
episodes) and it is considered a first-line agent for the treatment of bipolar
depression in adults (Fenn et al. 2006). Pooled geriatric data from two
randomized placebo-controlled trials support the efficacy of lamotrigine in
preventing bipolar depression in older patients (Sajatovic et al. 2005a, 2007).
Open studies and case reports suggest a role for lamotrigine in the treatment
of bipolar depression (Sajatovic et al. 2011), and possibly bipolar mania and
dementia-related agitation as well (Sajatovic et al. 2007).
In contrast with many other mood stabilizers and antidepressants,
lamotrigine does not seem to be associated with weight gain or to cause
significant drug interactions. It is also less likely than other mood stabilizers
to be associated with cognitive impairment (Gualtieri and Johnson 2006).
Typically, lamotrigine is well tolerated, but somnolence and rashes have
been observed in older patients. Rashes are the most common reason for
discontinuation, but their incidence is less frequent with lamotrigine than
with carbamazepine (Fenn et al. 2006). Severe rashes, including Stevens-
Johnson syndrome or toxic epidermal necrolysis, have been observed in
about 0.3% of adult patients (Messenheimer 1998). At the first sign of rash
or other evidence of hypersensitivity (e.g., fever, lymphadenopathy),
lamotrigine should be discontinued, and the patient should be evaluated. The
incidence of rashes can be reduced by using a low initial dose and a slow
titration.
Because valproate increases lamotrigine concentration, the initial and
target doses need to be halved in patients who are receiving divalproex and
the titration of lamotrigine needs to be slowed down. Conversely,
carbamazepine approximately halves lamotrigine concentrations, and the
initial lamotrigine dose needs to be doubled in patients who are receiving
carbamazepine.
Topiramate
Early reports of the efficacy of topiramate in younger patients with bipolar
disorder have not been confirmed by subsequent studies (Sommer et al.
2007). In younger patients, topiramate is one of the few psychotropic
medications that have been associated with weight loss. However, it also has
been associated with cognitive impairment that can be severe enough to
interfere with functioning (Gualtieri and Johnson 2006). Additionally,
because of the paucity of data pertaining to use of topiramate in older
psychiatric patients (Sommer et al. 2007), its use cannot be recommended in
these patients.
Buspirone
The anxiolytic buspirone, a partial 5-HT1A agonist, is rarely used.
Nevertheless, it may be beneficial for some patients with generalized anxiety
disorder or as an augmentation agent in treatment-resistant depression (Flint
2005; Trivedi et al. 2006). It appears to be well tolerated by elderly patients
without the sedation or addiction liability of the benzodiazepines (Steinberg
1994). Therefore, it may be helpful for some older patients who are prone to
falls, confusion, or chronic lung disease. Nonetheless, buspirone may take
several weeks to exert an anxiolytic effect, has no cross-tolerance with
benzodiazepines, and may cause dizziness, headache, or nervousness (Strand
et al. 1990). It is of limited use for panic or obsessive-compulsive disorders.
The pharmacokinetics of buspirone are not affected by age or gender, but
coadministration with verapamil, diltiazem, erythromycin, or itraconazole
will substantially increase buspirone concentrations, and its combination
with serotonergic medications may result in the serotonin syndrome
(Mahmood and Sahajwalla 1999).
Cognitive Enhancers
Cholinesterase Inhibitors
In addition to memantine (discussed in the next subsection), four
cholinesterase inhibitors have received FDA approval for the symptomatic
improvement of Alzheimer’s disease. Table 20–7 describes three of these
drugs: donepezil, galantamine, and rivastigmine. The fourth, tacrine, is no
longer recommended because of its potential hepatotoxic effects.
Cholinesterase inhibitors produce modest improvements in cognition and
function in patients with Alzheimer’s disease (Hansen et al. 2008;
Herrmann et al. 2013b), including those with severe Alzheimer’s disease
(Herrmann et al. 2013b; Howard et al. 2012; Winblad et al. 2006).
Therefore, a trial with a cholinesterase inhibitor is recommended in most
patients with Alzheimer’s disease; in the absence of convincing evidence
that one of the three cholinesterase inhibitors is more effective than the
others, the selection of a specific drug is based on its pharmacokinetic and
adverse effects profile (see Table 20–7) (Herrmann et al. 2013b).
Cholinesterase inhibitors have modest benefit of uncertain clinical
significance in vascular dementia (Kavirajan and Schneider 2007). They
may also have a role in the management of other cognitive disorders, such
as Lewy body dementia (Gustavsson et al. 2009; Rolinski et al. 2012),
dementia with Parkinson’s disease (Herrmann et al. 2013b: Rolinski et al.
2012), frontotemporal dementia (Herrmann et al. 2013b), mild cognitive
impairment (Diniz et al. 2009; Doody et al. 2009), and cognitive
impairment associated with late-life depression (Reynolds et al. 2011).
Available data are not consistent, however, and there is no agreement on the
role of cholinesterase inhibitors in the treatment or prevention of behavioral
or psychological symptoms associated with dementia (Freund-Levi et al.
2014; Gauthier et al. 2010; Herrmann et al. 2013b; Howard et al. 2007;
Lockhart et al. 2011; Rodda et al. 2009; Sink et al. 2005).
No evidence suggests that cholinesterase inhibitors alter the underlying
neuropathology of Alzheimer’s disease or its eventual progression. Indeed,
a rapid symptomatic deterioration may occur when cholinesterase inhibitors
are discontinued (Scarpini et al. 2011). In patients with diminished
cognitive reserve, even small anticholinergic effects can substantially
impair cognition (Mulsant et al. 2003; Nebes et al. 2005). Drugs with potent
anticholinergic effects directly antagonize cholinesterase inhibitors (Chew
et al. 2008; Modi et al. 2009). Thus, it is imperative that unnecessary
anticholinergic medications be discontinued before initiating a
cholinesterase inhibitor (Lu and Tune 2003; Modi et al. 2009).
The main adverse effects of cholinesterase inhibitors are concentration
dependent and result from their central and peripheral cholinergic actions.
Nausea, diarrhea, weight loss, bradycardia, syncope, and nightmares are
associated with all of the cholinesterase inhibitors and may lead to their
discontinuation (see Table 20–7; Gill et al. 2009; Hernandez et al. 2009;
Park-Wyllie et al. 2009); gastrointestinal adverse effects may be less
frequent with donepezil (Mayeux 2010). However, despite theoretical
concerns, the use of cholinesterase inhibitors appears to be safe in patients
with chronic airway disorders (Thacker and Schneeweiss 2006). Finally, in
a placebo-controlled study in older patients with a major depressive
disorder receiving maintenance antidepressant pharmacotherapy, donepezil
was associated with a higher rate of recurrence of depression than placebo
(Reynolds et al. 2011). With these adverse effects in mind, clinicians should
be aware of the drugs’ specific pathways of elimination and potential
pharmacokinetic drug interactions with CYP2D6 or CYP3A4 inhibitors and
with CYP3A4 inducers when prescribing donepezil and galantamine
(Pilotto et al. 2009; Seritan 2008). Rivastigmine is affected by renal
function, and FDA warnings have emphasized the need for careful dose
titration (and retitration if restarting) to prevent severe vomiting (Birks et al.
2009).
Conclusion
Substantial evidence now exists to guide the use of psychotropic
medications in older persons. Most available data pertain to: the use of
SSRI or SNRI antidepressants in the treatment of major depressive
disorders; the use of atypical antipsychotics in the treatment of behavioral
and psychological symptoms of dementia; and the use of cognitive
enhancers in the treatment of cognitive impairment due to Alzheimer’s
disease. Existing data support the short-term and long-term efficacy and the
relative safety of SSRI and SNRI antidepressants in the treatment of most
older individuals with a major depressive disorder or an anxiety disorder. In
contrast, the risks associated with the use of antipsychotics outweigh their
potential benefits in many older persons presenting with behavioral and
psychological symptoms of dementia. Cognitive enhancers appear
relatively safe in older individuals with dementia but their efficacy is
modest. Pharmacoepidemiological data show that benzodiazepines continue
to be prescribed to a larger number of older individuals despite their
toxicity. Empirical data to guide the use of mood stabilizers or
antipsychotics in older persons with bipolar disorder or schizophrenia are
lacking. Similarly, better data are needed to inform the selection,
sequencing, and combination psychotropic medications in older persons
whose symptoms do not respond to first-line pharmacological interventions.
Key Points
• Substantial evidence supports the short-term and long-term efficacy and
the relative safety of SSRI and SNRI antidepressants in the treatment of
most older persons with a major depressive disorder or an anxiety
disorder.
• In contrast, the risks associated with the use of antipsychotics outweigh
their potential benefits in many older persons presenting with behavioral
and psychological symptoms of dementia.
• Cognitive enhancers appear relatively safe in older persons with
dementia but their efficacy is modest.
• Pharmacoepidemiological data show that benzodiazepines continue to be
prescribed to many older persons despite their toxicity.
• Empirical data to guide the use of mood stabilizers or antipsychotics in
older persons with bipolar disorder or schizophrenia are lacking.
• Similarly, better data are needed to inform the selection, sequencing, and
combination of psychotropic medications in older persons whose
symptoms do not respond to first-line pharmacological interventions.
References
Aarsland D, Ballard C, Walker Z, et al: Memantine in patients with
Parkinson’s disease dementia or dementia with Lewy bodies: a double-
blind, placebo-controlled, multicentre trial. Lancet Neurol 8(7):613–
618, 2009 19520613
Aichhorn W, Weiss U, Marksteiner J, et al: Influence of age and gender on
risperidone plasma concentrations. J Psychopharmacol 19(4):395–401,
2005 15982995
Aiken CB: Pramipexole in psychiatry: a systematic review of the literature.
J Clin Psychiatry 68(8):1230–1236, 2007 17854248
Alexopoulos GS, Katz IR, Reynolds CF 3rd, et al: Pharmacotherapy of
depression in older patients: a summary of the expert consensus
guidelines. J Psychiatr Pract 7(6):361–376, 2001 15990550
Al Jurdi RK, Schulberg HC, Greenberg RL, et al: Characteristics associated
with inpatient versus outpatient status in older adults with bipolar
disorder. J Geriatr Psychiatry Neurol 25(1):62–68, 2012 22467848
Allard P, Gram L, Timdahl K, et al: Efficacy and tolerability of venlafaxine
in geriatric outpatients with major depression: a double-blind,
randomised 6-month comparative trial with citalopram. Int J Geriatr
Psychiatry 19(12):1123–1130, 2004 15526307
Altamura AC, De Novellis F, Guercetti G, et al: Fluoxetine compared with
amitriptyline in elderly depression: a controlled clinical trial. Int J Clin
Pharmacol Res 9(6):391–396, 1989 2699465
Alvir JM, Lieberman JA, Safferman AZ, et al: Clozapine-induced
agranulocytosis: incidence and risk factors in the United States. N Engl
J Med 329(3):162–167, 1993 8515788
American Diabetes Association; American Psychiatric Association;
American Association of Clinical Endocrinologists; North American
Association for the Study of Obesity: Consensus development
conference on antipsychotic drugs and obesity and diabetes. Diabetes
Care 27(2):596–601, 2004 14747245
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 60(4):
616–631, 2012 22376048
Andersen G, Vestergaard K, Lauritzen L: Effective treatment of poststroke
depression with the selective serotonin reuptake inhibitor citalopram.
Stroke 25(6):1099–1104, 1994 8202964
Andreescu C, Mulsant BH, Rothschild AJ, et al: Pharmacotherapy of major
depression with psychotic features: what is the evidence? Psychiatr
Ann 35:31–38, 2006
Aragona M, Inghilleri M: Increased ocular pressure in two patients with
narrow angle glaucoma treated with venlafaxine. Clin Neuropharmacol
21(2):130–131, 1998 9579301
Arranz FJ, Ros S: Effects of comorbidity and polypharmacy on the clinical
usefulness of sertraline in elderly depressed patients: an open
multicentre study. J Affect Disord 46(3):285–291, 1997 9547126
Atalay A, Turhan N, Aki OE: A challenging case of syndrome of
inappropriate secretion of antidiuretic hormone in an elderly patient
secondary to quetiapine. South Med J 100(8):832–833, 2007 17713312
Auerbach AD, Vittinghoff E, Maselli J, et al: Perioperative use of selective
serotonin reuptake inhibitors and risks for adverse outcomes of surgery.
JAMA Intern Med 173(12):1075–1081, 2013 23699725
Bailer U, Fischer P, Küfferle B, et al: Occurrence of mirtazapine-induced
delirium in organic brain disorder. Int Clin Psychopharmacol
15(4):239–243, 2000 10954066
Ballard C, Corbett A: Management of neuropsychiatric symptoms in people
with dementia. CNS Drugs 24(9):729–739, 2010 20806986
Ballard C, Hanney ML, Theodoulou M, et al; DART-AD investigators: The
dementia antipsychotic withdrawal trial (DART-AD): long-term
follow-up of a randomised placebo-controlled trial. Lancet Neurol
8(2):151–157, 2009 19138567
Ballokova A, Peel NM, Fialova D, et al: Use of benzodiazepines and
association with falls in older people admitted to hospital: a prospective
cohort study. Drugs Aging 31(4):299–310, 2014 24566878
Banerjee S, Hellier J, Dewey M, et al: Sertraline or mirtazapine for
depression in dementia (HTA-SADD): a randomised, multicentre,
double-blind, placebo-controlled trial. Lancet 378(9789):403–411,
2011 21764118
Barak Y, Wittenberg N, Naor S, et al: Clozapine in elderly psychiatric
patients: tolerability, safety, and efficacy. Compr Psychiatry 40(4):320–
325, 1999 10428193
Barak Y, Shamir E, Zemishlani H, et al: Olanzapine vs. haloperidol in the
treatment of elderly chronic schizophrenia patients. Prog
Neuropsychopharmacol Biol Psychiatry 26(6):1199–1202, 2002
12452546
Barak Y, Plopski I, Tadger S, et al: Escitalopram versus risperidone for the
treatment of behavioral and psychotic symptoms associated with
Alzheimer’s disease: a randomized double-blind pilot study. Int
Psychogeriatr 23(9):1515–1519, 2011 21492498
Barbui C, Esposito E, Cipriani A: Selective serotonin reuptake inhibitors
and risk of suicide: a systematic review of observational studies. CMAJ
180(3):291–297, 2009 19188627
Barone P, Scarzella L, Marconi R, et al; Depression/Parkinson Italian Study
Group: Pramipexole versus sertraline in the treatment of depression in
Parkinson’s disease: a national multicenter parallel-group randomized
study. J Neurol 253(5): 601–607, 2006 16607468
Barone P, Poewe W, Albrecht S, et al: Pramipexole for the treatment of
depressive symptoms in patients with Parkinson’s disease: a
randomised, double-blind, placebo-controlled trial. Lancet Neurol 9(6):
573–580, 2010 20452823
Baruch Y, Tadger S, Plopski I, et al: Asenapine for elderly bipolar manic
patients. J Affect Disord 145(1):130–132, 2013 22877962
Beers MH: Explicit criteria for determining potentially inappropriate
medication use by the elderly: an update. Arch Intern Med
157(14):1531–1536, 1997 9236554
Belgamwar RB, Fenton M: Olanzapine IM or Velotab for acutely
disturbed/agitated people with suspected serious mental illnesses.
Cochrane Database Syst Rev (2):CD003729, 2005 15846678
Bell JS, Taipale HT, Soini H, Pitkälä KH: Sedative load among long-term
care facility residents with and without dementia: a cross-sectional
study. Clin Drug Investig 30(1):63–70, 2010 19995099
Benazzi F: Urinary retention with venlafaxine-haloperidol combination.
Pharmacopsychiatry 30(1):27, 1997 9065967
Benazzi F: Serotonin syndrome with mirtazapine-fluoxetine combination.
Int J Geriatr Psychiatry 13(7):495–496, 1998 9695042
Benet LZ, Hoener B: Changes in plasma protein binding have little clinical
relevance. Clin Pharmacol Ther 71:115–121, 2002
Bergh S, Selbæk G, Engedal K: Discontinuation of antidepressants in
people with dementia and neuropsychiatric symptoms (DESEP study):
double blind, randomised, parallel group, placebo controlled trial. BMJ
344:e1566, 2012
Berhan A, Barker A: Vortioxetine in the treatment of adult patients with
major depressive disorder: a meta-analysis of randomized double-blind
controlled trials. BMC Psychiatry 14:276, 2014 25260373
Berkowitz A: Ziprasidone for dementia in elderly patients: case review. J
Psychiatr Pract 9(6):469–473, 2003 15985971
Beyer JL, Greenberg RL, Marino P, et al: Social support in late life mania:
GERI-BD. Int J Geriatr Psychiatry 29(10):1028–1032, 2014 24664811
Billioti de Gage S, Moride Y, Ducruet T, et al: Benzodiazepine use and risk
of Alzheimer’s disease: case-control study. BMJ 349:g5205, 2014
25208536
Birks J, Grimley Evans J, Iakovidou V, et al: Rivastigmine for Alzheimer’s
disease. Cochrane Database Syst Rev (2):CD001191, 2009 19370562
Bishara D, Taylor D: Adverse effects of clozapine in older patients:
epidemiology, prevention and management. Drugs Aging 31(1):11–20,
2014 24338220
Bodkin JA, Lasser RA, Wines JD Jr, et al: Combining serotonin reuptake
inhibitors and bupropion in partial responders to antidepressant
monotherapy. J Clin Psychiatry 58(4):137–145, 1997 9164423
Bond DJ, Lam RW, Yatham LN: Divalproex sodium versus placebo in the
treatment of acute bipolar depression: a systematic review and meta-
analysis. J Affect Disord 124(3):228–234, 2010 20044142
Bondareff W, Alpert M, Friedhoff AJ, et al: Comparison of sertraline and
nortriptyline in the treatment of major depressive disorder in late life.
Am J Psychiatry 157(5):729–736, 2000 10784465
Bose A, Li D, Gandhi C: Escitalopram in the acute treatment of depressed
patients aged 60 years or older. Am J Geriatr Psychiatry 16(1):14–20,
2008 18165459
Bossini L, Casolaro I, Koukouna D, et al: Off-label uses of trazodone: a
review. Expert Opin Pharmacother 13(12):1707–1717, 2012 22712761
Bradley AJ, Lenox-Smith AJ: Does adding noradrenaline reuptake
inhibition to selective serotonin reuptake inhibition improve efficacy in
patients with depression? A systematic review of meta-analyses and
large randomised pragmatic trials. J Psychopharmacol 27(8):740–758,
2013 23832963
Branconnier RJ, Cole JO, Ghazvinian S, et al: Clinical pharmacology of
bupropion and imipramine in elderly depressives. J Clin Psychiatry
44(5 Pt 2):130–133, 1983 6406441
Breier A, Sutton VK, Feldman PD, et al: Olanzapine in the treatment of
dopamimetic-induced psychosis in patients with Parkinson’s disease.
Biol Psychiatry 52(5): 438–445, 2002 12242060
Brodaty H, Ames D, Snowdon J, et al: A randomized placebo-controlled
trial of risperidone for the treatment of aggression, agitation, and
psychosis of dementia. J Clin Psychiatry 64(2):134–143, 2003
12633121
Buchanan D, Tourigny-Rivard MF, Cappeliez P, et al: National Guidelines
for seniors’ mental health: the assessment and treatment of depression.
Can J Geriatr 9 (suppl 2):S52–S58, 2006
Burrows AB, Salzman C, Satlin A, et al: A randomized, placebo-controlled
trial of paroxetine in nursing home residents with non-major
depression. Depress Anxiety 15(3):102–110, 2002 12001178
Camargos EF, Louzada LL, Quintas JL, et al: Trazodone improves sleep
parameters in Alzheimer disease patients: a randomized, double-blind,
and placebo-controlled study. Am J Geriatr Psychiatry 22(12):1565–
1574, 2014 24495406
Carta MG, Zairo F, Mellino G, et al: Add-on quetiapine in the treatment of
major depressive disorder in elderly patients with cerebrovascular
damage. Clin Pract Epidemiol Ment Health 3:28, 2007 18039392
Carvajal García-Pando A, García del Pozo J, Sánchez AS, et al:
Hepatotoxicity associated with the new antidepressants. J Clin
Psychiatry 63(2):135–137, 2002 11874214
Castro VM, Clements CC, Murphy SN, et al: QT interval and
antidepressant use: a cross sectional study of electronic health records.
BMJ 346:f288, 2013 23360890
Centorrino F, Price BH, Tuttle M, et al: EEG abnormalities during treatment
with typical and atypical antipsychotics. Am J Psychiatry 159(1):109–
115, 2002 11772698
Chalon SA, Granier LA, Vandenhende FR, et al: Duloxetine increases
serotonin and norepinephrine availability in healthy subjects: a double-
blind, controlled study. Neuropsychopharmacology 28(9):1685–1693,
2003 12784100
Chan WC, Lam LC, Choy CN, et al: A double-blind randomised
comparison of risperidone and haloperidol in the treatment of
behavioural and psychological symptoms in Chinese dementia patients.
Int J Geriatr Psychiatry 16(12):1156–1162, 2001 11748775
Cheah CY, Ladhams B, Fegan PG: Mirtazapine associated with profound
hyponatremia: two case reports. Am J Geriatr Pharmacother 6(2):91–
95, 2008 18675767
Chen P, Kales HC, Weintraub D, et al: Antidepressant treatment of veterans
with Parkinson’s disease and depression: analysis of a national sample.
J Geriatr Psychiatry Neurol 20(3):161–165, 2007 17712099
Chen Y, Patel NC, Guo JJ, et al: Antidepressant prophylaxis for poststroke
depression: a meta-analysis. Int Clin Psychopharmacol 22(3):159–166,
2007 17414742
Cheung G, Stapelberg J: Quetiapine for the treatment of behavioural and
psychological symptoms of dementia (BPSD): a meta-analysis of
randomised placebo-controlled trials. N Z Med J 124(1336):39–50,
2011 21946743
Chew ML, Mulsant BH, Pollock BG: Serum anticholinergic activity and
cognition in patients with moderate-to-severe dementia. Am J Geriatr
Psychiatry 13(6):535–538, 2005 15956274
Chew ML, Mulsant BH, Pollock BG, et al: A model of anticholinergic
activity of atypical antipsychotic medications. Schizophr Res 88(1–
3):63–72, 2006 16928430
Chew ML, Mulsant BH, Pollock BG, et al: Anticholinergic activity of 107
medications commonly used by older adults. J Am Geriatr Soc
56(7):1333–1341, 2008 18510583
Cipriani A, Furukawa TA, Salanti G, et al: Comparative efficacy and
acceptability of 12 new-generation antidepressants: a multiple-
treatments meta-analysis. Lancet 373(9665):746–758, 2009 19185342
Cipriani A, Koesters M, Furukawa TA, et al: Duloxetine versus other anti-
depressive agents for depression. Cochrane Database Syst Rev
10:CD006533, 2012 23076926
Clark WS, Street JS, Feldman PD, Breier A: The effects of olanzapine in
reducing the emergence of psychosis among nursing home patients
with Alzheimer’s disease. J Clin Psychiatry 62(1):34–40, 2001
11235926
Clayton AH, Kornstein SG, Rosas G, et al: An integrated analysis of the
safety and tolerability of desvenlafaxine compared with placebo in the
treatment of major depressive disorder. CNS Spectr 14(4):183–195,
2009 19407730
Clayton AH, Kornstein SG, Dunlop BW, et al: Efficacy and safety of
desvenlafaxine 50 mg/d in a randomized, placebo-controlled study of
perimenopausal and postmenopausal women with major depressive
disorder. J Clin Psychiatry 74(10): 1010–1017, 2013 24229754
Cohn CK, Shrivastava R, Mendels J, et al: Double-blind, multicenter
comparison of sertraline and amitriptyline in elderly depressed patients.
J Clin Psychiatry 51 (suppl B):28–33, 1990 2258379
Coley KC, Scipio TM, Ruby C, et al: Aripiprazole prescribing patterns and
side effects in elderly psychiatric inpatients. J Psychiatr Pract
15(2):150–153, 2009 19339850
Cooper C, Katona C, Lyketsos K, et al: A systematic review of treatments
for refractory depression in older people. Am J Psychiatry 168(7):681–
688, 2011 21454919
Coupland C, Dhiman P, Morriss R, et al: Antidepressant use and risk of
adverse outcomes in older people: population based cohort study. BMJ
343:d4551, 2011 21810886
Culang ME, Sneed JR, Keilp JG, et al: Change in cognitive functioning
following acute antidepressant treatment in late-life depression. Am J
Geriatr Psychiatry 17(10): 881–888, 2009 19916207
Culo S, Mulsant BH, Rosen J, et al: Treating neuropsychiatric symptoms in
dementia with Lewy bodies: a randomized controlled trial. Alzheimer
Dis Assoc Disord 24(4):360–364, 2010 20625270
Dahmen N, Marx J, Hopf HC, et al: Therapy of early poststroke depression
with venlafaxine: safety, tolerability, and efficacy as determined in an
open, uncontrolled clinical trial. Stroke 30(3):691–692, 1999 10066874
Darwish M, Kirby M, Hellriegel ET, et al: Systemic exposure to
armodafinil and its tolerability in healthy elderly versus young men: an
open-label, multiple-dose, parallel-group study. Drugs Aging
28(2):139–150, 2011 21275439
Davidson J, Watkins L, Owens M, et al: Effects of paroxetine and
venlafaxine XR on heart rate variability in depression. J Clin
Psychopharmacol 25(5):480–484, 2005 16160626
Davidson M, Harvey PD, Vervarcke J, et al: A long-term, multicenter,
open-label study of risperidone in elderly patients with psychosis. On
behalf of the Risperidone Working Group. Int J Geriatr Psychiatry
15(6):506–514, 2000 10861916
Davidson M, Emsley R, Kramer M, et al: Efficacy, safety and early
response of paliperidone extended-release tablets (paliperidone ER):
results of a 6-week, randomized, placebo-controlled study. Schizophr
Res 93(1–3):117–130, 2007 17466492
de Abajo FJ, García-Rodríguez LA: Risk of upper gastrointestinal tract
bleeding associated with selective serotonin reuptake inhibitors and
venlafaxine therapy: interaction with nonsteroidal anti-inflammatory
drugs and effect of acid-suppressing agents. Arch Gen Psychiatry
65(7):795–803, 2008 18606952
Declercq T, Petrovic M, Azermai M, et al: Withdrawal versus continuation
of chronic antipsychotic drugs for behavioural and psychological
symptoms in older people with dementia. Cochrane Database Syst Rev
28(3):CD007726, 2013
De Deyn PP, Rabheru K, Rasmussen A, et al: A randomized trial of
risperidone, placebo, and haloperidol for behavioral symptoms of
dementia. Neurology 53(5):946–955, 1999 10496251
De Deyn PP, Carrasco MM, Deberdt W, et al: Olanzapine versus placebo in
the treatment of psychosis with or without associated behavioral
disturbances in patients with Alzheimer’s disease. Int J Geriatr
Psychiatry 19(2):115–126, 2004 14758577
De Deyn P, Jeste DV, Swanink R, et al: Aripiprazole for the treatment of
psychosis in patients with Alzheimer’s disease: a randomized, placebo-
controlled study. J Clin Psychopharmacol 25(5):463–467, 2005a
16160622
De Deyn PP, Katz IR, Brodaty H, et al: Management of agitation,
aggression, and psychosis associated with dementia: a pooled analysis
including three randomized, placebo-controlled double-blind trials in
nursing home residents treated with risperidone. Clin Neurol Neurosurg
107(6):497–508, 2005b 15922506
De Deyn PP, Drenth AF, Kremer BP, et al: Aripiprazole in the treatment of
Alzheimer’s disease. Expert Opin Pharmacother 14(4):459–474, 2013
23350964
Devanand DP, Pelton GH, Marston K, et al: Sertraline treatment of elderly
patients with depression and cognitive impairment. Int J Geriatr
Psychiatry 18(2):123–130, 2003 12571820
Devanand DP, Juszczak N, Nobler MS, et al: An open treatment trial of
venlafaxine for elderly patients with dysthymic disorder. J Geriatr
Psychiatry Neurol 17(4):219–224, 2004 15533993
Devanand DP, Nobler MS, Cheng J, et al: Randomized, double-blind,
placebo-controlled trial of fluoxetine treatment for elderly patients with
dysthymic disorder. Am J Geriatr Psychiatry 13(1):59–68, 2005
15653941
Devanand DP, Mintzer J, Schultz SK, et al: Relapse risk after
discontinuation of risperidone in Alzheimer’s disease. N Engl J Med
367(16):1497–1507, 2012 23075176
Devlin JW, Roberts RJ, Fong JJ, et al: Efficacy and safety of quetiapine in
critically ill patients with delirium: a prospective, multicenter,
randomized, double-blind, placebo-controlled pilot study. Crit Care
Med 38(2):419–427, 2010 19915454
Devlin JW, Skrobik Y, Riker RR, et al: Impact of quetiapine on resolution
of individual delirium symptoms in critically ill patients with delirium:
a post-hoc analysis of a double-blind, randomized, placebo-controlled
study. Crit Care 15(5):R215, 2011 21923923
Devos D, Dujardin K, Poirot I, et al: Comparison of desipramine and
citalopram treatments for depression in Parkinson’s disease: a double-
blind, randomized, placebo-controlled study. Mov Disord 23(6):850–
857, 2008 18311826
Diem SJ, Blackwell TL, Stone KL, et al: Use of antidepressants and rates of
hip bone loss in older women: the study of osteoporotic fractures. Arch
Intern Med 167(12):1240–1245, 2007 17592096
Diniz BS, Pinto JA Jr, Gonzaga ML, et al: To treat or not to treat? A meta-
analysis of the use of cholinesterase inhibitors in mild cognitive
impairment for delaying progression to Alzheimer’s disease. Eur Arch
Psychiatry Clin Neurosci 259(4):248–256, 2009 19224111
Dolder C, Nelson M, Stump A: Pharmacological and clinical profile of
newer antidepressants: implications for the treatment of elderly
patients. Drugs Aging 27(8): 625–640, 2010 20658791
Doody RS, Ferris SH, Salloway S, et al: Donepezil treatment of patients
with MCI: a 48-week randomized, placebo-controlled trial. Neurology
72(18):1555–1561, 2009 19176895
Doraiswamy PM, Khan ZM, Donahue RM, et al: Quality of life in geriatric
depression: a comparison of remitters, partial responders, and
nonresponders. Am J Geriatr Psychiatry 9(4):423–428, 2001 11739069
Doraiswamy PM, Krishnan KR, Oxman T, et al: Does antidepressant
therapy improve cognition in elderly depressed patients? J Gerontol A
Biol Sci Med Sci 58(12):M1137–M1144, 2003 14684712
D’Souza R, Rajji TK, Mulsant BH, et al: Use of lithium in the treatment of
bipolar disorder in late-life. Curr Psychiatry Rep 13(6): 488–492, 2011
21847537
Dunlop BW, Crits-Christoph P, Evans DL, et al: Coadministration of
modafinil and a selective serotonin reuptake inhibitor from the
initiation of treatment of major depressive disorder with fatigue and
sleepiness: a double-blind, placebo-controlled study. J Clin
Psychopharmacol 27(6):614–619, 2007 18004129
Dunner DL, Cohn JB, Walshe T 3rd, et al: Two combined, multicenter
double-blind studies of paroxetine and doxepin in geriatric patients
with major depression. J Clin Psychiatry 53 (suppl):57–60, 1992
1531827
Dunner DL, Zisook S, Billow AA, et al: A prospective safety surveillance
study for bupropion sustained-release in the treatment of depression. J
Clin Psychiatry 59(7):366–373, 1998 9714265
Ellis T, Cudkowicz ME, Sexton PM, et al: Clozapine and risperidone
treatment of psychosis in Parkinson’s disease. J Neuropsychiatry Clin
Neurosci 12(3):364–369, 2000 10956570
Emre M, Tsolaki M, Bonuccelli U, et al; 11018 Study Investigators:
Memantine for patients with Parkinson’s disease dementia or dementia
with Lewy bodies: a randomised, double-blind, placebo-controlled
trial. Lancet Neurol 9(10):969–977, 2010 20729148
Evans M, Hammond M, Wilson K, et al: Treatment of depression in the
elderly: effect of physical illness on response. Int J Geriatr Psychiatry
12(12):1189–1194, 1997 9444543
Fabian TJ, Amico JA, Kroboth PD, et al: Paroxetine-induced hyponatremia
in older adults: a 12-week prospective study. Arch Intern Med
164(3):327–332, 2004 14769630
Fava M, Thase ME, DeBattista C, et al: Modafinil augmentation of
selective serotonin reuptake inhibitor therapy in MDD partial
responders with persistent fatigue and sleepiness. Ann Clin Psychiatry
19(3):153–159, 2007 17729016
Feighner JP, Cohn JB: Double-blind comparative trials of fluoxetine and
doxepin in geriatric patients with major depressive disorder. J Clin
Psychiatry 46(3 Pt 2):20–25, 1985 3882676
Feldman PD, Hay LK, Deberdt W, et al: Retrospective cohort study of
diabetes mellitus and antipsychotic treatment in a geriatric population
in the United States. J Am Med Dir Assoc 5(1):38–46, 2004 14706127
Fenn HH, Sommer BR, Ketter TA, et al: Safety and tolerability of mood-
stabilising anticonvulsants in the elderly. Expert Opin Drug Saf
5(3):401–416, 2006 16610969
Fernandez HH, Trieschmann ME, Burke MA, et al: Quetiapine for
psychosis in Parkinson’s disease versus dementia with Lewy bodies. J
Clin Psychiatry 63(6):513–515, 2002 12088163
Fick DM, Cooper JW, Wade WE, et al: Updating the Beers criteria for
potentially inappropriate medication use in older adults: results of a US
consensus panel of experts. Arch Intern Med 163(22):2716–2724, 2003
14662625
Finkel SI, Richter EM, Clary CM, et al: Comparative efficacy of sertraline
vs. fluoxetine in patients age 70 or over with major depression. Am J
Geriatr Psychiatry 7(3):221–227, 1999 10438693
Flint AJ: Generalised anxiety disorder in elderly patients: epidemiology,
diagnosis and treatment options. Drugs Aging 22(2):101–114, 2005
15733018
Flint AJ, Rifat SL: Nonresponse to first-line pharmacotherapy may predict
relapse and recurrence of remitted geriatric depression. Depress
Anxiety 13(3):125–131, 2001 11387732
Foland LC, Altshuler LL, Sugar CA, et al: Increased volume of the
amygdala and hippocampus in bipolar patients treated with lithium.
Neuroreport 19(2):221–224, 2008 18185112
Fontaine CS, Hynan LS, Koch K, et al: A double-blind comparison of
olanzapine versus risperidone in the acute treatment of dementia-
related behavioral disturbances in extended care facilities. J Clin
Psychiatry 64(6):726–730, 2003 12823090
Forester BP, Streeter CC, Berlow YA, et al: Brain lithium levels and effects
on cognition and mood in geriatric bipolar disorder: a lithium-7
magnetic resonance spectroscopy study. Am J Geriatr Psychiatry
17(1):13–23, 2009 18626002
Forlenza OV, Almeida OP, Stoppe A Jr, et al: Antidepressant efficacy and
safety of low-dose sertraline and standard-dose imipramine for the
treatment of depression in older adults: results from a double-blind,
randomized, controlled clinical trial. Int Psychogeriatr 13(1):75–84,
2001 11352337
Freudenreich O, Menza M: Zolpidem-related delirium: a case report. J Clin
Psychiatry 61(6):449–450, 2000 10901348
Freund-Levi Y, Jedenius E, Tysen-Bäckström AC, et al: Galantamine versus
risperidone treatment of neuropsychiatric symptoms in patients with
probable dementia: an open randomized trial. Am J Geriatr Psychiatry
22(4):341–348, 2014 24035407
Friedman RA, Leon AC: Expanding the black box—depression,
antidepressants, and the risk of suicide. N Engl J Med 356(23): 2343–
2346, 2007 17485726
Furlan PM, Kallan MJ, Ten Have T, et al: Cognitive and psychomotor
effects of paroxetine and sertraline on healthy elderly volunteers. Am J
Geriatr Psychiatry 9(4): 429–438, 2001 11739070
Gareri P, Cotroneo A, Lacava R, et al: Comparison of the efficacy of new
and conventional antipsychotic drugs in the treatment of behavioral and
psychological symptoms of dementia (BPSD). Arch Gerontol Geriatr
Suppl 9(9):207–215, 2004 15207416
Garfield LD, Müller DJ, Kennedy JL, et al: Genetic variation in the
serotonin transporter and HTR1B receptor predicts reduced bone
formation during serotonin reuptake inhibitor treatment in older adults.
World J Biol Psychiatry 15(5):404–410, 2014 24074042
Gartlehner G, Hansen RA, Morgan LC, et al: Comparative benefits and
harms of second-generation antidepressants for treating major
depressive disorder: an updated meta-analysis. Ann Intern Med
155(11):772–785, 2011 22147715
Gastó C, Navarro V, Marcos T, et al: Single-blind comparison of
venlafaxine and nortriptyline in elderly major depression. J Clin
Psychopharmacol 23(1):21–26, 2003 12544371
Gauthier S, Loft H, Cummings J: Improvement in behavioural symptoms in
patients with moderate to severe Alzheimer’s disease by memantine: a
pooled data analysis. Int J Geriatr Psychiatry 23(5):537–545, 2008
18058838
Gauthier S, Cummings J, Ballard C, et al: Management of behavioral
problems in Alzheimer’s disease. Int Psychogeriatr 22(3): 346–372,
2010 20096151
Gelenberg AJ, McGahuey C, Laukes C, et al: Mirtazapine substitution in
SSRI-induced sexual dysfunction. J Clin Psychiatry 61(5):356–360,
2000 10847310
Georgotas A, McCue RE, Hapworth W, et al: Comparative efficacy and
safety of MAOIs versus TCAs in treating depression in the elderly.
Biol Psychiatry 21(12): 1155–1166, 1986 3756264
Geretsegger C, Stuppaeck CH, Mair M, et al: Multicenter double blind
study of paroxetine and amitriptyline in elderly depressed inpatients.
Psychopharmacology (Berl) 119(3):277–281, 1995 7675961
Germaná C, Kempton MJ, Sarnicola A, et al: The effects of lithium and
anticonvulsants on brain structure in bipolar disorder. Acta Psychiatr
Scand 122(6):481–487, 2010 20560901
Gex-Fabry M, Balant-Gorgia AE, Balant LP: Therapeutic drug monitoring
of olanzapine: the combined effect of age, gender, smoking, and
comedication. Ther Drug Monit 25(1):46–53, 2003 12548144
Gill SS, Rochon PA, Herrmann N, et al: Atypical antipsychotic drugs and
risk of ischaemic stroke: population based retrospective cohort study.
BMJ 330(7489):445, 2005 15668211
Gill SS, Anderson GM, Fischer HD, et al: Syncope and its consequences in
patients with dementia receiving cholinesterase inhibitors: a
population-based cohort study. Arch Intern Med 169(9):867–873, 2009
19433698
Glass J, Lanctôt KL, Herrmann N, et al: Sedative hypnotics in older people
with insomnia: meta-analysis of risks and benefits. BMJ
331(7526):1169, 2005 16284208
Glassman AH, O’Connor CM, Califf RM, et al; Sertraline Antidepressant
Heart Attack Randomized Trial (SADHEART) Group: Sertraline
treatment of major depression in patients with acute MI or unstable
angina (published erratum appears in JAMA 288:1720, 2002). JAMA
288(6):701–709, 2002 12169073
Goetz CG, Blasucci LM, Leurgans S, et al: Olanzapine and clozapine:
comparative effects on motor function in hallucinating PD patients.
Neurology 55(6):789–794, 2000 10993997
González-Rodríguez A, Molina-Andreu O, Penadés R, et al: Effectiveness
of long-acting injectable antipsychotics in delusional disorders with
nonprominent hallucinations and without hallucinations. Int Clin
Psychopharmacol 29(3):177–180, 2014 24681811
Goodnick PJ, Hernandez M: Treatment of depression in comorbid medical
illness. Expert Opin Pharmacother 1(7):1367–1384, 2000 11249471
Gorwood P, Weiller E, Lemming O, et al: Escitalopram prevents relapse in
older patients with major depressive disorder. Am J Geriatr Psychiatry
15(7):581–593, 2007 17586783
Goss AJ, Kaser M, Costafreda SG, et al: Modafinil augmentation therapy in
unipolar and bipolar depression: a systematic review and meta-analysis
of randomized controlled trials. J Clin Psychiatry 74(11):1101–1107,
2013 24330897
Greco KE, Tune LE, Brown FW, et al: A retrospective study of the safety of
intramuscular ziprasidone in agitated elderly patients. J Clin Psychiatry
66(7):928–929, 2005 16013910
Greenblatt DJ, Harmatz JS, Shapiro L, et al: Sensitivity to triazolam in the
elderly. N Engl J Med 324(24):1691–1698, 1991 2034245
Grothe DR, Scheckner B, Albano D: Treatment of pain syndromes with
venlafaxine. Pharmacotherapy 24(5):621–629, 2004 15162896
Gualtieri CT, Johnson LG: Comparative neurocognitive effects of 5
psychotropic anticonvulsants and lithium. MedGenMed 8(3):46, 2006
17406176
Guay DR: Comment on the potential utility of the new atypical
antipsychotic lurasidone in the geriatric population. Consult Pharm
26(8):579–582, 2011 21840821
Guillibert E, Pelicier Y, Archambault JC, et al: A double-blind, multicentre
study of paroxetine versus clomipramine in depressed elderly patients.
Acta Psychiatr Scand Suppl 350:132–134, 1989 2530766
Gustavsen I, Bramness JG, Skurtveit S, et al: Road traffic accident risk
related to prescriptions of the hypnotics zopiclone, zolpidem,
flunitrazepam and nitrazepam. Sleep Med 9(8):818–822, 2008
18226959
Gustavsson A, Van Der Putt R, Jönsson L, et al: Economic evaluation of
cholinesterase inhibitor therapy for dementia: comparison of
Alzheimer’s disease and Dementia with Lewy bodies. Int J Geriatr
Psychiatry 24(10):1072–1078, 2009 19639600
Hajek T, Cullis J, Novak T, et al: Hippocampal volumes in bipolar
disorders: opposing effects of illness burden and lithium treatment.
Bipolar Disord 14(3):261–270, 2012a 22548899
Hajek T, Kopecek M, Höschl C, et al: Smaller hippocampal volumes in
patients with bipolar disorder are masked by exposure to lithium: a
meta-analysis. J Psychiatry Neurosci 37(5):333–343, 2012b 22498078
Han CS, Kim YK: A double-blind trial of risperidone and haloperidol for
the treatment of delirium. Psychosomatics 45(4): 297–301, 2004
15232043
Hansen RA, Gartlehner G, Lohr KN, et al: Efficacy and safety of second-
generation antidepressants in the treatment of major depressive
disorder. Ann Intern Med 143(6):415–426, 2005 16172440
Hansen RA, Gartlehner G, Webb AP, et al: Efficacy and safety of donepezil,
galantamine, and rivastigmine for the treatment of Alzheimer’s disease:
a systematic review and meta-analysis. Clin Interv Aging 3(2):211–
225, 2008 18686744
Harvey AT, Rudolph RL, Preskorn SH: Evidence of the dual mechanisms of
action of venlafaxine. Arch Gen Psychiatry 57(5):503–509, 2000
10807491
Harvey PD, Napolitano JA, Mao L, et al: Comparative effects of
risperidone and olanzapine on cognition in elderly patients with
schizophrenia or schizoaffective disorder. Int J Geriatr Psychiatry
18(9):820–829, 2003 12949850
Health Canada Advisory: Remeron/Remeron RD (mirtazapine)—Abnormal
Heart Rhythm. March 2014. Available at:
http://healthycanadians.gc.ca/recall-alert-rappel-avis/hc-
sc/2014/38709a-eng.php. Accessed June 9, 2014.
Hemmelgarn B, Suissa S, Huang A, et al: Benzodiazepine use and the risk
of motor vehicle crash in the elderly. JAMA 278(1): 27–31, 1997
9207334
Henry G, Williamson D, Tampi RR: Efficacy and tolerability of
antidepressants in the treatment of behavioral and psychological
symptoms of dementia, a literature review of evidence. Am J
Alzheimers Dis Other Demen 26(3):169–183, 2011 21429956
Hernandez RK, Farwell W, Cantor MD, et al: Cholinesterase inhibitors and
incidence of bradycardia in patients with dementia in the Veterans
Affairs New England healthcare system. J Am Geriatr Soc
57(11):1997–2003, 2009 19793162
Herrmann N, Mamdani M, Lanctôt KL: Atypical antipsychotics and risk of
cerebrovascular accidents. Am J Psychiatry 161(6): 1113–1115, 2004
15169702
Herrmann N, Rothenburg LS, Black SE, et al: Methylphenidate for the
treatment of apathy in Alzheimer disease: prediction of response using
dextroamphetamine challenge. J Clin Psychopharmacol 28(3):296–301,
2008 18480686
Herrmann N, Cappell J, Eryavec GM, et al: Changes in nursing burden
following memantine for agitation and aggression in long-term care
residents with moderate to severe Alzheimer’s disease: an open-label
pilot study. CNS Drugs 25(5):425–433, 2011 21476613
Herrmann N, Gauthier S, Boneva N, Lemming OM; 10158 Investigators: A
randomized, double-blind, placebo-controlled trial of memantine in a
behaviorally enriched sample of patients with moderate-to-severe
Alzheimer’s disease. Int Psychogeriatr 25(6):919–927, 2013a
23472619
Herrmann N, Lanctôt KL, Hogan DB: Pharmacological recommendations
for the symptomatic treatment of dementia: the Canadian Consensus
Conference on the Diagnosis and Treatment of Dementia 2012.
Alzheimers Res Ther 8(suppl 1):S5, 2013b
Hesse LM, He P, Krishnaswamy S, et al: Pharmacogenetic determinants of
interindividual variability in bupropion hydroxylation by cytochrome
P450 2B6 in human liver microsomes. Pharmacogenetics 14(4):225–
238, 2004 15083067
Hibbard KR, Propst A, Frank DE, et al: Fatalities associated with
clozapine-related constipation and bowel obstruction: a literature
review and two case reports. Psychosomatics 50(4):416–419, 2009
19687183
Hien TT, Cumming RG, Cameron ID, et al: Atypical antipsychotic
medications and risk of falls in residents of aged care facilities. J Am
Geriatr Soc 53(8):1290–1295, 2005 16078953
Houlihan DJ, Mulsant BH, Sweet RA, et al: A naturalistic study of
trazodone in the treatment of behavioral complications of dementia.
Am J Geriatr Psychiatry 2(1): 78–85, 1994 21629010
Howanitz E, Pardo M, Smelson DA, et al: The efficacy and safety of
clozapine versus chlorpromazine in geriatric schizophrenia. J Clin
Psychiatry 60(1):41–44, 1999 10074877
Howard RJ, Juszczak E, Ballard CG, et al; CALM-AD Trial Group:
Donepezil for the treatment of agitation in Alzheimer’s disease. N Engl
J Med 357(14):1382–1392, 2007 17914039
Howard R, McShane R, Lindesay J, et al: Donepezil and memantine for
moderate-to-severe Alzheimer’s disease. N Engl J Med 366(10):893–
903, 2012 22397651
Howard WT, Warnock JK: Bupropion-induced psychosis. Am J Psychiatry
156(12):2017–2018, 1999 10588428
Høyberg OJ, Maragakis B, Mullin J, et al: A double-blind multicentre
comparison of mirtazapine and amitriptyline in elderly depressed
patients. Acta Psychiatr Scand 93(3):184–190, 1996 8739664
Hung CC, Lin CH, Lan TH, et al: The association of selective serotonin
reuptake inhibitors use and stroke in geriatric population. Am J Geriatr
Psychiatry 21(8):811–815, 2013 23567390
Hutchison LC: Mirtazapine and bone marrow suppression: a case report. J
Am Geriatr Soc 49(8):1129–1130, 2001 11555082
Jaskiw GE, Thyrum PT, Fuller MA, et al: Pharmacokinetics of quetiapine in
elderly patients with selected psychotic disorders. Clin Pharmacokinet
43(14):1025–1035, 2004 15530131
Jeste DV, Barak Y, Madhusoodanan S, et al: International multisite double-
blind trial of the atypical antipsychotics risperidone and olanzapine in
175 elderly patients with chronic schizophrenia. Am J Geriatr
Psychiatry 11(6):638–647, 2003 14609804
Jin Y, Pollock BG, Frank E, et al: Effect of age, weight, and CYP2C19
genotype on escitalopram exposure. J Clin Pharmacol 50(1):62–72,
2010 19841156
Johnson EM, Whyte E, Mulsant BH, et al: Cardiovascular changes
associated with venlafaxine in the treatment of late-life depression. Am
J Geriatr Psychiatry 14(9): 796–802, 2006 16943176
Joo JH, Lenze EJ, Mulsant BH, et al: Risk factors for falls during treatment
of late-life depression. J Clin Psychiatry 63(10):936–941, 2002
12416604
Jorge RE, Acion L, Moser D, et al: Escitalopram and enhancement of
cognitive recovery following stroke. Arch Gen Psychiatry 67(2):187–
196, 2010 20124118
Juurlink DN, Mamdani MM, Kopp A, et al: The risk of suicide with
selective serotonin reuptake inhibitors in the elderly. Am J Psychiatry
163(5):813–821, 2006 16648321
Kando JC, Tohen M, Castillo J, et al: The use of valproate in an elderly
population with affective symptoms. J Clin Psychiatry 57(6):238–240,
1996 8666559
Kane J, Canas F, Kramer M, et al: Treatment of schizophrenia with
paliperidone extended-release tablets: a 6-week placebo-controlled
trial. Schizophr Res 90(1–3): 147–161, 2007 17092691
Karaiskos D, Pappa D, Tzavellas E, et al: Pregabalin augmentation of
antidepressants in older patients with comorbid depression and
generalized anxiety disorder-an open-label study. Int J Geriatr
Psychiatry 28(1):100–105, 2013 22431439
Karp JF, Weiner D, Seligman K, et al: Body pain and treatment response in
late-life depression. Am J Geriatr Psychiatry 13(3):188–194, 2005
15728749
Karp JF, Whyte EM, Lenze EJ, et al: Rescue pharmacotherapy with
duloxetine for selective serotonin reuptake inhibitor nonresponders in
late-life depression: outcome and tolerability. J Clin Psychiatry
69(3):457–463, 2008 18251622
Karp JF, Weiner DK, Dew MA, et al: Duloxetine and care management
treatment of older adults with comorbid major depressive disorder and
chronic low back pain: results of an open-label pilot study. Int J Geriatr
Psychiatry 25(6):633–642, 2010 19750557
Kasckow JW, Mohamed S, Thallasinos A, et al: Citalopram augmentation
of antipsychotic treatment in older schizophrenia patients. Int J Geriatr
Psychiatry 16(12): 1163–1167, 2001 11748776
Kasper S, de Swart H, Friis Andersen H: Escitalopram in the treatment of
depressed elderly patients. Am J Geriatr Psychiatry 13(10):884–891,
2005 16223967
Katila H, Mezhebovsky I, Mulroy A, et al: Randomized, double-blind study
of the efficacy and tolerability of extended release quetiapine fumarate
(quetiapine XR) monotherapy in elderly patients with major depressive
disorder. Am J Geriatr Psychiatry 21(8):769–784, 2013 23567397
Katona CLE, Hunter BN, Bray J: A double-blind comparison of the
efficacy and safely of paroxetine and imipramine in the treatment of
depression with dementia. Int J Geriatr Psychiatry 13(2):100–108, 1998
9526179
Katz IR, Jeste DV, Mintzer JE, et al; Risperidone Study Group: Comparison
of risperidone and placebo for psychosis and behavioral disturbances
associated with dementia: a randomized, double-blind trial. J Clin
Psychiatry 60(2):107–115, 1999 10084637
Katz IR, Reynolds CF 3rd, Alexopoulos GS, et al: Venlafaxine ER as a
treatment for generalized anxiety disorder in older adults: pooled
analysis of five randomized placebo-controlled clinical trials. J Am
Geriatr Soc 50(1):18–25, 2002 12028242
Kavirajan H: Memantine: a comprehensive review of safety and efficacy.
Expert Opin Drug Saf 8(1):89–109, 2009 19236221
Kavirajan H, Schneider LS: Efficacy and adverse effects of cholinesterase
inhibitors and memantine in vascular dementia: a meta-analysis of
randomised controlled trials. Lancet Neurol 6(9):782–792, 2007
17689146
Kennedy JS, Jeste D, Kaiser CJ, et al: Olanzapine vs haloperidol in geriatric
schizophrenia: analysis of data from a double-blind controlled trial. Int
J Geriatr Psychiatry 18(11):1013–1020, 2003 14618553
Kennedy J, Deberdt W, Siegal A, et al: Olanzapine does not enhance
cognition in non-agitated and non-psychotic patients with mild to
moderate Alzheimer’s dementia. Int J Geriatr Psychiatry 20(11):1020–
1027, 2005 16250069
Kiev A, Masco HL, Wenger TL, et al: The cardiovascular effects of
bupropion and nortriptyline in depressed outpatients. Ann Clin
Psychiatry 6(2):107–115, 1994 7804386
Kim KY, Bader GM, Kotlyar V, et al: Treatment of delirium in older adults
with quetiapine. J Geriatr Psychiatry Neurol 16(1):29–31, 2003
12641370
Kinon BJ, Stauffer VL, McGuire HC, et al: The effects of antipsychotic
drug treatment on prolactin concentrations in elderly patients. J Am
Med Dir Assoc 4(4):189–194, 2003 12837139
Kirby D, Harrigan S, Ames D: Hyponatraemia in elderly psychiatric
patients treated with selective serotonin reuptake inhibitors and
venlafaxine: a retrospective controlled study in an inpatient unit. Int J
Geriatr Psychiatry 17(3):231–237, 2002 11921151
Kleijer BC, Heerdink ER, Egberts TC, et al: Antipsychotic drug use and the
risk of venous thromboembolism in elderly patients. J Clin
Psychopharmacol 30(5):526–530, 2010 20814323
Kohen I, Preval H, Southard R, et al: Naturalistic study of intramuscular
ziprasidone versus conventional agents in agitated elderly patients:
retrospective findings from a psychiatric emergency service. Am J
Geriatr Pharmacother 3(4):240–245, 2005 16503319
Kohen I, Gordon ML, Manu P: Serotonin syndrome in elderly patients
treated for psychotic depression with atypical antipsychotics and
antidepressants: two case reports. CNS Spectr 12(8):596–598, 2007
17667887
Kok RM, Nolen WA, Heeren TJ: Venlafaxine versus nortriptyline in the
treatment of elderly depressed inpatients: a randomised, double-blind,
controlled trial. Int J Geriatr Psychiatry 22(12):1247–1254, 2007
17562523
Koller EA, Cross JT, Doraiswamy PM, et al: Pancreatitis associated with
atypical antipsychotics: from the Food and Drug Administration’s
MedWatch surveillance system and published reports.
Pharmacotherapy 23(9):1123–1130, 2003 14524644
Kornstein SG, Clayton AH, Soares CN, et al: Analysis by age and sex of
efficacy data from placebo-controlled trials of desvenlafaxine in
outpatients with major depressive disorder. J Clin Psychopharmacol
30(3):294–299, 2010a 20473066
Kornstein SG, Jiang Q, Reddy S, et al: Short-term efficacy and safety of
desvenlafaxine in a randomized, placebo-controlled study of
perimenopausal and postmenopausal women with major depressive
disorder. J Clin Psychiatry 71(8):1088–1096, 2010b 20797382
Kornstein SG, Clayton A, Bao W, Guico-Pabia CJ: Post hoc analysis of the
efficacy and safety of desvenlafaxine 50 mg/day in a randomized,
placebo-controlled study of perimenopausal and postmenopausal
women with major depressive disorder. Menopause 21(8):799–806,
2014 24448103
Kotlyar M, Brauer LH, Tracy TS, et al: Inhibition of CYP2D6 activity by
bupropion. J Clin Psychopharmacol 25(3):226–229, 2005 15876900
Kripke DF, Langer RD, Kline LE: Hypnotics’ association with mortality or
cancer: a matched cohort study. BMJ Open 2(1): e000850, 2012
22371848
Kuehn BM: FDA warns antipsychotic drugs may be risky for elderly.
JAMA 293(20): 2462, 2005 15914734
Kurlan R, Cummings J, Raman R, Thal L; Alzheimer’s Disease
Cooperative Study Group: Quetiapine for agitation or psychosis in
patients with dementia and parkinsonism. Neurology 68(17):1356–
1363, 2007 17452579
Kyle CJ, Petersen HE, Overø KF: Comparison of the tolerability and
efficacy of citalopram and amitriptyline in elderly depressed patients
treated in general practice. Depress Anxiety 8(4):147–153, 1998
9871816
Lam RW, Lönn SL, Despiégel N: Escitalopram versus serotonin
noradrenaline reuptake inhibitors as second step treatment for patients
with major depressive disorder: a pooled analysis. Int Clin
Psychopharmacol 25(4):199–203, 2010 20357664
Landi F, Onder G, Cesari M, et al; Silver Network Home Care Study
Group: Psychotropic medications and risk for falls among community-
dwelling frail older people: an observational study. J Gerontol A Biol
Sci Med Sci 60(5):622–626, 2005 15972615
Langballe EM, Engdahl B, Nordeng H, et al: Short- and long-term mortality
risk associated with the use of antipsychotics among 26,940 dementia
outpatients: a population-based study. Am J Geriatr Psychiatry
22(4):321–331, 2014 24016844
Laroche ML, Charmes JP, Nouaille Y, et al: Is inappropriate medication use
a major cause of adverse drug reactions in the elderly? Br J Clin
Pharmacol 63(2):177–186, 2007 17166186
Larsen KA, Kelly SE, Stern TA, et al: Administration of olanzapine to
prevent postoperative delirium in elderly joint-replacement patients: a
randomized, controlled trial. Psychosomatics 51(5):409–418, 2010
20833940
Lasser RA, Bossie CA, Zhu Y, et al: Efficacy and safety of long-acting
risperidone in elderly patients with schizophrenia and schizoaffective
disorder. Int J Geriatr Psychiatry 19(9):898–905, 2004 15352149
Lavretsky H, Park S, Siddarth P, et al: Methylphenidate-enhanced
antidepressant response to citalopram in the elderly: a double-blind,
placebo-controlled pilot trial. Am J Geriatr Psychiatry 14(2):181–185,
2006 16473984
Lavretsky H, Siddarth P, Irwin MR: Improving depression and enhancing
resilience in family dementia caregivers: a pilot randomized placebo-
controlled trial of escitalopram. Am J Geriatr Psychiatry 18(2):154–
162, 2010 20104071
Lee PE, Gill SS, Freedman M, et al: Atypical antipsychotic drugs in the
treatment of behavioural and psychological symptoms of dementia:
systematic review. BMJ 329(7457):75, 2004 15194601
Lenze EJ, Mulsant BH, Shear MK, et al: Anxiety symptoms in elderly
patients with depression: what is the best approach to treatment? Drugs
Aging 19(10):753–760, 2002 12390052
Lenze EJ, Mulsant BH, Shear MK, et al: Efficacy and tolerability of
citalopram in the treatment of late-life anxiety disorders: results from
an 8-week randomized, placebo-controlled trial. Am J Psychiatry
162(1):146–150, 2005 15625213
Lenze EJ, Rollman BL, Shear MK, et al: Escitalopram for older adults with
generalized anxiety disorder: a randomized controlled trial. JAMA
301(3):295–303, 2009 19155456
Leopold NA: Risperidone treatment of drug-related psychosis in patients
with parkinsonism. Mov Disord 15(2):301–304, 2000 10752580
Lessard E, Yessine MA, Hamelin BA, et al: Influence of CYP2D6 activity
on the disposition and cardiovascular toxicity of the antidepressant
agent venlafaxine in humans. Pharmacogenetics 9(4):435–443, 1999
10780263
Leufkens TR, Lund JS, Vermeeren A: Highway driving performance and
cognitive functioning the morning after bedtime and middle-of-the-
night use of gaboxadol, zopiclone and zolpidem. J Sleep Res
18(4):387–396, 2009 19552733
Liebowitz MR, Tourian KA: Efficacy, safety, and tolerability of
desvenlafaxine 50 mg/d for the treatment of major depressive disorder:
a systematic review of clinical trials. Prim Care Companion J Clin
Psychiatry 12(3): 2010 20944767
Lindenmayer JP, Nasrallah H, Pucci M, et al: A systematic review of
psychostimulant treatment of negative symptoms of schizophrenia:
challenges and therapeutic opportunities. Schizophr Res 147(2–3):
241–252, 2013 23619055
Liperoti R, Pedone C, Lapane KL, et al: Venous thromboembolism among
elderly patients treated with atypical and conventional antipsychotic
agents. Arch Intern Med 165(22):2677–2682, 2005 16344428
Liperoti R, Onder G, Lapane KL, et al: Conventional or atypical
antipsychotics and the risk of femur fracture among elderly patients:
results of a case-control study. J Clin Psychiatry 68(6):929–934, 2007
17592919
Lipscombe LL, Lévesque L, Gruneir A, et al: Antipsychotic drugs and
hyperglycemia in older patients with diabetes. Arch Intern Med
169(14):1282–1289, 2009 19636029
Liu B, Anderson G, Mittmann N, et al: Use of selective serotonin-reuptake
inhibitors or tricyclic antidepressants and risk of hip fractures in elderly
people. Lancet 351(9112):1303–1307, 1998 9643791
Lockhart IA, Orme ME, Mitchell SA: The efficacy of licensed-indication
use of donepezil and memantine monotherapies for treating
behavioural and psychological symptoms of dementia in patients with
Alzheimer’s disease: systematic review and meta-analysis. Dement
Geriatr Cogn Dis Extra 1(1):212–227, 2011 22163246
Looper KJ: Potential medical and surgical complications of serotonergic
antidepressant medications. Psychosomatics 48(1):1–9, 2007 17209143
Löppönen P, Tetri S, Juvela S, et al: Association between warfarin
combined with serotonin-modulating antidepressants and increased
case fatality in primary intracerebral hemorrhage: a population-based
study. J Neurosurg 120(6):1358–1363, 2014 24506245
Lotrich FE, Rabinovitz M, Gironda P, et al: Depression following pegylated
interferon-alpha: characteristics and vulnerability. J Psychosom Res
63(2):131–135, 2007 17662748
Lu CJ, Tune LE: Chronic exposure to anticholinergic medications adversely
affects the course of Alzheimer disease. Am J Geriatr Psychiatry
11(4):458–461, 2003 12837675
Lyketsos CG, DelCampo L, Steinberg M, et al: Treating depression in
Alzheimer disease: efficacy and safety of sertraline therapy, and the
benefits of depression reduction: the DIADS. Arch Gen Psychiatry
60(7):737–746, 2003 12860778
Macritchie KA, Lloyd AJ, Bastin ME, et al: White matter microstructural
abnormalities in euthymic bipolar disorder. Br J Psychiatry 196(1):52–
58, 2010 20044661
Madhusoodanan S, Zaveri D: Paliperidone use in the elderly. Curr Drug Saf
5(2):149–152, 2010 20406162
Madhusoodanan S, Suresh P, Brenner R, et al: Experience with the atypical
antipsychotics—risperidone and olanzapine in the elderly. Ann Clin
Psychiatry 11(3):113–118, 1999 10482120
Madhusoodanan S, Brenner R, Alcantra A: Clinical experience with
quetiapine in elderly patients with psychotic disorders. J Geriatr
Psychiatry Neurol 13(1):28–32, 2000 10753004
Mahapatra SN, Hackett D: A randomised, double-blind, parallel-group
comparison of venlafaxine and dothiepin in geriatric patients with
major depression. Int J Clin Pract 51(4):209–213, 1997 9287259
Maher AR, Theodore G: Summary of the comparative effectiveness review
on off-label use of atypical antipsychotics. J Manag Care Pharm 18(5)
(suppl B):S1–S20, 2012 22784311
Maher AR, Maglione M, Bagley S, et al: Efficacy and comparative
effectiveness of atypical antipsychotic medications for off-label uses in
adults: a systematic review and meta-analysis. JAMA 306(12): 1359–
1369, 2011 21954480
Mahmood I, Sahajwalla C: Clinical pharmacokinetics and
pharmacodynamics of buspirone, an anxiolytic drug. Clin
Pharmacokinet 36(4):277–287, 1999 10320950
Mamo DC, Sweet RA, Mulsant BH, et al: Effect of nortriptyline and
paroxetine on extrapyramidal signs and symptoms: A prospective
double-blind study in depressed elderly patients. Am J Geriatr
Psychiatry 8(3):226–231, 2000 10910421
Mamo D, Graff A, Mizrahi R, et al: Differential effects of aripiprazole on
D(2), 5-HT(2), and 5-HT(1A) receptor occupancy in patients with
schizophrenia: a triple tracer PET study. Am J Psychiatry 164(9):1411–
1417, 2007 17728427
Manning KJ, Alexopoulos GS, Banerjee S, et al: Executive functioning
complaints and escitalopram treatment response in late-life depression.
Am J Geriatr Psychiatry Nov 27, 2013 [Epub ahead of print] 24388222
Mariappan P, Ballantyne Z, N’Dow JM, et al: Serotonin and noradrenaline
reuptake inhibitors (SNRI) for stress urinary incontinence in adults.
Cochrane Database Syst Rev (3):CD004742, 2005 16034945
Marsh L, Lyketsos C, Reich SG: Olanzapine for the treatment of psychosis
in patients with Parkinson’s disease and dementia. Psychosomatics
42(6):477–481, 2001 11815682
Martin H, Slyk MP, Deymann S, et al: Safety profile assessment of
risperidone and olanzapine in long-term care patients with dementia. J
Am Med Dir Assoc 4(4): 183–188, 2003 12837138
Matsunaga S, Kishi T, Iwata N: Memantine for Lewy body disorders:
systematic review and meta-analysis. Am J Geriatr Psychiatry Dec 4,
2013 [Epub ahead of print] 24406251
Mayeux R: Clinical practice. Early Alzheimer’s disease. N Engl J Med
362(23): 2194–2201, 2010 20558370
Mazeh D, Shahal B, Aviv A, et al: A randomized, single-blind, comparison
of venlafaxine with paroxetine in elderly patients suffering from
resistant depression. Int Clin Psychopharmacol 22(6):371–375, 2007
17917556
McCue RE, Joseph M: Venlafaxine- and trazodone-induced serotonin
syndrome. Am J Psychiatry 158(12):2088–2089, 2001 11729039
Meagher DJ, McLoughlin L, Leonard M, et al: What do we really know
about the treatment of delirium with antipsychotics? Ten key issues for
delirium pharmacotherapy. Am J Geriatr Psychiatry 21(12): 1223–
1238, 2013 23567421
Messenheimer JA: Rash in adult and pediatric patients treated with
lamotrigine. Can J Neurol Sci 25(4):S14–S18, 1998 9827240
Meyers BS, Flint AJ, Rothschild AJ, et al: A double-blind randomized
controlled trial of olanzapine plus sertraline vs olanzapine plus placebo
for psychotic depression: the study of pharmacotherapy of psychotic
depression (STOP-PD). Arch Gen Psychiatry 66:838–847, 2009
19652123
Micca JL, Hoffmann VP, Lipkovich I, et al: Retrospective analysis of
diabetes risk in elderly patients with dementia in olanzapine clinical
trials. Am J Geriatr Psychiatry 14(1):62–70, 2006 16407583
Mintzer JE, Tune LE, Breder CD, et al: Aripiprazole for the treatment of
psychoses in institutionalized patients with Alzheimer dementia: a
multicenter, randomized, double-blind, placebo-controlled assessment
of three fixed doses. Am J Geriatr Psychiatry 15(11):918–931, 2007
17974864
Mittal D, Jimerson NA, Neely EP, et al: Risperidone in the treatment of
delirium: results from a prospective open-label trial. J Clin Psychiatry
65(5):662–667, 2004 15163252
Modi A, Weiner M, Craig BA, et al: Concomitant use of anticholinergics
with acetylcholinesterase inhibitors in Medicaid recipients with
dementia and residing in nursing homes. J Am Geriatr Soc 57(7):
1238–1244, 2009 19467148
Molho ES, Factor SA: Worsening of motor features of parkinsonism with
olanzapine. Mov Disord 14(6):1014–1016, 1999 10584679
Montejo AL, Llorca G, Izquierdo JA, Rico-Villademoros F; Spanish
Working Group for the Study of Psychotropic-Related Sexual
Dysfunction: Incidence of sexual dysfunction associated with
antidepressant agents: a prospective multicenter study of 1022
outpatients. J Clin Psychiatry 62 (suppl 3):10–21, 2001 11229449
Montgomery S, Chatamra K, Pauer L, et al: Efficacy and safety of
pregabalin in elderly people with generalised anxiety disorder. Br J
Psychiatry 193(5):389–394, 2008 18978320
Montgomery SA, Fava M, Padmanabhan SK, et al: Discontinuation
symptoms and taper/poststudy-emergent adverse events with
desvenlafaxine treatment for major depressive disorder. Int Clin
Psychopharmacol 24(6):296–305, 2009 19779354
Montgomery SA, Gommoll CP, Chen C, et al: Efficacy of levomilnacipran
extended-release in major depressive disorder: pooled analysis of 5
double-blind, placebo-controlled trials. CNS Spectr. 2014 Jun 5:1–9.
[Epub ahead of print] 24902007
Mortensen JK, Larsson H, Johnsen SP, Andersen G: Post stroke use of
selective serotonin reuptake inhibitors and clinical outcome among
patients with ischemic stroke: a nationwide propensity score-matched
follow-up study. Stroke 44(2): 420–426, 2013 23306326
Mukai Y, Tampi RR: Treatment of depression in the elderly: a review of the
recent literature on the efficacy of single- versus dual-action
antidepressants. Clin Ther 31(5):945–961, 2009 19539096
Müller-Oerlinghausen B, Lewitzka U: Lithium reduces pathological
aggression and suicidality: a mini-review. Neuropsychobiology
62(1):43–49, 2010 20453534
Mulsant BH: Challenges of the treatment of neuropsychiatric symptoms
associated with dementia. Am J Geriatr Psychiatry 22(4):317–320,
2014 24635993
Mulsant BH, Alexopoulos GS, Reynolds CF 3rd, et al; PROSPECT Study
Group: Pharmacological treatment of depression in older primary care
patients: the PROSPECT algorithm. Int J Geriatr Psychiatry 16(6):585–
592, 2001a 11424167
Mulsant BH, Pollock BG, Nebes R, et al: A twelve-week, double-blind,
randomized comparison of nortriptyline and paroxetine in older
depressed inpatients and outpatients. Am J Geriatr Psychiatry
9(4):406–414, 2001b 11739067
Mulsant BH, Pollock BG, Kirshner M, et al: Serum anticholinergic activity
in a community-based sample of older adults: relationship with
cognitive performance. Arch Gen Psychiatry 60(2):198–203, 2003
12578438
Mulsant BH, Gharabawi GM, Bossie CA, et al: Correlates of
anticholinergic activity in patients with dementia and psychosis treated
with risperidone or olanzapine. J Clin Psychiatry 65(12):1708–1714,
2004 15641877
Mulsant BH, Blumberger DM, Ismail Z, et al: A systematic approach to the
pharmacotherapy of geriatric major depression. Clin Geriatr Med
30(3):517–534, 2014
Murray V, von Arbin M, Bartfai A, et al: Double-blind comparison of
sertraline and placebo in stroke patients with minor depression and less
severe major depression. J Clin Psychiatry 66(6):708–716, 2005
15960563
Nagaraja D, Jayashree S: Randomized study of the dopamine receptor
agonist piribedil in the treatment of mild cognitive impairment. Am J
Psychiatry 158(9):1517–1519, 2001 11532743
Nandagopal JJ, DelBello MP: Selegiline transdermal system: a novel
treatment option for major depressive disorder. Expert Opin
Pharmacother 10(10):1665–1673, 2009 19527191
National Collaborating Centre for Mental Health: Management of
Depression in Primary and Secondary Care (Clinical Guideline 23).
London, National Institute for Clinical Excellence, 2004
Navarro V, Gastó C, Torres X, et al: Citalopram versus nortriptyline in late-
life depression: a 12-week randomized single-blind study. Acta
Psychiatr Scand 103(6): 435–440, 2001 11401657
Nebes RD, Pollock BG, Meltzer CC, et al: Serum anticholinergic activity,
white matter hyperintensities, and cognitive performance. Neurology
65:1487–1489, 2005 16275844
Nelson JC, Wohlreich MM, Mallinckrodt CH, et al: Duloxetine for the
treatment of major depressive disorder in older patients. Am J Geriatr
Psychiatry 13(3):227–235, 2005 15728754
Nelson JC, Delucchi K, Schneider L: Suicidal thinking and behavior during
treatment with sertraline in late-life depression. Am J Geriatr
Psychiatry 15(7):573–580, 2007 17586782
Nelson JC, Delucchi K, Schneider LS: Efficacy of second generation
antidepressants in late-life depression: a meta-analysis of the evidence.
Am J Geriatr Psychiatry 16(7):558–567, 2008 18591576
Newhouse PA, Krishnan KR, Doraiswamy PM, et al: A double-blind
comparison of sertraline and fluoxetine in depressed elderly
outpatients. J Clin Psychiatry 61(8):559–568, 2000 10982198
Nicholas LM, Ford AL, Esposito SM, et al: The effects of mirtazapine on
plasma lipid profiles in healthy subjects. J Clin Psychiatry 64(8):883–
889, 2003 12927002
Nieuwstraten CE, Dolovich LR: Bupropion versus selective serotonin-
reuptake inhibitors for treatment of depression. Ann Pharmacother
35(12):1608–1613, 2001 11793630
Nikaido AM, Ellinwood EH Jr, Heatherly DG, et al: Age-related increase in
CNS sensitivity to benzodiazepines as assessed by task difficulty.
Psychopharmacology (Berl) 100(1):90–97, 1990 1967500
Noaghiul S, Narayan M, Nelson JC: Divalproex treatment of mania in
elderly patients. Am J Geriatr Psychiatry 6(3):257–262, 1998 9659958
Nyth AL, Gottfries CG: The clinical efficacy of citalopram in treatment of
emotional disturbances in dementia disorders. A Nordic multicentre
study. Br J Psychiatry 157:894–901, 1990 1705151
Nyth AL, Gottfries CG, Lyby K, et al: A controlled multicenter clinical
study of citalopram and placebo in elderly depressed patients with and
without concomitant dementia. Acta Psychiatr Scand 86(2): 138–145,
1992 1529737
Oakes TM, Katona C, Liu P, et al: Safety and tolerability of duloxetine in
elderly patients with major depressive disorder: a pooled analysis of
two placebo-controlled studies. Int Clin Psychopharmacol 28(1):1–11,
2013 23138680
O’Connor DW, Sierakowski C, Chin LF, et al: The safety and tolerability of
clozapine in aged patients: a retrospective clinical file review. World J
Biol Psychiatry 11(6):788–791, 2010 20586532
Oganesian A, Shilling AD, Young-Sciame R, et al: Desvenlafaxine and
venlafaxine exert minimal in vitro inhibition of human cytochrome
P450 and P-glycoprotein activities. Psychopharmacol Bull 42(2):47–
63, 2009 19629022
Olafsson K, Jørgensen S, Jensen HV, et al: Fluvoxamine in the treatment of
demented elderly patients: a double-blind, placebo-controlled study.
Acta Psychiatr Scand 85(6):453–456, 1992 1642129
Oslin DW, Streim JE, Katz IR, et al: Heuristic comparison of sertraline with
nortriptyline for the treatment of depression in frail elderly patients.
Am J Geriatr Psychiatry 8(2):141–149, 2000 10804075
Oslin DW, Ten Have TR, Streim JE, et al: Probing the safety of medications
in the frail elderly: evidence from a randomized clinical trial of
sertraline and venlafaxine in depressed nursing home residents. J Clin
Psychiatry 64(8):875–882, 2003 12927001
Pact V, Giduz T: Mirtazapine treats resting tremor, essential tremor, and
levodopa-induced dyskinesias. Neurology 53(5):1154, 1999 10496290
Pae CU, Lee SJ, Lee CU, et al: A pilot trial of quetiapine for the treatment
of patients with delirium. Hum Psychopharmacol 19(2):125–127, 2004
14994323
Papakostas GI, Thase ME, Fava M, et al: Are antidepressant drugs that
combine serotonergic and noradrenergic mechanisms of action more
effective than the selective serotonin reuptake inhibitors in treating
major depressive disorder? A meta-analysis of studies of newer agents.
Biol Psychiatry 62(11):1217–1227, 2007 17588546
Parellada E, Baeza I, de Pablo J, et al: Risperidone in the treatment of
patients with delirium. J Clin Psychiatry 65(3):348–353, 2004
15096074
Park-Wyllie LY, Mamdani MM, Li P, et al: Cholinesterase inhibitors and
hospitalization for bradycardia: a population-based study. PLoS Med
6(9):e1000157, 2009 19787032
Parkinson Study Group: Low-dose clozapine for the treatment of drug-
induced psychosis in Parkinson’s disease. N Engl J Med 340(10):757–
763, 1999 10072410
Pedersen L, Klysner R: Antagonism of selective serotonin reuptake
inhibitor–induced nausea by mirtazapine. Int Clin Psychopharmacol
12(1):59–60, 1997 9179636
Percudani M, Barbui C, Fortino I, et al: Second-generation antipsychotics
and risk of cerebrovascular accidents in the elderly. J Clin
Psychopharmacol 25(5):468–470, 2005 16160623
Perry NK: Venlafaxine-induced serotonin syndrome with relapse following
amitriptyline. Postgrad Med J 76(894):254–256, 2000 10727586
Petracca GM, Chemerinski E, Starkstein SE: A double-blind, placebo-
controlled study of fluoxetine in depressed patients with Alzheimer’s
disease. Int Psychogeriatr 13(2):233–240, 2001 11495397
Phanjoo AL, Wonnacott S, Hodgson A: Double-blind comparative
multicentre study of fluvoxamine and mianserin in the treatment of
major depressive episode in elderly people. Acta Psychiatr Scand
83(6):476–479, 1991 1909083
Physicians’ Desk Reference: Full Prescribing Information—Brintellix
(vortioxetine). 2013. Available at: http://www.pdr.net/full-prescribing-
information/brintellix?druglabelid=3348. Accessed June 9, 2014
Pilotto A, Franceschi M, D’Onofrio G, et al: Effect of a CYP2D6
polymorphism on the efficacy of donepezil in patients with Alzheimer
disease. Neurology 73(10):761–767, 2009 19738170
Pinquart M, Duberstein PR, Lyness JM: Treatments for later-life depressive
conditions: a meta-analytic comparison of pharmacotherapy and
psychotherapy. Am J Psychiatry 163(9):1493–1501, 2006 16946172
Poewe W: Treatment of dementia with Lewy bodies and Parkinson’s
disease dementia. Mov Disord 20 (suppl 12):S77–S82, 2005 16092095
Pollock BG, Mulsant BH, Sweet R, et al: An open pilot study of citalopram
for behavioral disturbances of dementia. Plasma levels and real-time
observations. Am J Geriatr Psychiatry 5(1):70–78, 1997 9169247
Pollock BG, Laghrissi-Thode F, Wagner WR: Evaluation of platelet
activation in depressed patients with ischemic heart disease after
paroxetine or nortriptyline treatment. J Clin Psychopharmacol 20(2):
137–140, 2000 10770450
Pollock BG, Mulsant BH, Rosen J, et al: Comparison of citalopram,
perphenazine, and placebo for the acute treatment of psychosis and
behavioral disturbances in hospitalized, demented patients. Am J
Psychiatry 159(3):460–465, 2002 11870012
Pollock BG, Mulsant BH, Rosen J, et al: A double-blind comparison of
citalopram and risperidone for the treatment of behavioral and
psychotic symptoms associated with dementia. Am J Geriatr Psychiatry
15(11):942–952, 2007 17846102
Pollock B, Forsyth C, Bies R: The critical role of clinical pharmacology in
geriatric psychopharmacology. Clin Pharmacol Ther 85(1):89–93, 2009
19037202
Pomara N, Deptula D, Medel M, et al: Effects of diazepam on recall
memory: relationship to aging, dose, and duration of treatment.
Psychopharmacol Bull 25(1):144–148, 1989 2772114
Porsteinsson AP, Drye LT, Pollock BG, et al; CitAD Research Group: Effect
of citalopram on agitation in Alzheimer disease: the CitAD randomized
clinical trial. JAMA 311(7):682–691, 2014 24549548
Post RM, Frye MA, Denicoff KD, et al: Beyond lithium in the treatment of
bipolar illness. Neuropsychopharmacology 19(3):206–219, 1998
9653709
Prakanrattana U, Prapaitrakool S: Efficacy of risperidone for prevention of
postoperative delirium in cardiac surgery. Anaesth Intensive Care
35(5):714–719, 2007 17933157
Rabey JM, Prokhorov T, Miniovitz A, et al: Effect of quetiapine in
psychotic Parkinson’s disease patients: a double-blind labeled study of
3 months’ duration. Mov Disord 22(3):313–318, 2007 17034006
Rabins PV, Lyketsos CG: Antipsychotic drugs in dementia: what should be
made of the risks? JAMA 294(15):1963–1965, 2005 16234504
Rado J, Janicak PG: Pharmacological and clinical profile of recently
approved second-generation antipsychotics: implications for treatment
of schizophrenia in older patients. Drugs Aging 29(10):783–791, 2012
23018584
Rahman MK, Akhtar MJ, Savla NC, et al: A double-blind, randomised
comparison of fluvoxamine with dothiepin in the treatment of
depression in elderly patients. Br J Clin Pract 45(4):255–258, 1991
1810358
Rais AR, Williams K, Rais T, et al: Use of intramuscular ziprasidone for the
control of acute psychosis or agitation in an inpatient geriatric
population: an open-label study. Psychiatry (Edgmont) 7(1):17–24,
2010 20386633
Raji MA, Brady SR: Mirtazapine for treatment of depression and
comorbidities in Alzheimer disease. Ann Pharmacother 35(9):1024–
1027, 2001 11573849
Rajji TK, Mulsant BH, Lotrich FE, et al: Use of antidepressants in late-life
depression. Drugs Aging 25(10):841–853, 2008 18808208
Rajji TK, Uchida H, Ismail Z, et al: Clozapine and global cognition in
schizophrenia. J Clin Psychopharmacol 30(4):431–436, 2010 20631560
Rampello L, Chiechio S, Nicoletti G, et al: Prediction of the response to
citalopram and reboxetine in post-stroke depressed patients.
Psychopharmacology (Berl) 173(1–2):73–78, 2004 14685645
Rapaport MH, Schneider LS, Dunner DL, et al: Efficacy of controlled-
release paroxetine in the treatment of late-life depression. J Clin
Psychiatry 64(9):1065–1074, 2003 14628982
Raskin J, Wiltse CG, Siegal A, et al: Efficacy of duloxetine on cognition,
depression, and pain in elderly patients with major depressive disorder:
an 8-week, double-blind, placebo-controlled trial. Am J Psychiatry
164(6):900–909, 2007 17541049
Rasmussen A, Lunde M, Poulsen DL, et al: A double-blind, placebo-
controlled study of sertraline in the prevention of depression in stroke
patients. Psychosomatics 44(3):216–221, 2003 12724503
Ray WA, Thapa PB, Gideon P: Benzodiazepines and the risk of falls in
nursing home residents. J Am Geriatr Soc 48(6):682–685, 2000
10855607
Ray WA, Chung CP, Murray KT, et al: Atypical antipsychotic drugs and the
risk of sudden cardiac death. N Engl J Med 360(3): 225–235, 2009
19144938
Reifler BV, Teri L, Raskind M, et al: Double-blind trial of imipramine in
Alzheimer’s disease patients with and without depression. Am J
Psychiatry 146(1):45–49, 1989 2643356
Reisberg B, Doody R, Stöffler A, et al; Memantine Study Group:
Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med
348(14):1333–1341, 2003 12672860
Rej S, Beaulieu S, Segal M, et al: Lithium dosing and serum concentrations
across the age spectrum: from early adulthood to the tenth decade of
life. Drugs Aging 31(12):911–916, 2014a 25331906
Rej S, Shulman K, Herrmann N, et al: Prevalence and correlates of renal
disease in older lithium users: a population-based study. Am J Geriatr
Psychiatry 22(11): 1075–1082, 2014b 24566239
Rektorová I, Rektor I, Bares M, et al: Pramipexole and pergolide in the
treatment of depression in Parkinson’s disease: a national multicentre
prospective randomized study. Eur J Neurol 10(4):399–406, 2003
12823492
Reynolds CF3rd, Dew MA, Pollock BG, et al: Maintenance treatment of
major depression in old age. N Engl J Med 354(11): 1130–1138, 2006
16540613
Reynolds CF 3rd, Butters MA, Lopez O, et al: Maintenance treatment of
depression in old age: a randomized, double-blind, placebo-controlled
evaluation of the efficacy and safety of donepezil combined with
antidepressant pharmacotherapy. Arch Gen Psychiatry 68(1):51–60,
2011 21199965
Reznik I, Rosen Y, Rosen B: An acute ischaemic event associated with the
use of venlafaxine: a case report and proposed pathophysiological
mechanisms. J Psychopharmacol 13(2):193–195, 1999 10475726
Richards JB, Papaioannou A, Adachi JD, et al; Canadian Multicentre
Osteoporosis Study Research Group: Effect of selective serotonin
reuptake inhibitors on the risk of fracture. Arch Intern Med
167(2):188–194, 2007 17242321
Ridout F, Meadows R, Johnsen S, et al: A placebo controlled investigation
into the effects of paroxetine and mirtazapine on measures related to
car driving performance. Hum Psychopharmacol 18(4): 261–269, 2003
12766930
Ritchie CW, Chiu E, Harrigan S, et al: The impact upon extra-pyramidal
side effects, clinical symptoms and quality of life of a switch from
conventional to atypical antipsychotics (risperidone or olanzapine) in
elderly patients with schizophrenia. Int J Geriatr Psychiatry 18(5):432–
440, 2003 12766921
Ritchie CW, Chiu E, Harrigan S, et al: A comparison of the efficacy and
safety of olanzapine and risperidone in the treatment of elderly patients
with schizophrenia: an open study of six months duration. Int J Geriatr
Psychiatry 21(2): 171–179, 2006 16416458
Rive B, Gauthier S, Costello S, et al: Synthesis and comparison of the meta-
analyses evaluating the efficacy of memantine in moderate to severe
stages of Alzheimer’s disease. CNS Drugs 27(7):573–582, 2013
23765561
Robinson DS, Amsterdam JD: The selegiline transdermal system in major
depressive disorder: a systematic review of safety and tolerability. J
Affect Disord 105(1–3): 15–23, 2008 17568687
Robinson M, Oakes TM, Raskin J, et al: Acute and long-term treatment of
late-life major depressive disorder: duloxetine versus placebo. Am J
Geriatr Psychiatry 22(1): 34–45, 2014 24314888
Robinson RG, Schultz SK, Castillo C, et al: Nortriptyline versus fluoxetine
in the treatment of depression and in short-term recovery after stroke: a
placebo-controlled, double-blind study. Am J Psychiatry 157(3):351–
359, 2000 10698809
Robinson RG, Jorge RE, Moser DJ, et al: Escitalopram and problem-
solving therapy for prevention of poststroke depression: a randomized
controlled trial. JAMA 299(20):2391–2400, 2008 18505948
Rocca P, Calvarese P, Faggiano F, et al: Citalopram versus sertraline in late-
life nonmajor clinically significant depression: a 1-year follow-up
clinical trial. J Clin Psychiatry 66(3):360–369, 2005 15766303
Rochon PA, Stukel TA, Sykora K, et al: Atypical antipsychotics and
parkinsonism. Arch Intern Med 165(16):1882–1888, 2005 16157833
Rodda J, Morgan S, Walker Z: Are cholinesterase inhibitors effective in the
management of the behavioral and psychological symptoms of
dementia in Alzheimer’s disease? A systematic review of randomized,
placebo-controlled trials of donepezil, rivastigmine and galantamine.
Int Psychogeriatr 21(5):813–824, 2009 19538824
Rolinski M, Fox C, Maidment I, et al: Cholinesterase inhibitors for
dementia with Lewy bodies, Parkinson’s disease dementia and
cognitive impairment in Parkinson’s disease. Cochrane Database Syst
Rev 3:CD006504, 2012 22419314
Roose SP, Dalack GW, Glassman AH, et al: Cardiovascular effects of
bupropion in depressed patients with heart disease. Am J Psychiatry
148(4):512–516, 1991 1900980
Roose SP, Nelson JC, Salzman C, et al; Mirtazapine in the Nursing Home
Study Group: Open-label study of mirtazapine orally disintegrating
tablets in depressed patients in the nursing home. Curr Med Res Opin
19(8):737–746, 2003 14687445
Roose SP, Miyazaki M, Devanand D, et al: An open trial of venlafaxine for
the treatment of late-life atypical depression. Int J Geriatr Psychiatry
19(10):989–994, 2004a 15449363
Roose SP, Sackeim HA, Krishnan KR, et al; Old-Old Depression Study
Group: Antidepressant pharmacotherapy in the treatment of depression
in the very old: a randomized, placebo-controlled trial. Am J Psychiatry
161(11):2050–2059, 2004b 15514406
Rosen J, Sweet R, Pollock BG, et al: Nortriptyline in the hospitalized
elderly: tolerance and side effect reduction. Psychopharmacol Bull
29(2):327–331, 1993 8290682
Rosenberg C, Lauritzen L, Brix J, et al: Citalopram versus amitriptyline in
elderly depressed patients with or without mild cognitive dysfunction:
a Danish multicentre trial in general practice. Psychopharmacol Bull
40(1):63–73, 2007 17285097
Rosenberg PB, Drye LT, Martin BK, et al; DIADS-2 Research Group:
Sertraline for the treatment of depression in Alzheimer disease. Am J
Geriatr Psychiatry 18(2):136–145, 2010 20087081
Ross J: Discontinuation of lithium augmentation in geriatric patients with
unipolar depression: a systematic review. Can J Psychiatry 53(2):117–
120, 2008 18357930
Rossini D, Serretti A, Franchini L, et al: Sertraline versus fluvoxamine in
the treatment of elderly patients with major depression: a double-blind,
randomized trial. J Clin Psychopharmacol 25(5):471–475, 2005
16160624
Rush AJ, Trivedi MH, Wisniewski SR, et al: Acute and longer-term
outcomes in depressed outpatients requiring one or several treatment
steps: a STAR*D report. Am J Psychiatry 163(11):1905–1917, 2006a
17074942
Rush AJ, Trivedi MH, Wisniewski SR, et al; STAR*D Study Team:
Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for
depression. N Engl J Med 354(12):1231–1242, 2006b 16554525
Rush A, Wisniewski S, Warden D, et al: Selecting among second-step
antidepressant medication monotherapies: predictive value of clinical,
demographic, or first-step treatment features. Arch Gen Psychiatry
65:870–880, 2008 18678792
Sackeim HA, Haskett RF, Mulsant BH, et al: Continuation
pharmacotherapy in the prevention of relapse following
electroconvulsive therapy: a randomized controlled trial. JAMA
285(10):1299–1307, 2001 11255384
Saiz-Ruiz J, Ibañez A, Díaz-Marsá M, et al: Nefazodone in the treatment of
elderly patients with depressive disorders: a prospective, observational
study. CNS Drugs 16(9):635–643, 2002 12153334
Sajatovic M, Jaskiw G, Konicki PE, et al: Outcome of clozapine therapy for
elderly patients with refractory primary psychosis. Int J Geriatr
Psychiatry 12(5):553–558, 1997 9193964
Sajatovic M, Gyulai L, Calabrese JR, et al: Maintenance treatment
outcomes in older patients with bipolar I disorder. Am J Geriatr
Psychiatry 13(4):305–311, 2005a 15845756
Sajatovic M, Madhusoodanan S, Coconcea N: Managing bipolar disorder in
the elderly: defining the role of the newer agents. Drugs Aging
22(1):39–54, 2005b 15663348
Sajatovic M, Blow FC, Ignacio RV: Psychiatric comorbidity in older adults
with bipolar disorder. Int J Geriatr Psychiatry 21(6): 582–587, 2006
16783798
Sajatovic M, Ramsay E, Nanry K, Thompson T: Lamotrigine therapy in
elderly patients with epilepsy, bipolar disorder or dementia. Int J
Geriatr Psychiatry 22(10):945–950, 2007 17326238
Sajatovic M, Coconcea N, Ignacio RV, et al: Aripiprazole therapy in 20
older adults with bipolar disorder: a 12-week, open-label trial. J Clin
Psychiatry 69(1):41–46, 2008 18312036
Sajatovic M, Gildengers A, Al Jurdi RK, et al: Multisite, open-label,
prospective trial of lamotrigine for geriatric bipolar depression: a
preliminary report. Bipolar Disord 13(3):294–302, 2011 21676132
Sajatovic M, Forester BP, Gildengers A, Mulsant BH: Aging changes and
medical complexity in late-life bipolar disorder: emerging research
findings that may help advance care. Neuropsychiatry 3(6):621–633,
2013 24999372
Sajatovic M, Dines P, Fuentes-Casiano E, et al: Asenapine in the treatment
of older adults with bipolar disorder. Int J Geriatr Psychiatry. 2014 Oct
21. [Epub ahead of print] 25335125
Salzman C, Jeste DV, Meyer RE, et al: Elderly patients with dementia-
related symptoms of severe agitation and aggression: consensus
statement on treatment options, clinical trials methodology, and policy.
J Clin Psychiatry 69(6):889–898, 2008 18494535
Sasaki Y, Matsuyama T, Inoue S, et al: A prospective, open-label, flexible-
dose study of quetiapine in the treatment of delirium. J Clin Psychiatry
64(11):1316–1321, 2003 14658945
Satel SL, Nelson JC: Stimulants in the treatment of depression: a critical
overview. J Clin Psychiatry 50(7):241–249, 1989 2567730
Sato Y, Kondo I, Ishida S, et al: Decreased bone mass and increased bone
turnover with valproate therapy in adults with epilepsy. Neurology
57(3):445–449, 2001 11502911
Savaskan E, Müller SE, Böhringer A, et al: Antidepressive therapy with
escitalopram improves mood, cognitive symptoms, and identity
memory for angry faces in elderly depressed patients. Int J
Neuropsychopharmacol 11(3):381–388, 2008 17697395
Scarpini E, Bruno G, Zappalà G, et al: Cessation versus continuation of
galantamine treatment after 12 months of therapy in patients with
Alzheimer’s disease: a randomized, double blind, placebo controlled
withdrawal trial. J Alzheimers Dis 26(2):211–220, 2011 21606568
Schatzberg A, Roose S: A double-blind, placebo-controlled study of
venlafaxine and fluoxetine in geriatric outpatients with major
depression. Am J Geriatr Psychiatry 14(4):361–370, 2006 16582045
Schatzberg AF, Kremer C, Rodrigues HE, et al; Mirtazapine vs. Paroxetine
Study Group: Double-blind, randomized comparison of mirtazapine
and paroxetine in elderly depressed patients. Am J Geriatr Psychiatry
10(5):541–550, 2002 12213688
Schiff GD, Galanter WL, Duhig J, et al: Principles of conservative
prescribing. Arch Intern Med 171(16):1433–1440, 2011 21670331
Schneider LS, Nelson JC, Clary CM, et al; Sertraline Elderly Depression
Study Group: An 8-week multicenter, parallel-group, double-blind,
placebo-controlled study of sertraline in elderly outpatients with major
depression. Am J Psychiatry 160(7): 1277–1285, 2003 12832242
Schneider LS, Dagerman KS, Insel P: Risk of death with atypical
antipsychotic drug treatment for dementia: meta-analysis of
randomized placebo-controlled trials. JAMA 294(15):1934–1943, 2005
16234500
Schneider LS, Dagerman K, Insel PS: Efficacy and adverse effects of
atypical antipsychotics for dementia: meta-analysis of randomized,
placebo-controlled trials. Am J Geriatr Psychiatry 14(3):191–210,
2006a 16505124
Schneider LS, Tariot PN, Dagerman KS, et al; CATIE-AD Study Group:
Effectiveness of atypical antipsychotic drugs in patients with
Alzheimer’s disease. N Engl J Med 355(15):1525–1538, 2006b
17035647
Schöne W, Ludwig M: A double-blind study of paroxetine compared with
fluoxetine in geriatric patients with major depression. J Clin
Psychopharmacol 13 (6 suppl 2): 34S–39S, 1993 8106654
Schueler YB, Koesters M, Wieseler B, et al: A systematic review of
duloxetine and venlafaxine in major depression, including unpublished
data. Acta Psychiatr Scand 123(4):247–265, 2011 20831742
Schwaninger M, Ringleb P, Winter R, et al: Elevated plasma concentrations
of homocysteine in antiepileptic drug treatment. Epilepsia 40(3):345–
350, 1999 10080517
Scott J, Greenwald BS, Kramer E, Shuwall M: Atypical (second generation)
antipsychotic treatment response in very late-onset schizophrenia-like
psychosis. Int Psychogeriatr 23(5):742–748, 2011 21118614
Semel D, Murphy TK, Zlateva G, et al: Evaluation of the safety and
efficacy of pregabalin in older patients with neuropathic pain: results
from a pooled analysis of 11 clinical studies. BMC Fam Pract 11:85,
2010 21054853
Semenchuk MR, Sherman S, Davis B: Double-blind, randomized trial of
bupropion SR for the treatment of neuropathic pain. Neurology
57(9):1583–1588, 2001 11706096
Serebruany VL, Glassman AH, Malinin AI, et al; Sertraline AntiDepressant
Heart Attack Randomized Trial Study Group: Platelet/endothelial
biomarkers in depressed patients treated with the selective serotonin
reuptake inhibitor sertraline after acute coronary events: the Sertraline
AntiDepressant Heart Attack Randomized Trial (SADHART) Platelet
Substudy. Circulation 108(8):939–944, 2003 12912814
Seritan AL: Prevent drug-drug interactions with cholinesterase inhibitors:
avoid adverse events when prescribing medications for patients with
dementia. Curr Psychiatry 7:57–67, 2008
Shea ML, Garfield LD, Teitelbaum S, et al: Serotonin-norepinephrine
reuptake inhibitor therapy in late-life depression is associated with
increased marker of bone resorption. Osteoporos Int 24(5):1741–1749,
2013 23358607
Sheffrin M, Driscoll HC, Lenze EJ, et al: Pilot study of augmentation with
aripiprazole for incomplete response in late-life depression: getting to
remission. J Clin Psychiatry 70(2):208–213, 2009 19210951
Sheikh JI, Cassidy EL, Doraiswamy PM, et al: Efficacy, safety, and
tolerability of sertraline in patients with late-life depression and
comorbid medical illness. J Am Geriatr Soc 52(1):86–92, 2004a
14687320
Sheikh JI, Lauderdale SA, Cassidy EL: Efficacy of sertraline for panic
disorder in older adults: a preliminary open-label trial. Am J Geriatr
Psychiatry 12(2):230, 2004b 15010352
Shelton C, Entsuah R, Padmanabhan SK, Vinall PE: Venlafaxine XR
demonstrates higher rates of sustained remission compared to
fluoxetine, paroxetine or placebo. Int Clin Psychopharmacol 20(4):
233–238, 2005 15933485
Shelton RC, Tollefson GD, Tohen M, et al: A novel augmentation strategy
for treating resistant major depression. Am J Psychiatry 158(1):131–
134, 2001 11136647
Shin D, Oh YH, Eom CS, Park SM: Use of selective serotonin reuptake
inhibitors and risk of stroke: a systematic review and meta-analysis. J
Neurol 261(4):686–695, 2014 24477492
Shulman KI: Lithium for older adults with bipolar disorder: Should it still
be considered a first-line agent? Drugs Aging 27(8):607–615, 2010
20658789
Shulman KI, Walker SE: A reevaluation of dietary restrictions for
irreversible monoamine oxidase inhibitors. Psychiatr Ann 31:378–384,
2001
Shulman KI, Fischer HD, Herrmann N, et al: Current prescription patterns
and safety profile of irreversible monoamine oxidase inhibitors: a
population-based cohort study of older adults. J Clin Psychiatry
70(12):1681–1686, 2009 19852903
Sink KM, Holden KF, Yaffe K: Pharmacological treatment of
neuropsychiatric symptoms of dementia: a review of the evidence.
JAMA 293(5):596–608, 2005 15687315
Skrobik YK, Bergeron N, Dumont M, et al: Olanzapine vs haloperidol:
treating delirium in a critical care setting. Intensive Care Med
30(3):444–449, 2004 14685663
Smeraldi E, Rizzo F, Crespi G: Double-blind, randomized study of
venlafaxine, clomipramine and trazodone in geriatric patients with
major depression. Prim Care Psychiatry 4:189–195, 1998
Smith D, Dempster C, Glanville J, et al: Efficacy and tolerability of
venlafaxine compared with selective serotonin reuptake inhibitors and
other antidepressants: a meta-analysis. Br J Psychiatry 180:396–404,
2002 11983635
Sneed JR, Culang ME, Keilp JG, et al: Antidepressant medication and
executive dysfunction: a deleterious interaction in late-life depression.
Am J Geriatr Psychiatry 18(2):128–135, 2010 20104069
Soares CN, Thase ME, Clayton A, et al: Desvenlafaxine and escitalopram
for the treatment of postmenopausal women with major depressive
disorder. Menopause 17(4):700–711, 2010 20539246
Sommer BR, Fenn HH, Ketter TA: Safety and efficacy of anticonvulsants in
elderly patients with psychiatric disorders: oxcarbazepine, topiramate
and gabapentin. Expert Opin Drug Saf 6(2):133–145, 2007 17367259
Sonnenberg CM, Deeg DJ, Comijs HC, et al: Trends in antidepressant use
in the older population: results from the LASA study over a period of
10 years. J Affect Disord 111(2-3):299–305, 2008 18442857
Spier SA: Use of bupropion with SRIs and venlafaxine. Depress Anxiety
7(2):73–75, 1998 9614595
Spina E, Trifirò G, Caraci F: Clinically significant drug interactions with
newer antidepressants. CNS Drugs 26(1):39–67, 2012 22171584
Sproule BA, Hardy BG, Shulman KI: Differential pharmacokinetics of
lithium in elderly patients. Drugs Aging 16(3):165–177, 2000
10803857
Stahl SM, Entsuah R, Rudolph RL: Comparative efficacy between
venlafaxine and SSRIs: a pooled analysis of patients with depression.
Biol Psychiatry 52(12):1166–1174, 2002 12488062
Steffens DC, Doraiswamy PM, McQuoid DR: Bupropion SR in the
naturalistic treatment of elderly patients with major depression. Int J
Geriatr Psychiatry 16(9): 862–865, 2001 11571765
Steffens DC, Nelson JC, Eudicone JM, et al: Efficacy and safety of
adjunctive aripiprazole in major depressive disorder in older patients: a
pooled subpopulation analysis. Int J Geriatr Psychiatry 26(6): 564–572,
2011 20827794
Steinberg JR: Anxiety in elderly patients. A comparison of azapirones and
benzodiazepines. Drugs Aging 5(5):335–345, 1994 7833587
Strand M, Hetta J, Rosen A, et al: A double-blind, controlled trial in
primary care patients with generalized anxiety: a comparison between
buspirone and oxazepam. J Clin Psychiatry 51 (suppl):40–45, 1990
2211567
Street JS, Clark WS, Gannon KS, et al; The HGEU Study Group:
Olanzapine treatment of psychotic and behavioral symptoms in patients
with Alzheimer disease in nursing care facilities: a double-blind,
randomized, placebo-controlled trial. Arch Gen Psychiatry 57(10):968–
976, 2000 11015815
Streim JE, Porsteinsson AP, Breder CD, et al: A randomized, double-blind,
placebo-controlled study of aripiprazole for the treatment of psychosis
in nursing home patients with Alzheimer disease. Am J Geriatr
Psychiatry 16(7):537–550, 2008 18591574
Suh GH, Son HG, Ju YS, et al: A randomized, double-blind, crossover
comparison of risperidone and haloperidol in Korean dementia patients
with behavioral disturbances. Am J Geriatr Psychiatry 12(5): 509–516,
2004 15353389
Sultzer DL, Gray KF, Gunay I, et al: A double-blind comparison of
trazodone and haloperidol for treatment of agitation in patients with
dementia. Am J Geriatr Psychiatry 5(1):60–69, 1997 9169246
Suppes T, Eudicone J, McQuade R, et al: Efficacy and safety of aripiprazole
in subpopulations with acute manic or mixed episodes of bipolar I
disorder. J Affect Disord 107(1-3):145–154, 2008 17904226
Suzuki T, Remington G, Uchida H, et al: Management of schizophrenia in
late life with antipsychotic medications: a qualitative review. Drugs
Aging 28(12):961–980, 2011 22117095
Szuba MP, Leuchter AF: Falling backward in two elderly patients taking
bupropion. J Clin Psychiatry 53(5):157–159, 1992 1592841
Tadger S, Paleacu D, Barak Y: Quetiapine augmentation of antidepressant
treatment in elderly patients suffering from depressive symptoms: a
retrospective chart review. Arch Gerontol Geriatr 53(1):104–105, 2011
20678815
Tahir TA, Eeles E, Karapareddy V, et al: A randomized controlled trial of
quetiapine versus placebo in the treatment of delirium. J Psychosom
Res 69(5):485–490, 2010 20955868
Taragano FE, Lyketsos CG, Mangone CA, et al: A double-blind,
randomized, fixed-dose trial of fluoxetine vs. amitriptyline in the
treatment of major depression complicating Alzheimer’s disease.
Psychosomatics 38(3):246–252, 1997 9136253
Tariot PN, Erb R, Podgorski CA, et al: Efficacy and tolerability of
carbamazepine for agitation and aggression in dementia. Am J
Psychiatry 155(1):54–61, 1998 9433339
Tariot PN, Salzman C, Yeung PP, et al: Long-Term use of quetiapine in
elderly patients with psychotic disorders. Clin Ther 22(9):1068–1084,
2000 11048905
Tariot PN, Farlow MR, Grossberg GT, et al; Memantine Study Group:
Memantine treatment in patients with moderate to severe Alzheimer
disease already receiving donepezil: a randomized controlled trial.
JAMA 291(3):317–324, 2004 14734594
Tariot PN, Raman R, Jakimovich L, et al; Alzheimer’s Disease Cooperative
Study; Valproate Nursing Home Study Group: Divalproex sodium in
nursing home residents with possible or probable Alzheimer Disease
complicated by agitation: a randomized, controlled trial. Am J Geriatr
Psychiatry 13(11):942–949, 2005 16286437
Tariot PN, Schneider LS, Cummings J, et al; Alzheimer’s Disease
Cooperative Study Group: Chronic divalproex sodium to attenuate
agitation and clinical progression of Alzheimer disease. Arch Gen
Psychiatry 68(8):853–861, 2011 21810649
Tashkin D, Kanner R, Bailey W, et al: Smoking cessation in patients with
chronic obstructive pulmonary disease: a double-blind, placebo-
controlled, randomised trial. Lancet 357(9268):1571–1575, 2001
11377644
Tedeschini E, Levkovitz Y, Iovieno N, et al: Efficacy of antidepressants for
late-life depression: a meta-analysis and meta-regression of placebo-
controlled randomized trials. J Clin Psychiatry 72(12):1660–1668,
2011 22244025
Teri L, Logsdon RG, Peskind E, et al; Alzheimer’s Disease Cooperative
Study: Treatment of agitation in AD: a randomized, placebo-controlled
clinical trial. Neurology 55(9):1271–1278, 2000 11087767
Thacker EL, Schneeweiss S: Initiation of acetylcholinesterase inhibitors and
complications of chronic airways disorders in elderly patients. Drug
Saf 29(11):1077–1085, 2006 17061913
Thaler KJ, Morgan LC, Van Noord M, et al: Comparative effectiveness of
second-generation antidepressants for accompanying anxiety, insomnia,
and pain in depressed patients: a systematic review. Depress Anxiety
29(6):495–505, 2012 22553134
Thase ME: Effects of venlafaxine on blood pressure: a meta-analysis of
original data from 3744 depressed patients. J Clin Psychiatry
59(10):502–508, 1998 9818630
Thase ME: What role do atypical antipsychotic drugs have in treatment-
resistant depression? J Clin Psychiatry 63(2):95–103, 2002 11874227
Thase ME, Entsuah AR, Rudolph RL: Remission rates during treatment
with venlafaxine or selective serotonin reuptake inhibitors. Br J
Psychiatry 178:234–241, 2001 11230034
Thase ME, Entsuah R, Cantillon M, et al: Relative antidepressant efficacy
of venlafaxine and SSRIs: sex-age interactions. J Womens Health
(Larchmt) 14(7):609–616, 2005a 16181017
Thase ME, Haight BR, Richard N, et al: Remission rates following
antidepressant therapy with bupropion or selective serotonin reuptake
inhibitors: a meta-analysis of original data from 7 randomized
controlled trials. J Clin Psychiatry 66(8):974–981, 2005b 16086611
Thase ME, Tran PV, Wiltse C, et al: Cardiovascular profile of duloxetine, a
dual reuptake inhibitor of serotonin and norepinephrine. J Clin
Psychopharmacol 25(2):132–140, 2005c 15738744
Thompson DS: Mirtazapine for the treatment of depression and nausea in
breast and gynecological oncology. Psychosomatics 41(4):356–359,
2000 10906359
Tollefson GD, Bosomworth JC, Heiligenstein JH, et al; The Fluoxetine
Collaborative Study Group: A double-blind, placebo-controlled clinical
trial of fluoxetine in geriatric patients with major depression. Int
Psychogeriatr 7(1):89–104, 1995 7579025
Trappler B, Cohen CI: Use of SSRIs in “very old” depressed nursing home
residents. Am J Geriatr Psychiatry 6(1):83–89, 1998 9469218
Trick L, Stanley N, Rigney U, Hindmarch I: A double-blind, randomized,
26-week study comparing the cognitive and psychomotor effects and
efficacy of 75 mg (37.5 mg b.i.d.) venlafaxine and 75 mg (25 mg mane,
50 mg nocte) dothiepin in elderly patients with moderate major
depression being treated in general practice. J Psychopharmacol
18(2):205–214, 2004 15260909
Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team:
Medication augmentation after the failure of SSRIs for depression. N
Engl J Med 354(12):1243–1252, 2006 16554526
Tsai CS, Wu CL, Chou SY, et al: Prevention of poststroke depression with
milnacipran in patients with acute ischemic stroke: a double-blind
randomized placebo-controlled trial. Int Clin Psychopharmacol
26(5):263–267, 2011 21811172
Tsuboi T, Suzuki T, Uchida H: A tipping point in drug dosing in late-life
schizophrenia. Curr Psychiatry Rep 13(3):225–233, 2011 21327902
Tzimos A, Samokhvalov V, Kramer M, et al: Safety and tolerability of oral
paliperidone extended-release tablets in elderly patients with
schizophrenia: a double-blind, placebo-controlled study with six-month
open-label extension. Am J Geriatr Psychiatry 16(1):31–43, 2008
18165460
Uchida H, Mamo DC, Mulsant BH, et al: Increased antipsychotic sensitivity
in elderly patients: evidence and mechanisms. J Clin Psychiatry
70(3):397–405, 2009 19192476
U.S. Food and Drug Administration: FDA Drug Safety Communication:
Revised recommendations for Celexa (citalopram hydrobromide)
related to a potential risk of abnormal heart rhythms with high doses.
March 2012. Available at:
http://www.fda.gov/Drugs/DrugSafety/ucm297391.htm. Accessed June
10, 2014.
van Iersel MB, Zuidema SU, Koopmans RT, et al: Antipsychotics for
behavioural and psychological problems in elderly people with
dementia: a systematic review of adverse events. Drugs Aging
22(10):845–858, 2005 16245958
Varanese S, Perfetti B, Gilbert-Wolf R, et al: Modafinil and armodafinil
improve attention and global mental status in Lewy bodies disorders:
preliminary evidence. Int J Geriatr Psychiatry 28(10):1095–1097, 2013
24038163
Verhey FR, Verkaaik M, Lousberg R; Olanzapine-Haloperidol in Dementia
Study group: Olanzapine versus haloperidol in the treatment of
agitation in elderly patients with dementia: results of a randomized
controlled double-blind trial. Dement Geriatr Cogn Disord 21(1):1–8,
2006 16244481
Vis PM, van Baardewijk M, Einarson TR: Duloxetine and venlafaxine-XR
in the treatment of major depressive disorder: a meta-analysis of
randomized clinical trials. Ann Pharmacother 39(11):1798–1807, 2005
16189284
von Wolff A, Hölzel LP, Westphal A, et al: Selective serotonin reuptake
inhibitors and tricyclic antidepressants in the acute treatment of chronic
depression and dysthymia: a systematic review and meta-analysis. J
Affect Disord 144(1–2):7–15, 2013 22963896
Vozoris NT, Fischer HD, Wang X, et al: Benzodiazepine drug use and
adverse respiratory outcomes among older adults with COPD. Eur
Respir J 44(2):332–340, 2014 24743966
Wakelin JS: Fluvoxamine in the treatment of the older depressed patient;
double-blind, placebo-controlled data. Int Clin Psychopharmacol
1(3):221–230, 1986 3104446
Walker Z, Grace J, Overshot R, et al: Olanzapine in dementia with Lewy
bodies: a clinical study. Int J Geriatr Psychiatry 14(6):459–466, 1999
10398356
Wallace AE, Kofoed LL, West AN: Double-blind, placebo-controlled trial
of methylphenidate in older, depressed, medically ill patients. Am J
Psychiatry 152(6):929–931, 1995 7755127
Wang HR, Woo YS, Bahk WM: Atypical antipsychotics in the treatment of
delirium. Psychiatry Clin Neurosci 67(5):323–331, 2013 23859663
Wang PS, Bohn RL, Glynn RJ, et al: Zolpidem use and hip fractures in
older people. J Am Geriatr Soc 49(12):1685–1690, 2001 11844004
Wang PS, Schneeweiss S, Avorn J, et al: Risk of death in elderly users of
conventional vs. atypical antipsychotic medications. N Engl J Med
353(22):2335–2341, 2005 16319382
Wang YP, Chen YT, Tsai CF, et al: Short-term use of serotonin reuptake
inhibitors and risk of upper gastrointestinal bleeding. Am J Psychiatry
171(1):54–61, 2014 24030313
Wehmeier PM, Kluge M, Maras A, et al: Fluoxetine versus trimipramine in
the treatment of depression in geriatric patients. Pharmacopsychiatry
38(1):13–16, 2005 15706460
Weich S, Pearce HL, Croft P, et al: Effect of anxiolytic and hypnotic drug
prescriptions on mortality hazards: retrospective cohort study. BMJ
348:g1996, 2014
Weihs KL, Settle EC Jr, Batey SR, et al: Bupropion sustained release versus
paroxetine for the treatment of depression in the elderly. J Clin
Psychiatry 61(3):196–202, 2000 10817105
Weintraub D, Rosenberg PB, Drye LT, et al; DIADS-2 Research Group:
Sertraline for the treatment of depression in Alzheimer disease: week-
24 outcomes. Am J Geriatr Psychiatry 18(4):332–340, 2010 20220589
Wenzel-Seifert K, Wittmann M, Haen E: QTc prolongation by psychotropic
drugs and the risk of torsade de pointes. Dtsch Arztebl Int
108(41):687–693, 2011 22114630
Whyte EM, Basinski J, Farhi P, et al: Geriatric depression treatment in
nonresponders to selective serotonin reuptake inhibitors. J Clin
Psychiatry 65(12):1634–1641, 2004 15641868
Whyte EM, Romkes M, Mulsant BH, et al: CYP2D6 genotype and
venlafaxine-XR concentrations in depressed elderly. Int J Geriatr
Psychiatry 21(6):542–549, 2006 16642541
Wilcock GK, Ballard CG, Cooper JA, et al: Memantine for
agitation/aggression and psychosis in moderately severe to severe
Alzheimer’s disease: a pooled analysis of 3 studies. J Clin Psychiatry
69(3):341–348, 2008 18294023
Wilner KD, Tensfeldt TG, Baris B, et al: Single- and multiple-dose
pharmacokinetics of ziprasidone in healthy young and elderly
volunteers. Br J Clin Pharmacol 49(Suppl 1):15S–20S, 2000 10771449
Wilson K, Mottram P: A comparison of side effects of selective serotonin
reuptake inhibitors and tricyclic antidepressants in older depressed
patients: a meta-analysis. Int J Geriatr Psychiatry 19(8):754–762, 2004
15290699
Winblad B, Kilander L, Eriksson S, et al; Severe Alzheimer’s Disease
Study Group: Donepezil in patients with severe Alzheimer’s disease:
double-blind, parallel-group, placebo-controlled study. Lancet
367(9516):1057–1065, 2006 16581404
Wingen M, Bothmer J, Langer S, et al: Actual driving performance and
psychomotor function in healthy subjects after acute and subchronic
treatment with escitalopram, mirtazapine, and placebo: a crossover
trial. J Clin Psychiatry 66(4):436–443, 2005 15816785
Wohlreich MM, Sullivan MD, Mallinckrodt CH, et al: Duloxetine for the
treatment of recurrent major depressive disorder in elderly patients:
treatment outcomes in patients with comorbid arthritis. Psychosomatics
50(4):402–412, 2009 19687181
Wylie ME, Miller MD, Shear MK, et al: Fluvoxamine pharmacotherapy of
anxiety disorders in later life: preliminary open-trial data. J Geriatr
Psychiatry Neurol 13(1): 43–48, 2000 10753007
Yang CH, Tsai SJ, Hwang JP: The efficacy and safety of quetiapine for
treatment of geriatric psychosis. J Psychopharmacol 19(6): 661–666,
2005 16272189
Young RC, Gyulai L, Mulsant BH, et al: Pharmacotherapy of bipolar
disorder in old age: review and recommendations. Am J Geriatr
Psychiatry 12(4):342–357, 2004 15249272
Young RC, Schulberg HC, Gildengers AG, et al: Conceptual and
methodological issues in designing a randomized, controlled treatment
trial for geriatric bipolar disorder: GERI-BD. Bipolar Disord 12(1):56–
67, 2010 20148867
Yuan Y, Tsoi K, Hunt RH: Selective serotonin reuptake inhibitors and risk
of upper GI bleeding: confusion or confounding? Am J Med
119(9):719–727, 2006 16945603
Zhang H, Lu Y, Liu M, et al: Strategies for prevention of postoperative
delirium: a systematic review and meta-analysis of randomized trials.
Crit Care 17(2):R47, 2013 23506796
Zheng L, Mack WJ, Dagerman KS, et al: Metabolic changes associated
with second-generation antipsychotic use in Alzheimer’s disease
patients: the CATIE-AD study. Am J Psychiatry 166(5):583–590, 2009
19369318
Zhong KX, Tariot PN, Mintzer J, et al: Quetiapine to treat agitation in
dementia: a randomized, double-blind, placebo-controlled study. Curr
Alzheimer Res 4(1): 81–93, 2007 17316169
Zimmer B, Kant R, Zeiler D, et al: Antidepressant efficacy and
cardiovascular safety of venlafaxine in young vs old patients with
comorbid medical disorders. Int J Psychiatry Med 27(4):353–364, 1997
9565731
Suggested Readings
Antidepressant Medications
Mulsant BH, Blumberger DM, Ismail Z, et al: A systematic approach to the
pharmacotherapy of geriatric major depression. Clin Geriatr Med
30(3):517–534, 2014
Antipsychotic Medications
Maher AR, Maglione M, Bagley S, et al: Efficacy and comparative
effectiveness of atypical antipsychotic medications for off-label uses in
adults: a systematic review and meta-analysis. JAMA 306(12): 1359–
1369, 2011 21954480
Meagher DJ, McLoughlin L, Leonard M, et al: What do we really know
about the treatment of delirium with antipsychotics? Ten key issues for
delirium pharmacotherapy. Am J Geriatr Psychiatry 21(12): 1223–
1238, 2013 2356742
Suzuki T, Remington G, Uchida H, et al: Management of schizophrenia in
late life with antipsychotic medications: a qualitative review. Drugs
Aging 28(12):961–980, 2011 22117095
Mood Stabilizers
Sajatovic M, Forester BP, Gildengers A, Mulsant BH: Aging changes and
medical complexity in late-life bipolar disorder: emerging research
findings that may help advance care. Neuropsychiatry 3(6): 621–633,
2013 24999372
Cognitive Enhancers
Herrmann N, Lanctôt KL, Hogan DB: Pharmacological recommendations
for the symptomatic treatment of dementia: the Canadian Consensus
Conference on the Diagnosis and Treatment of Dementia 2012.
Alzheimers Res Ther 8 (suppl 1): S5, 2013 24565367
CHAPTER 21
History
In 1935, the Hungarian neuropsychiatrist Ladislas Meduna chemically
induced seizures in a small series of patients with schizophrenia (Shorter
and Healy 2007). His rationale for doing so was based on the hypothesis
(later shown to be incorrect) that individuals with epilepsy had a reduced
incidence of schizophrenia. Having achieved some partial success in
therapeutic outcome, Meduna’s new pharmacoconvulsive therapy was
greeted with great acclaim as a means to manage what had been an
otherwise untreatable illness. When this treatment modality spread to Italy
shortly thereafter, another neuropsychiatrist, Ugo Cerletti, was impressed by
the efficacy of this new treatment but was also frustrated by its technical
complexity (Shorter and Healy 2007). From his work as an experimental
epileptologist, Cerletti was aware of an electrical model of seizure induction
that had been used in animal research and that offered the opportunity for
more efficient and less complex treatment than pharmacoconvulsive
therapy. After further experimentation, Cerletti and his assistant Lucio Bini
were successful in using electrical seizure induction to treat one of their
patients with schizophrenia, and ECT rapidly replaced pharmacoconvulsive
therapy throughout the world.
Although ECT was first used in the treatment of schizophrenia,
clinicians soon realized that its highest therapeutic potency was in treating
mood disorders. The peak in ECT utilization was from the early 1940s
through the mid-1950s, at which point the first effective antipsychotic and
antidepressant medications came into clinical use. Although these new
psychopharmacological agents obviated the need for ECT in many
individuals, trials comparing the antidepressant efficacy of ECT and these
medications indicate that ECT remains the most rapid and effective means
to induce remission (Husain et al. 2004; UK ECT Review Group 2003).
In the early days of ECT, there was considerable fear concerning the use
of ECT in older adults, largely because of the medical comorbidity that is
common in this age group. The cardiovascular physiology of ECT was not
yet well understood, and more recent procedural innovations—such as
oxygenation, muscular relaxation, general anesthesia, and physiological
monitoring—had not yet been instituted. Instead, ECT during that early
period was accomplished in a distinctly nonmedical setting, often in
psychiatrists’ offices, without the presence of other medical staff or medical
support resources.
As ECT methodology became more refined, practitioners became more
willing to use it with previously underserved populations, including older
adults. In Texas, which has mandatory reporting of ECT use, 19% of ECT
treatment courses were administered to individuals age 65 years or older
between September 1, 2012, and August 31, 2013 (Texas Department of
State Health Services 2014).
Indications for ECT
Diagnostic Indications
The most common diagnostic indication for ECT is major depression
(American Psychiatric Association 2001). A significant body of literature
not only supports the efficacy of ECT for major depression but suggests that
ECT is the most rapid and effective treatment for this condition (Husain et
al. 2004). This literature includes a series of randomized, double-blind,
placebo-controlled studies in which the placebo control was “sham ECT,”
whereby subjects received all aspects of a usual clinical ECT treatment
except the electrical stimulus (Brandon 1986). Evidence from meta-
analytical studies also suggests greater efficacy of ECT than of
antidepressant medication (Janicak et al. 1985; UK ECT Review Group
2003). Although this type of analysis has not been carried out comparing
ECT with newer antidepressant medications, there are some data supporting
an advantage for ECT compared with selective serotonin reuptake inhibitors
(Folkerts et al. 1997).
Regarding subtypes of depression, ECT appears to be effective in
treating both melancholic and severe nonmelancholic depression (Sackeim
and Rush 1995), as well as bipolar and unipolar major depression (Weiner
and Krystal 2001). In addition, ECT may be particularly effective in
treating psychotic major depression (Petrides et al. 2001; Sobin et al. 1996).
Although ECT is used more frequently for major depression than for
other illnesses and the vast majority of ECT research studies have focused
on this condition, evidence suggests that ECT has efficacy in treating a
number of other mental disorders. Two reviews of the efficacy of ECT in
the treatment of bipolar disorder, including mania, mixed states, and
depressive episodes, have reported substantial efficacy (Dierckx et al. 2012;
Versiani et al. 2011). In this regard, ECT has been reported to achieve a
response rate as high as 80%, to have efficacy equal to that of lithium, and
to have a significant advantage over lithium in patients who have not
responded to lithium or antipsychotic medication. The relative efficacy of
ECT compared with anticonvulsant antimanic agents has not yet been
studied, and no systematic studies exist to indicate the utility of ECT in
individuals with rapid-cycling bipolar disorder.
Another disorder for which ECT has efficacy is schizophrenia. Although
ECT was first employed as a treatment for this condition, the superior
response to ECT of patients with mood disorders was soon evident.
Following the development of antipsychotic medications in the late 1950s,
the use of ECT as a treatment for schizophrenia gradually declined.
Regardless, a number of studies have suggested that antipsychotic
medications and ECT have comparable efficacy (Pompili et al. 2013). In
addition, evidence suggests that for treatment of acute psychotic episodes,
the combination of antipsychotic medications and ECT may have greater
efficacy than either ECT or medications alone (Chanpattana et al. 2010;
Klapheke 1993). The combination of clozapine and ECT appears to be
particularly potent, even in patients whose psychotic ideation has been
nonresponsive to clozapine alone (Kho et al. 2004). However, no evidence
indicates that ECT has efficacy for the treatment of deficit or “negative”
symptoms of schizophrenia.
The presence of affective symptoms appears to increase the likelihood of
response to ECT in individuals with schizophrenia (Pompili et al. 2013). In
this regard, case reports and case series suggest that individuals with
schizoaffective disorder may respond better to ECT than do patients with
schizophrenia (American Psychiatric Association 2001). Catatonia, which
can be associated with both schizophrenia and mood disorders, is highly
responsive to ECT (Krystal and Coffey 1997), even when this condition is
associated with medical conditions such as systemic lupus erythematosus,
uremia, hepatic encephalopathy, porphyria, and hyperparathyroidism
(Fricchione et al. 1990; Rummans and Bassingthwaighte 1991).
Some evidence suggests that ECT may also be a useful treatment for
Parkinson’s disease when medication management fails or is not tolerated
(Fregni et al. 2005). However, it should also be noted that patients with
Parkinson’s disease may be at increased risk for developing cognitive side
effects and delirium with ECT (Figiel et al. 1991). In addition, the benefits
gained with ECT in regard to Parkinson’s symptoms and signs are transient,
necessitating the use of maintenance ECT in such cases (Wengel et al.
1998).
Mortality
Although it is difficult to establish an accurate mortality rate associated
with any medical procedure, it has been estimated that the overall mortality
rate for ECT is roughly 2 deaths per 100,000 treatments (Shiwach et al.
2001). A more recent study from the Veterans Administration patient
population found no mortalities in 73,440 treatments administered from
1999 to 2010 (Watts et al. 2011). This relatively low mortality rate appears
to be comparable to the rate associated with minor surgery and has been
considered to be less than that associated with pharmacotherapy with
tricyclic antidepressants (Sackeim 1998). Furthermore, some studies have
suggested that depressed inpatients who receive ECT have a lower
mortality rate after discharge than individuals who receive other types of
treatment (Philibert et al. 1995). It is important to understand, however, that
the likelihood of ECT-related death in high-risk populations—most
commonly in older adults—can be substantially higher than that mentioned
above. Still, even in such situations, the risk of undertaking ECT may be
lower than the risk of not doing it.
Cardiovascular Disorders
Fluctuations in pulse and blood pressure that occur during ECT treatments
may be associated with cardiovascular complications (Weiner et al. 2000).
Immediately after the stimulus, there is an increase in parasympathetic tone,
which can lead to a sudden but transient decrease in heart rate, not
uncommonly presenting as a brief period of asystole. The subsequent
induced seizure, however, is associated with a sympathetic surge that
markedly increases both blood pressure and heart rate. This sympathetic
surge is then followed by a relative increase in parasympathetic tone as the
induced seizure ends.
Despite these autonomic fluctuations, cardiovascular complications from
ECT rarely occur in patients without preexisting cardiovascular risk factors
(Takada et al. 2005; Weiner et al. 2000). The risk is increased in patients
with recent myocardial infarction, uncompensated congestive heart failure,
severe valvular disease, unstable aneurysm, unstable angina or active
cardiac ischemia, uncontrolled hypertension, high-grade atrioventricular
block, symptomatic ventricular arrhythmia, and supraventricular arrhythmia
with uncontrolled ventricular rate (American Psychiatric Association 2001;
Applegate 1997). For patients with these conditions, a consultation with a
cardiologist is recommended to help with the risk-benefit analysis and for
treatment modifications that may decrease risks. It has been proposed that
an assessment of functional cardiac status (such as a stress test) should be
considered in 1) men younger than age 60 and women under age 70 with
definite angina, 2) men older than age 60 and women over age 70 with
probable angina, 3) all patients with angina and two risk factors for
myocardial infarction, and 4) those with clinically significant extracardiac
vascular disease (Applegate 1997). A finding of good functional status
indicates that the risk is low; otherwise, further cardiac evaluation is
indicated.
Individuals with coronary artery disease are at risk for ischemia during
both the periods of relative parasympathetic tone and the periods of
increased sympathetic system tone (Christopher 2003; Weiner et al. 2000).
When parasympathetic tone is increased, there is a risk of ischemia due to
hypoperfusion, whereas when sympathetic activity rises, the increased
cardiac workload can lead to complications. The risks of complications due
to these factors can be decreased pharmacologically. Anticholinergic
medications such as atropine can be used to decrease the occurrence and
severity of bradycardia, β-adrenergic blockers can be used to decrease
cardiac workload, and nitrates or calcium channel blockers may be used to
decrease the risks of ischemia (Weiner et al. 2000). Typically, patients who
are receiving medications for the treatment of coronary artery disease at the
time of referral for ECT are maintained on those medications throughout
the ECT course, including administration before ECT on treatment days
(Applegate 1997). Changes to the medication regimen or the addition of
other medications to decrease the risks of complications should generally be
considered in conjunction with a cardiology consultant.
Patients with pathologies associated with low cardiac output such as
heart failure or those with severe valvular disease are at particular risk
during the sympathetic surge because of the increase in afterload and
decreased diastolic filling time (Stern et al. 1997). Such patients should not
be administered large volumes of fluid (Rayburn 1997). A number of
medications have been suggested as means to decrease risks in these
situations, including β-adrenergic blockers, α-adrenergic blockers, nitrites,
digitalis, and anticholinergic agents; however, the use of such agents is
controversial (Weiner et al. 2000). No single regimen appears to be optimal
for all patients with these diseases, and the treatment plan should be
individualized on the basis of risk-benefit considerations.
Arrhythmias may increase the risks of ECT (Takada et al. 2005).
Bradyarrhythmias are typically best managed with atropine to prevent
exacerbation during increases in parasympathetic tone. Because of the
anticonvulsant effects of lidocaine, ventricular ectopy should be treated
with other agents before the ECT treatment (Hood and Mecca 1983). There
is a risk that individuals with atrial fibrillation may experience spontaneous
cardioversion with ECT that can lead to an embolic event (Petrides and
Fink 1996). For this reason, consideration should be given to
echocardiography (to rule out a mural thrombus) and the use of
anticoagulants (Weiner et al. 2000).
Endocrinological Disorders
Several endocrinological disorders require special consideration before
ECT. The most commonly encountered is diabetes mellitus. Patients with
diabetes are more likely than other ECT patients to have problems
stemming from the need to fast from midnight until the time of the ECT
treatment. Insulin doses may need to be adjusted, and pretreatment
intravenous glucose administration can be considered if indicated (Weiner
et al. 2000).
In patients with hyperthyroidism and pheochromocytoma, β-adrenergic
blockers are typically administered to prevent triggering of a thyroid storm
or hypertensive crisis, which can be elicited by the sympathetic surge
(Weiner et al. 2000). In cases of pheochromocytoma, α-adrenergic and
tyrosine hydroxylase blockers may be needed.
Metabolic Disorders
The metabolic problems that are of primary concern are hyperkalemia and
hypokalemia, both of which may lead to cardiac arrhythmias. The former is
of particular concern because of the transient rise in serum potassium
caused by succinylcholine and the muscle activity that may occur during the
induced seizures (Christopher 2003; Weiner et al. 2000). In individuals with
hyperkalemia, prolonged paralysis and associated apnea induced by
succinylcholine may be seen. Although it is best to correct these conditions
before administering ECT, in cases where correction is not possible, the use
of paralytic agents other than succinylcholine should be considered.
Hematological Disorders
Thrombophlebitis carries with it the risk of embolism with ECT, a risk that
is generally easily avoided with the use of anticoagulant medications. The
use of warfarin has been recommended, with a goal of achieving an
international normalized ratio (prothrombin time normalized to the
laboratory control value) between 1.5 and 2.5 (Petrides and Fink 1996).
Pulmonary Disorders
Patients with asthma or chronic obstructive pulmonary disease have an
increased risk of posttreatment bronchospasm, which should be mitigated
by the use of bronchodilators (Weiner et al. 2000). Theophylline should be
avoided if possible, or the dose should be kept to a minimum because of an
increased risk of prolonged seizures.
Gastrointestinal Disorders
Patients with gastroesophageal reflux are commonly encountered in the
practice of ECT. Complications of aspiration may be diminished with the
use of a pretreatment histamine type 2 antagonist the night before and the
morning of treatment (Weiner et al. 2000). To increase gastric emptying,
pretreatment metoclopramide may be considered, and sodium citrate may
also be used to neutralize the acidity of stomach contents. Although it has
not been reported, fecal impaction has been mentioned as a risk factor for
intestinal rupture with ECT (Weiner et al. 2000). Consequently, in patients
referred for ECT, it is important to address constipation, which is
particularly common in the elderly population.
Genitourinary Disorders
Urinary retention could, in theory, lead to bladder rupture with ECT. As a
result, it has been recommended that patients void before ECT, and urinary
catheterization should be considered in those with significant obstruction or
difficulty urinating (Weiner et al. 2000).
Musculoskeletal Disorders
Musculoskeletal conditions, such as osteoporosis, unstable fractures, and
loose or damaged teeth, are common in older adults and carry an increased
risk of complications with ECT. Patients with osteoporosis or with recent or
unstable fractures are at risk for bone damage during the induced
convulsion, and should be considered high fall risks. The contraction of the
jaw muscles that leads to teeth clenching cannot be diminished with the use
of paralytic agents because it occurs by direct electrical stimulation of the
muscle tissue and not via neuromuscular transmission. Therefore, the use of
a mouth guard is always necessary. However, those with loose or damaged
teeth may require customized devices, dental treatment, or tooth extraction
before ECT.
Pre-ECT Evaluation
Informed Consent
The collaborative aspect of decision making has been formalized as the
legal doctrine of informed consent (American Psychiatric Association
2001). No patient with the capacity to give voluntary consent should be
treated with ECT without his or her written, informed consent. Although
there is no clear consensus about how to determine capacity to give consent,
this capacity has generally been interpreted as evidence that the patient can
understand information about the procedure and can act responsibly on the
basis of this information (American Psychiatric Association 2001). The
process of determining competency, the process for giving ECT
involuntarily in cases of emergency, and the specific procedures regarding
informed consent should be carried out as specified by applicable state
statutes. Because of the increased likelihood of cognitive dysfunction in
older adults, capacity to consent is of particular concern (Rabheru 2001).
At a minimum, written consent should be obtained before the initial
course of ECT, before a treatment course involving an unusually large
number of treatments, and before initiating continuation or maintenance
ECT (American Psychiatric Association 2001). To adequately convey the
risks and benefits, the consent form should include the following
information: 1) a description of treatment alternatives; 2) a detailed
description of how, when, and where ECT will be carried out; 3) a
discussion of options regarding electrode placement; 4) the typical range of
number of treatments; 5) a statement that there is no guarantee that the
treatment will be successful; 6) a statement that continuation or
maintenance treatment of some kind will be necessary; 7) a discussion of
the possible risks, including death, cardiac dysfunction, confusion, and
memory impairment; 8) a statement that the consent also applies to
emergency treatment that may be clinically necessary at times when the
patient is unconscious; 9) a listing of patient requirements during the ECT
course, such as taking nothing by mouth after midnight before treatment;
10) a statement that there has been an opportunity to ask questions and an
indication of who can be contacted with further questions; and 11) a
statement that consent is voluntary and can be withdrawn at any time
(American Psychiatric Association 2001).
Informed consent involves more than just signing a consent form; it
requires a consent discussion with the patient or surrogate (and, if possible,
a significant other). The consent discussion should include any significant
differences in likelihood of benefit or extent of risk from that depicted in
the consent form, and mention of such discussion should be briefly
documented in the patient’s medical record.
Management of Medications
Each pre-ECT assessment should include an evaluation of the patient’s
medications and recommendations about how medications should be taken
before and during the ECT course. Medications that are needed to decrease
medical risks should be continued, but their dosing and timing of
administration may need to be changed (American Psychiatric Association
2001). Similarly, orders should be written specifying dosage and timing of
any medications that are to be added to decrease risks based on the pre-ECT
evaluation. Other nonpsychotropic medications should be withheld until
after the treatment on ECT days or—in the case of those that interfere with
or increase the risks of ECT—should be discontinued.
Regarding the use of psychotropic medications during the ECT course,
there are considerable differences of opinion and great variation in practice.
The only situation in which compelling evidence exists for a potentiating
effect of psychotropic medication on ECT is the use of antipsychotic agents
in individuals with schizophrenia (and inferentially in those with psychotic
depression) (American Psychiatric Association 2001). The literature
regarding the benefits of antidepressant medication as a means to augment
the ECT response is unclear, although it does not appear that such a
combination is associated with significantly increased risk. The primary
reasons that have been given for the use of antidepressant medication
augmentation are that it is difficult at times to accomplish drug withdrawal
before instituting ECT, that there is a desire to decrease the risk of early
relapse after ECT, and that “there is nothing to lose.”
When possible, antidepressant medications should be chosen that have
relatively fewer effects on cardiac function. The following psychotropic
medications are among those that are best avoided or maintained at the
lowest possible levels: 1) lithium—it may increase the risks for delirium or
prolonged seizures; 2) benzodiazepines—their anticonvulsant properties
may decrease efficacy (but can be reversed with flumazenil at the time of
ECT) (Krystal et al. 1998); and 3) antiepileptic drugs—their anticonvulsant
properties may decrease efficacy, but they may be needed in those with
epilepsy or with very brittle bipolar disorder (in which case the medication
should be withheld the night before and the morning of treatment if
possible).
ECT Technique
Anesthetic Considerations
ECT is a procedure involving general anesthesia. Airway management, the
administration of medications necessary for anesthesia, and the handling of
medical emergencies during and immediately following the ECT procedure
are the responsibility of the anesthesia provider (American Psychiatric
Association 2001). Appropriate medical backup should be present,
particularly for high-risk cases.
The patient is ventilated by mask with 100% oxygen throughout the
procedure, beginning at least a few minutes before anesthesia induction and
lasting until a satisfactory level of spontaneous respiration is maintained
during the postictal period. General anesthesia is usually provided by
intravenous methohexital, typically 1 mg/kg (American Psychiatric
Association 2001; Ding and White 2002; Saito 2005). Because seizure
threshold (the amount of electricity necessary to induce a seizure) appears
to increase with age, and stimulus output of ECT devices is limited by the
U.S. Food and Drug Administration (FDA), difficulties in seizure induction
can be experienced with older adults, particularly late in an index ECT
course. In such situations, methohexital dosage can be slightly diminished
by concurrent usage of a short-acting sedative narcotic such as remifentanil,
or the anesthetic itself can be switched to one with less anticonvulsant
properties (e.g., etomidate or ketamine).
After loss of consciousness, the muscle relaxant succinylcholine is
administered intravenously, again with a typical dosage of 1 mg/kg
(American Psychiatric Association 2001). When the patient’s muscles are
relaxed (ascertainable by disappearance of relaxant-induced fasciculations
and loss of deep tendon reflexes or twitch response to a peripheral nerve
stimulator), the electrical stimulus can be delivered.
An anticholinergic medication, such as glycopyrrolate or atropine, may
be administered before anesthesia to minimize the risk of stimulus-related
asystole and the occurrence of postictal oral secretions. However, most
practitioners use such agents selectively because they potentiate seizure-
related tachycardia. When seizure-related hypertension and tachycardia are
severe or when prophylaxis is indicated on the basis of preexisting
cardiovascular disease, β-blocking medications (e.g., labetalol) are often
used to minimize these effects. Again, it is best to use such agents
selectively. When necessary, postictal agitation or delirium can be managed
with the use of intravenous midazolam (1 mg), haloperidol (2–5 mg), or the
α2 agonist dexmedetomidine (10–40 mcg), as well as by providing
reassurance and maintaining a quiet, low-light environment for the postictal
recovery process. Dexmedetomidine is interesting in that if it is
administered preictally, it will blunt the sympathetically mediated rise in
heart rate and blood pressure during the induced seizure (Aydogan et al.
2013).
With older adults, it is important to recognize that lower doses of
medications may be indicated because of altered metabolism or tolerance.
In addition, time to effect may be longer in older adults. On the other hand,
under some circumstances, higher doses may be necessary (e.g., more
relaxant agent may be needed for an individual with osteoporosis).
Physiological Monitoring
During ECT, as with any procedure utilizing general anesthesia, vital signs
and pulse oximetry are monitored throughout the procedure and during the
immediate postictal period until stabilization occurs. After spontaneous
respiration resumes and vital signs and oxygen saturation are trending
toward baseline, the patient is moved to a postanesthesia care unit or area
(previously referred to as a recovery room), where monitoring of vital signs
and oxygenation continues.
Both the motor and EEG representations of seizure activity are
monitored during ECT. To allow monitoring of the motor response in a
patient whose muscles are relaxed, a blood pressure cuff is placed around
the ankle and inflated to approximately 200 mm Hg just before
administration of the muscle relaxant. This action prevents muscle activity
distal to the cuff from being suppressed during the seizure. Ictal EEG
recordings are made, using recording leads placed on the head, in
conjunction with amplification and display capabilities built into the ECT
device. It is recommended that two EEG channels be recorded so that
seizure activity from both the left and the right cerebral hemispheres can be
monitored. Such recording can be accomplished by placing one pair of
recording electrodes over the left prefrontal and left mastoid areas and the
other pair over the homologous areas on the right.
Stimulus Dosing
All contemporary ECT devices used in the United States utilize a
bidirectional, constant-current, brief-pulse stimulus waveform. This
waveform is more efficient than the older sine-wave stimulus for inducing
seizures and allows ECT to be administered with fewer adverse cognitive
effects (Weiner et al. 1986). More recently, these devices have also
incorporated the use of the ultrabrief pulse stimulus, defined as a pulse
duration of less than 0.5 milliseconds, which is a more efficient way to
induce seizures (Loo et al. 2007) and is associated with less memory
impairment, although it may also be associated with a slightly more delayed
antidepressant effect (Sackeim et al. 2008). Regardless of stimulus
waveform type, however, the paradigm for the choice of stimulus dose
intensity dosing remains as controversial as the choice of electrode
placement. The disagreement centers on whether to dose with respect to an
empirically determined seizure threshold estimate obtained at the first
treatment (dose-titration technique) or to use a formula based on factors
such as age, gender, and electrode placement to make this decision
(formula-based technique) (American Psychiatric Association 2001).
Coffey et al. (1995) found that the dose-titration technique is better in
that it offers a more precise means to determine the patient’s seizure
threshold (which can vary manyfold) and thereby allows the practitioner to
more effectively control stimulus intensity. In practice, seizure threshold is
estimated at the first treatment by incrementally increasing the dose from a
low level until a seizure is induced.
Regardless of the dosing paradigm, compelling evidence suggests that
stimulus intensity for unilateral brief pulse ECT should be somewhere
between 2.5 and 8 times seizure threshold (in terms of electrical charge),
with the range reflecting uncertainty as to the minimum dose necessary to
optimize therapeutic outcome (McCall et al. 2000). In this regard, it is
important to note that increasing stimulus intensity also increases the
severity of ECT-associated memory impairment, although to a lesser degree
than a switch to bilateral ECT. Stimulus intensity is less of an issue with
bilateral brief pulse ECT, in which a stimulus 1.5 times seizure threshold
appears to be sufficient. However, stimulus intensity is of most concern
with ultrabrief pulse ECT, certainly for unilateral electrode placement but
likely also for bitemporal and bifrontal placement (Sackeim et al. 2008).
Although evidence-based guidance in this regard is presently limited, it is
recommended for ultrabrief pulse stimuli that a stimulus intensity 6–8 times
seizure threshold be used for unilateral ECT and 4–6 times seizure
threshold for bitemporal treatments.
As alluded to above (see “Anesthetic Considerations”), seizure threshold
is a function of age, with substantially higher thresholds being present in
older adults. This higher threshold leaves older adults at greater risk for
being unable to receive a stimulus of sufficient intensity, because the
maximum output of ECT devices used in the United States is limited by
FDA regulations (Krystal et al. 2000a). The risk that the threshold will
exceed the maximum available stimulus intensity is greatest late in the
treatment course, because seizure threshold rises to a varying extent with
the number of treatments.
Maintenance Therapy
The conditions for which ECT is used are typically recurrent. The risk of
relapse, particularly during the first 6 months, is extremely high (Tew et al.
2007), necessitating an aggressive program of maintenance treatment to
minimize the likelihood of relapse. This maintenance treatment may be
pharmacological or in the form of continued ECT (at a greatly lowered
frequency).
Maintenance ECT
The high relapse rate following ECT, even with pharmacological
maintenance therapy, has created renewed interest in the practice of
maintenance ECT. After a flurry of largely positive case-series reports
(Rabheru and Persad 1997), a randomized trial of maintenance ECT versus
pharmacotherapy in patients treated for major depression was carried out,
with results indicating that the efficacy of maintenance ECT over a 6-month
period was comparable to that obtained by combination pharmacotherapy
with both lithium and nortriptyline (Kellner et al. 2006).
Although there are no established guidelines for a maintenance ECT
regimen, practitioners typically start with weekly treatments for 2–4 weeks,
followed by another 1–2 months of biweekly treatments, followed by 3-
week and then 4-week intervals. After 12 months, treatments are either
stopped or continued at an even lower frequency. Although many
maintenance ECT patients do well with such a regimen, others appear to
require more frequent treatments or even supplementation with
psychotropic medications. Others eventually relapse, leading to another
index ECT course or a switch to alternative treatment modalities. On the
basis of data from a multicenter trial of maintenance ECT versus
maintenance pharmacotherapy, Lisanby et al. (2008) suggested use of a
symptom-based approach to maintenance ECT for patients who have
responded well to an index course of ECT for major depression. With this
approach, the patient is administered four weekly treatments followed by
close monitoring for at least 6 months; recurrence of depressive
symptomatology is followed by one or two ECT treatments during the
following week. A further multicenter trial incorporating this technique is
currently under way.
Key Points
• ECT is the most rapid and effective treatment for major depressive
episodes in older adults.
• Major risks of ECT are largely a function of medical comorbidity.
• An index ECT treatment course can be ended once a therapeutic plateau
has been reached.
• The risk for relapse after an acute course of ECT is high, and aggressive
maintenance treatment is needed.
• Other brain stimulation therapies for the treatment of depression include
transcranial magnetic stimulation, transcranial direct current stimulation,
vagus nerve stimulation, and deep brain stimulation.
• Most of the brain stimulation therapies have been studied primarily in
the general adult population, and data to support their efficacy in older
adults are limited.
• Future clinical trials are needed to test the benefit of these therapies in
the geriatric population.
References
Acharya D, Harper DG, Achtyes ED, et al: Safety and utility of acute
electroconvulsive therapy for agitation and aggression in dementia. Int
J Geriatr Psychiatry May 26, 2014 [Epub ahead of print] 24838521
American Psychiatric Association: The Practice of Electroconvulsive
Therapy: Recommendations for Treatment, Training, and Privileging.
Washington, DC, American Psychiatric Press, 2001
Applegate RJ: Diagnosis and management of ischemic heart disease in the
patient scheduled to undergo electroconvulsive therapy. Convuls Ther
13(3):128–144, 1997 9342129
Avery D, Lubrano A: Depression treated with imipramine and ECT: the
DeCarolis study reconsidered. Am J Psychiatry 136(4B): 559–562,
1979 426143
Aydogan MS, Yücel A, Begec Z, et al: The hemodynamic effects of
dexmedetomidine and esmolol in electroconvulsive therapy: a
retrospective comparison. J ECT 29(4):308–311, 2013 23774056
Bailine SH, Rifkin A, Kayne E, et al: Comparison of bifrontal and
bitemporal ECT for major depression. Am J Psychiatry 157(1): 121–
123, 2000 10618025
Berlim MT, Van den Eynde F, Daskalakis ZJ: Efficacy and acceptability of
high frequency repetitive transcranial magnetic stimulation (rTMS)
versus electroconvulsive therapy (ECT) for major depression: a
systematic review and meta-analysis of randomized trials. Depress
Anxiety 30(7): 614–623, 2013 23349112
Birkenhäger TK, Pluijms EM, Ju MR, et al: Influence of age on the efficacy
of electroconvulsive therapy in major depression: a retrospective study.
J Affect Disord 126(1–2):257–261, 2010 20303601
Bosboom PR, Deijen JB: Age-related cognitive effects of ECT and ECT-
induced mood improvement in depressive patients. Depress Anxiety
23(2):93–101, 2006 16400627
Brandon S: Efficacy of ECT in depression: controlled trials.
Psychopharmacol Bull 22:465–468, 1986
Brunoni AR, Ferrucci R, Fregni F, et al: Transcranial direct current
stimulation for the treatment of major depressive disorder: a summary
of preclinical, clinical and translational findings. Prog
Neuropsychopharmacol Biol Psychiatry 39(1):9–16, 2012 22651961
Chanpattana W, Sackeim HA, Fregni F, et al: Electroconvulsive therapy in
treatment-resistant schizophrenia: prediction of response and the nature
of symptomatic improvement. J ECT 26(4):289–298, 2010 20375701
Christopher EJ: Electroconvulsive therapy in the medically ill. Curr
Psychiatry Rep 5(3):225–230, 2003 12773277
Coffey CE, Lucke J, Weiner RD, et al: Seizure threshold in
electroconvulsive therapy: I. Initial seizure threshold. Biol Psychiatry
37(10):713–720, 1995 7640326
Currier MB, Murray GB, Welch CC: Electroconvulsive therapy for post-
stroke depressed geriatric patients. J Neuropsychiatry Clin Neurosci
4(2):140–144, 1992 1627974
Daban C, Martinez-Aran A, Cruz N, et al: Safety and efficacy of vagus
nerve stimulation in treatment-resistant depression. A systematic
review. J Affect Disord 110(1–2):1–15, 2008 18374988
Dierckx B, Heijnen WT, van den Broek WW, et al: Efficacy of
electroconvulsive therapy in bipolar versus unipolar major depression:
a meta-analysis. Bipolar Disord 14(2):146–150, 2012 22420590
Ding Z, White PF: Anesthesia for electroconvulsive therapy. Anesth Analg
94(5):1351–1364, 2002 11973219
Figiel GS, Coffey CE, Djang WT, et al: Brain magnetic resonance imaging
findings in ECT-induced delirium. J Neuropsychiatry Clin Neurosci
2(1):53–58, 1990 2136061
Figiel GS, Hassen MA, Zorumski C, et al: ECT-induced delirium in
depressed patients with Parkinson’s disease. J Neuropsychiatry Clin
Neurosci 3(4):405–411, 1991 1821261
Folkerts HW, Michael N, Tolle R, et al: Electroconvulsive therapy vs.
paroxetine in treatment-resistant depression—a randomized study. Acta
Psychiatr Scand 96(5): 334–342, 1997 9395150
Fregni F, Simon DK, Wu A, et al: Non-invasive brain stimulation for
Parkinson’s disease: a systematic review and meta-analysis of the
literature. J Neurol Neurosurg Psychiatry 76(12):1614–1623, 2005
16291882
Fricchione GL, Kaufman LD, Gruber BL, et al: Electroconvulsive therapy
and cyclophosphamide in combination for severe neuropsychiatric
lupus with catatonia. Am J Med 88(4):442–443, 1990 2327432
George MS, Lisanby SH, Avery D, et al: Daily left prefrontal transcranial
magnetic stimulation therapy for major depressive disorder: a sham-
controlled randomized trial. Arch Gen Psychiatry 67(5):507–516, 2010
20439832
Hood DD, Mecca RS: Failure to initiate electroconvulsive seizures in a
patient pretreated with lidocaine. Anesthesiology 58(4):379–381, 1983
6837980
Husain MM, Rush AJ, Fink M, et al: Speed of response and remission in
major depressive disorder with acute electroconvulsive therapy (ECT):
a Consortium for Research in ECT (CORE) report. J Clin Psychiatry
65(4):485–491, 2004 15119910
Huuhka M, Korpisammal L, Haataja R, et al: One-year outcome of elderly
inpatients with major depressive disorder treated with ECT and
antidepressants. J ECT 20(3):179–185, 2004 15343003
Janicak PG, Davis JM, Gibbons RD, et al: Efficacy of ECT: a meta-
analysis. Am J Psychiatry 142(3):297–302, 1985 3882006
Kamat SM, Lefevre PJ, Grossberg GT: Electroconvulsive therapy in the
elderly. Clin Geriatr Med 19(4):825–839, 2003 15024814
Kellner CH, Knapp RG, Petrides G, et al: Continuation electroconvulsive
therapy vs pharmacotherapy for relapse prevention in major
depression: a multisite study from the Consortium for Research in
Electroconvulsive Therapy (CORE). Arch Gen Psychiatry
63(12):1337–1344, 2006 17146008
Kellner CH, Knapp R, Husain MM, et al: Bifrontal, bitemporal and right
unilateral electrode placement in ECT: randomised trial. Br J
Psychiatry 196(3):226–234, 2010 20194546
Kerner N, Prudic J: Current electroconvulsive therapy practice and research
in the geriatric population. Neuropsychiatry (London) 4(1):33–54,
2014 24778709
Kho KH, Blansjaar BA, de Vries S, et al: Electroconvulsive therapy for the
treatment of clozapine nonresponders suffering from schizophrenia—
an open label study. Eur Arch Psychiatry Clin Neurosci 254(6): 372–
379, 2004 15538604
Klapheke MM: Combining ECT and antipsychotic agents: benefits and
risks. Convuls Ther 9(4):241–255, 1993 11941220
Krystal AD, Coffey CE: Neuropsychiatric considerations in the use of
electroconvulsive therapy. J Neuropsychiatry Clin Neurosci 9(2):283–
292, 1997 9144111
Krystal AD, Weiner RD, Coffey CE: The ictal EEG as a marker of adequate
stimulus intensity with unilateral ECT. J Neuropsychiatry Clin
Neurosci 7(3):295–303, 1995 7580187
Krystal AD, Watts BV, Weiner RD, et al: The use of flumazenil in the
anxious and benzodiazepine-dependent ECT patient. J ECT 14(1):5–
14, 1998 9661088
Krystal AD, Dean MD, Weiner RD, et al: ECT stimulus intensity: are
present ECT devices too limited? Am J Psychiatry 157(6): 963–967,
2000a 10831477
Krystal AD, Weiner RD, Lindahl V, et al: The development and
retrospective testing of an electroencephalographic seizure quality-
based stimulus dosing paradigm with ECT. J ECT 16(4):338–349,
2000b 11314871
Kuehn BM: Scientists probe deep brain stimulation: some promise for brain
injury, psychiatric illness. JAMA 298(19):2249–2251, 2007 18029821
Kujala I, Rosenvinge B, Bekkelund SI: Clinical outcome and adverse
effects of electroconvulsive therapy in elderly psychiatric patients. J
Geriatr Psychiatry Neurol 15(2): 73–76, 2002 12083596
Lisanby SH, Sampson S, Husain MM, et al: Toward individualized post-
electroconvulsive therapy care: piloting the Symptom-Titrated,
Algorithm-Based Longitudinal ECT (STABLE) intervention. J ECT
24(3):179–182, 2008 18708943
Loo C, Sheehan P, Pigot M, Lyndon W: A report on mood and cognitive
outcomes with right unilateral ultrabrief pulsewidth (0.3 ms) ECT and
retrospective comparison with standard pulsewidth right unilateral
ECT. J Affect Disord 103(1–3):277–281, 2007 17706790
Lozano AM, Mayberg HS, Giacobbe P, et al: Subcallosal cingulate gyrus
deep brain stimulation for treatment-resistant depression. Biol
Psychiatry 64(6):461–467, 2008 18639234
Manly DT, Oakley SP Jr, Bloch RM: Electroconvulsive therapy in old-old
patients. Am J Geriatr Psychiatry 8(3):232–236, 2000 10910422
Mayberg HS, Lozano AM, Voon V, et al: Deep brain stimulation for
treatment-resistant depression. Neuron 45(5):651–660, 2005 15748841
McCall WV, Reboussin DM, Weiner RD, Sackeim HA: Titrated moderately
suprathreshold vs fixed high-dose right unilateral electroconvulsive
therapy: acute antidepressant and cognitive effects. Arch Gen
Psychiatry 57(5):438–444, 2000 10807483
Murray GB, Shea V, Conn DK: Electroconvulsive therapy for poststroke
depression. J Clin Psychiatry 47:258–260, 1986 3700345
Nahas Z, Li X, Kozel FA, et al: Safety and benefits of distance-adjusted
prefrontal transcranial magnetic stimulation in depressed patients 55–
75 years of age: a pilot study. Depress Anxiety 19(4):249–256, 2004
15274174; correction in Depress Anxiety 27(1):90, 2010
Nuttall GA, Bowersox MR, Douglass SB, et al: Morbidity and mortality in
the use of electroconvulsive therapy. J ECT 20(4): 237–241, 2004
15591857
O’Connor DW, Gardner B, Eppingstall B, et al: Cognition in elderly
patients receiving unilateral and bilateral electroconvulsive therapy: a
prospective, naturalistic comparison. J Affect Disord 124(3):235–240,
2010 20053457
O’Connor MK, Knapp R, Husain M, et al: The influence of age on the
response of major depression to electroconvulsive therapy: a C.O.R.E.
Report. Am J Geriatr Psychiatry 9(4):382–390, 2001 11739064
O’Reardon JP, Solvason HB, Janicak PG, et al: Efficacy and safety of
transcranial magnetic stimulation in the acute treatment of major
depression: a multisite randomized controlled trial. Biol Psychiatry
62(11): 1208–1216, 2007 17573044
Paul SM, Extein I, Calil HM, et al: Use of ECT with treatment-resistant
depressed patients at the National Institute of Mental Health. Am J
Psychiatry 138:486–489, 1981 6111228
Petrides G, Fink M: Atrial fibrillation, anticoagulation, and
electroconvulsive therapy. Convuls Ther 12:91–98, 1996 8744168
Petrides G, Fink M, Husain MM, et al: ECT remission rates in psychotic
versus nonpsychotic depressed patients: a report from CORE. J ECT
17(4):244–253, 2001 11731725
Philibert RA, Richards L, Lynch CF, et al: Effect of ECT on mortality and
clinical outcome in geriatric unipolar depression. J Clin Psychiatry
56(9):390–394, 1995 7665536
Pompili M, Lester D, Dominici G, et al: Indications for electroconvulsive
treatment in schizophrenia: a systematic review. Schizophr Res 146(1–
3):1–9, 2013 23499244
Prudic J, Haskett RF, Mulsant B, et al: Resistance to antidepressant
medications and short-term clinical response to ECT. Am J Psychiatry
153:985–992, 1996 8678194
Quitkin FM, McGrath PJ, Stewart JW, et al: Can the effects of
antidepressants be observed in the first two weeks of treatment?
Neuropsychopharmacology 15(4):390–394, 1996 8887993
Rabheru K: The use of electroconvulsive therapy in special patient
populations. Can J Psychiatry 46(8):710–719, 2001 11692973
Rabheru K, Persad E: A review of continuation and maintenance
electroconvulsive therapy. Can J Psychiatry 42(5):476–484, 1997
9220110
Rasmussen KG, Varghese R, Stevens SR, et al: Electrode placement and
ictal EEG indices in electroconvulsive therapy. J Neuropsychiatry Clin
Neurosci 19(4):453–457, 2007 18070850
Rayburn BK: Electroconvulsive therapy in patients with heart failure or
valvular heart disease. Convuls Ther 13:145–156, 1997 9342130
Riva-Posse P, Hermida AP, McDonald WM: The role of electroconvulsive
and neuromodulation therapies in the treatment of geriatric depression.
Psychiatr Clin North Am 36(4):607–630, 2013 24229660
Rodez C, Trevino K, McClintock SM, et al: Advances in neurostimulation
therapies for depression, in Advances in Psychology Research, Volume
63. Edited by Columbus A. Hauppauge NY, Nova Science Publishers,
2009, pp 151–164
Rummans TA, Bassingthwaighte ME: Severe medical and neurologic
complications associated with near-lethal catatonia treated with
electroconvulsive therapy. Convuls Ther 7(2):121–124, 1991 11941111
Sackeim HA: The use of electroconvulsive therapy in late-life depression,
in Clinical Geriatric Psychopharmacology, 3rd Edition. Edited by
Salzman C. Baltimore, MD, Williams & Wilkins, 1998, pp 262–309
Sackeim HA, Rush AJ: Melancholia and response to ECT. Am J Psychiatry
152(8): 1242–1243, 1995 7625490
Sackeim HA, Prudic J, Devanand DP, et al: The impact of medication
resistance and continuation pharmacotherapy on relapse following
response to electroconvulsive therapy in major depression. J Clin
Psychopharmacol 10(2):96–104, 1990 2341598
Sackeim HA, Prudic J, Devanand DP, et al: Effects of stimulus intensity and
electrode placement on the efficacy and cognitive effects of
electroconvulsive therapy. N Engl J Med 328(12):839–846, 1993
8441428
Sackeim HA, Haskett RF, Mulsant BH, et al: Continuation
pharmacotherapy in the prevention of relapse following
electroconvulsive therapy: a randomized controlled trial. JAMA
285(10):1299–1307, 2001a 11255384
Sackeim HA, Rush AJ, George MS, et al: Vagus nerve stimulation (VNS)
for treatment-resistant depression: efficacy, side effects, and predictors
of outcome. Neuropsychopharmacology 25(5):713–728, 2001b
11682255
Sackeim HA, Prudic J, Fuller R, et al: The cognitive effects of
electroconvulsive therapy in community settings.
Neuropsychopharmacology 32(1):244–254, 2007 16936712
Sackeim HA, Prudic J, Nobler MS, et al: Effects of pulse width and
electrode placement on the efficacy and cognitive effects of
electroconvulsive therapy. Brain Stimulat 1(2):71–83, 2008 19756236
Saito S: Anesthesia management for electroconvulsive therapy:
hemodynamic and respiratory management. J Anesth 19(2): 142–149,
2005 15875132
Schlaepfer TE, Cohen MX, Frick C, et al: Deep brain stimulation to reward
circuitry alleviates anhedonia in refractory major depression.
Neuropsychopharmacology 33(2):368–377, 2008 17429407
Schlaepfer TE, Bewernick BH, Kayser S, et al: Deep brain stimulation of
the human reward system for major depression—rationale, outcomes
and outlook. Neuropsychopharmacology 39(6):1303–1314, 2014
24513970
Shiwach RS, Reid WH, Carmody TJ: An analysis of reported deaths
following electroconvulsive therapy in Texas, 1993–1998. Psychiatr
Serv 52(8):1095–1097, 2001 11474057
Shorter E, Healy D: Shock Therapy: A History of Electroconvulsive
Treatment in Mental Illness. New Brunswick, NJ, Rutgers University
Press, 2007
Sobin C, Prudic J, Devanand DP, et al: Who responds to electroconvulsive
therapy? A comparison of effective and ineffective forms of treatment.
Br J Psychiatry 169(3):322–328, 1996 8879718
Stern L, Hirschmann S, Grunhaus L: ECT in patients with major depressive
disorder and low cardiac output. Convuls Ther 13(2):68–73, 1997
9253526
Stoppe A, Louzã M, Rosa M, et al: Fixed high-dose electroconvulsive
therapy in the elderly with depression: a double-blind, randomized
comparison of efficacy and tolerability between unilateral and bilateral
electrode placement. J ECT 22(2):92–99, 2006 16801822
Takada JY, Solimene MC, da Luz PL, et al: Assessment of the
cardiovascular effects of electroconvulsive therapy in individuals older
than 50 years. Braz J Med Biol Res 38(9):1349–1357, 2005 16138218
Tess AV, Smetana GW: Medical evaluation of patients undergoing
electroconvulsive therapy. N Engl J Med 360(14):1437–1444, 2009
19339723
Texas Department of State Health Services: FY13 Summary of ECT use in
all facilities, 2014. Available at:
http://www.dshs.state.tx.us/mhsa/bhmd/ect/. Accessed April 6, 2014.
Tew JD Jr, Mulsant BH, Haskett RF, et al: Acute efficacy of ECT in the
treatment of major depression in the old-old. Am J Psychiatry
156(12):1865–1870, 1999 10588398
Tew JD Jr, Mulsant BH, Haskett RF, et al: Relapse during continuation
pharmacotherapy after acute response to ECT: a comparison of usual
care versus protocolized treatment. Ann Clin Psychiatry 19(1):1–4,
2007 17453654
Tielkes CE, Comijs HC, Verwijk E, et al: The effects of ECT on cognitive
functioning in the elderly: a review. Int J Geriatr Psychiatry 23(8):789–
795, 2008 18311845
Tomac TA, Rummans TA, Pileggi TS, et al: Safety and efficacy of
electroconvulsive therapy in patients over age 85. Am J Geriatr
Psychiatry 5(2):126–130, 1997 9106376
UK ECT Review Group: Efficacy and safety of electroconvulsive therapy
in depressive disorders: a systematic review and meta-analysis. Lancet
361(9360):799–808, 2003 12642045
van der Wurff FB, Stek ML, Hoogendijk WJ, et al: The efficacy and safety
of ECT in depressed older adults: a literature review. Int J Geriatr
Psychiatry 18(10):894–904, 2003 14533122
van Schaik AM, Comijs HC, Sonnenberg CM, et al: Efficacy and safety of
continuation and maintenance electroconvulsive therapy in depressed
elderly patients: a systematic review. Am J Geriatr Psychiatry 20(1):5–
17, 2012 22183009
Versiani M, Cheniaux E, Landeira-Fernandez J: Efficacy and safety of
electroconvulsive therapy in the treatment of bipolar disorder: a
systematic review. J ECT 27(2):153–164, 2011 20562714
Verwijk E, Comijs HC, Kok RM, et al: Short-and long-term neurocognitive
functioning after electroconvulsive therapy in depressed elderly: a
prospective naturalistic study. Int Psychogeriatr 26(2):315–324, 2014
24280446
Wani A, Trevino K, Marnell P, Hussain MM: Advances in brain stimulation
for depression. Ann Clin Psychiatry 25(3):217–224, 2013 23926577
Watts BV, Groft A, Bagian JP, et al: An examination of mortality and other
adverse events related to electroconvulsive therapy using a national
adverse event report system. J ECT 27(2):105–108, 2011 20966769
Weiner RD, Krystal AD: Electroconvulsive therapy, in Treatments of
Psychiatric Disorders, 3rd Edition. Edited by Gabbard GO, Rush AJ.
Washington, DC, American Psychiatric Press, 2001, pp 1267–1293
Weiner RD, Rogers HJ, Davidson JR, et al: Effects of stimulus parameters
on cognitive side effects. Ann N Y Acad Sci 462:315–325, 1986
3458412
Weiner RD, Coffey CE, Krystal AD: The monitoring and management of
electrically induced seizures. Psychiatr Clin North Am 14(4):845–869,
1991 1771151
Weiner RD, Coffey CE, Krystal AD: Electroconvulsive therapy in the
medical and neurologic patient, in Psychiatric Care of the Medical
Patient, 2nd Edition. Edited by Stoudemire A, Fogel BS, Greenberg D.
New York, Oxford University Press, 2000, pp 419–428
Wengel SP, Burke WJ, Pfeiffer RF, et al: Maintenance electroconvulsive
therapy for intractable Parkinson’s disease. Am J Geriatr Psychiatry
6(3):263–269, 1998 9659959
Wesson ML, Wilkinson AM, Anderson DN, et al: Does age predict the
long-term outcome of depression treated with ECT? (a prospective
study of the long-term outcome of ECT-treated depression with respect
to age). Int J Geriatr Psychiatry 12(1): 45–51, 1997 9050423
Zielinski RJ, Roose SP, Devanand DP, et al: Cardiovascular complications
of ECT in depressed patients with cardiac disease. Am J Psychiatry
150(6):904–909, 1993 8494067
Suggested Readings
Abrams R: Electroconvulsive Therapy, 4th Edition. New York, Oxford
University Press, 2002
American Psychiatric Association: The Practice of ECT: Recommendations
for Treatment, Training, and Privileging. Washington, DC, American
Psychiatric Press, 2001
Dukakis K, Tye L: Shock: The Healing Power of Electroconvulsive
Therapy. New York, Avery, 2006
Endler NS: Holiday of Darkness, Revised. Toronto, ON, Wall & Davis,
1990
Fink M: Electroconvulsive Therapy: A Guide for Professionals and Their
Patients, 2nd Edition. New York, Oxford University Press, 2010
Higgins ES, George MS: Brain Stimulation Therapies for Clinicians.
Washington, DC, American Psychiatric Publishing, 2009
Mankad MV, Beyer JL, Weiner RD, et al: Clinical Manual of
Electroconvulsive Therapy. Washington, DC, American Psychiatric
Publishing, 2010
Manning M: Undercurrents: A Life Beneath the Surface. San Francisco,
CA, Harper Collins, 1995
Reti IM: Brain Stimulation: Methodologies and Interventions. New York,
Wiley, 2015
Shorter E, Healy D: Shock Therapy: A History of Electroconvulsive
Treatment in Mental Illness. New Brunswick, NJ, Rutgers University
Press, 2007
CHAPTER 22
When patients present with mental difficulties for the first time, the
clinician needs to consider a diverse laundry list of possible nutritional
concerns. Although the effects of nutritional deficits might be transitory, the
symptoms can be disabling and are likely to obscure any other underlying
mechanisms of impairment. Chronic inadequate food intake is a concern
and can lead to weakness, fatigue, and other vague symptoms due to
negative energy balance. Certain vitamin deficiencies can have cognitive
components, as we address in the later subsection “Vitamins.” Dehydration
is an important contributor to delirium and confusion in older adults, who
are known to have a diminished thirst response compared with younger
adults (Luckey and Parsa 2003). Lack of compliance with therapeutic
dietary restrictions and instructions can also contribute to poor mental
status. For example, when diabetic control is poor, the acute cognitive
effects of both hyperglycemia and hypoglycemia can be marked. Poor
glycemic control can also contribute to poor cognitive outcomes in the long
term (Ravona-Springer et al. 2012; West et al. 2014). Gregg et al. (2000)
used repeated clinical neuropsychological testing to evaluate the
relationship of diabetes and disease duration to cognitive function in a large
population-based study of older women and found an increased risk of
cognitive decline in individuals with diabetes mellitus.
Alcohol
Although there are distinct disadvantages to advocating regular alcohol
consumption for adults of any age, there is a considerable amount of
evidence linking modest to moderate alcohol intake (no more than one daily
drink for women and two daily drinks for men) with benefits related to
preservation of cognitive function. A longitudinal study of 5,033 older men
and women assessed cognitive performance over 7 years and associated
light to moderate wine consumption with beneficial effects on cognitive
performance compared with low or no alcohol intake (Arntzen et al. 2010).
Weyerer et al. (2011) found that light to moderate alcohol consumption was
associated with significant reduction in the incidence of dementia and
Alzheimer’s disease in 3,202 men and women ages 75 years and older.
Furthermore, in a meta-analysis of 74 studies of older adults, Neafsey and
Collins (2011) found that compared with abstinence, moderate alcohol
consumption reduced risk of overall dementia by 23%, Alzheimer’s disease
by 27%, and vascular dementia by 34%. Despite these benefits, the
protective effect of moderate alcohol intake was diminished in individuals
with the allele producing the ε4 type of apolipoprotein E (APOE*E4)
(Panza et al. 2012).
It remains to be determined whether alcohol has direct benefits to
cognition or whether alcohol use is associated with other factors (e.g.,
physiological, cultural) that promote these benefits; however, a number of
potential mechanisms linking alcohol consumption and cognitive
consumption have been proposed. Moderate alcohol intake has been found
to reduce brain infarcts and white matter lesions and to reduce vascular risk
by increasing prostacyclin concentrations, reducing generation of
thromboxane A2, inhibiting platelet function, and increasing high-density
lipoprotein cholesterol levels (Frisardi et al. 2010; Panza et al. 2012).
Additionally, polyphenols in wine provide antioxidant effects that may be
protective for cognitive decline.
Vitamins
Epidemiological studies, including longitudinal observations, have been
conducted to examine the relationships between vitamin intake and incident
dementia. The most commonly studied vitamins are folate and vitamin B12,
the deficiencies of which have been linked with symptoms of dementia in
older adults (Clarke 2007). Two longitudinal studies of adults older than age
65 years who were dementia free at baseline looked at predictors of incident
dementia over 2–6 years. Intakes of folate at baseline (assessed by a food
frequency questionnaire) and serum folate levels were associated with a
decreased risk of dementia, including Alzheimer’s disease, independent of
other risk factors (Kim et al. 2008a; Luchsinger et al. 2007). However,
Cochrane reviews for folate (Malouf and Grimley Evans 2008) and for
vitamin B12 (Malouf and Areosa Sastre 2003) have not supported a
cognitive benefit for these nutrients, and there are other such findings in the
literature (Eussen et al. 2006; Paulionis et al. 2005). The inconsistencies
across the folate literature may relate to the relatively more beneficial
influence of naturally occurring folate than of supplemental folic acid, and
to the correlation of folate-rich foods with other beneficial foods such as
fruits and vegetables. Regarding vitamin B12, the varied findings may
indicate that only deficient states promote dementia.
A study by Przybelski and Binkley (2007) suggested that vitamin D has
a role in cognitive function. Other studies have linked low plasma
antioxidant levels with cognitive impairment (N=589 elderly subjects)
(Akbaraly et al. 2007) and brain white matter hyperintense lesions (N=355
subjects ages 45–75 years) (Schmidt et al. 1996). It is likely that other
nutrients will also emerge as candidates for promoting optimal cognitive
status. Already, some investigators have tried interventions using nutritional
supplements to enhance cognitive function in the elderly, with limited
success (Wouters-Wesseling et al. 2005). This lack of success is consistent
with general lack of support for nutritional supplements for promoting
health in a healthy population, and the need for a food-based approach for
prevention of dementia. It is therefore not surprising that dietary patterns
have been shown to influence risk of cognitive decline. The available
scientific evidence supports a Mediterranean dietary pattern for the
prevention of late-life cognitive decline and dementia (Lourida et al. 2013),
rather than a focus on promoting single nutrients.
Diet may promote (or prevent) depression by influencing one’s risk of vascular diseases, including
atherosclerosis and diabetes. These vascular diseases are known to promote late-life depression. In
addition, diet may directly promote (or prevent) depression by altering neuronal health or
neurotransmitter levels. Potential moderators of both pathways include ischemic brain lesions and
inflammation. CRP=C-reactive protein.
The inconsistent findings regarding depression risk and individual
nutrients or foods have led some researchers to focus on dietary patterns.
This is a reasonable approach because nutrients and foods vary together in
the diet. The Mediterranean diet has received the most attention, and studies
have yielded promising results; greater adherence to this dietary pattern has
been found to decrease incident risk of depression (Sánchez-Villegas et al.
2009; Skarupski et al. 2013). A meta-analysis examined the role of the
Mediterranean diet in preventing mental and cognitive disorders
(Psaltopoulou et al. 2013). Higher adherence to a Mediterranean diet was
found to reduce risk not only for depression (relative risk [RR]=0.68; 95%
confidence interval [CI]=0.54–0.86), but also for stroke (RR=0.71; 95%
CI=0.57–0.89) and cognitive impairment (RR=0.60; 95% CI= 0.43–0.83).
Mood Disorders
Mood disorders, including major depression and bipolar illness, and
situational conditions, such as bereavement and living alone, can
significantly affect health behaviors, including diet and physical activity.
Depression among older adults is often characterized by reduced appetite
and weight loss (Kolasa et al. 1995; Morley 1996; Pirlich and Lochs 2001;
Reife 1995). In fact, many geriatricians consider depression to be the most
common cause of poor food intake and nutritional frailty among elderly
individuals (Morley 2001). The loneliness and loss experienced by many
elderly individuals affect both mood and dietary status, contributing to the
anorexia and malnutrition of aging (Ferry et al. 2005). Locher et al. (2005)
conducted in-depth interviews with isolated elderly individuals and reported
that when eating alone, they consumed on average 114 fewer calories per
meal than when someone joined them at mealtime. In a French survey of
150 adults ages 80 years and older who lived alone, Ferry et al. (2005)
documented high rates of undernutrition. In these situations, inadequate
nutrient intakes may lead to muscle loss, an increased risk of bone fractures,
and nursing home admission (Christensen and Somers 1994).
Although depression can interfere with food intake by affecting general
interest in daily activities, there are also possible physiological mechanisms
whereby depression could lead to decreased appetite and weight loss. One
potential explanation is that corticotrophin-releasing factor (CRF), a potent
anorectic agent known to be elevated in depressed individuals, may depress
appetite (Morley 1996). Some have blamed antidepressants for the weight
loss associated with depression; however, this relationship was observed
long before the advent of psychotropic medications. In addition, only a
subset of antidepressants is known to cause weight loss.
Patients who suffer from atypical depression may experience weight gain
as a result of increased appetite and inactivity. Obesity may develop or be
exacerbated in this situation. In addition to changes in quantity of food
consumed by individuals with depression, food quality may decline.
Carbohydrate-rich foods lacking in micronutrients (i.e., food of low nutrient
density) may be preferred by depressed individuals (Wurtman and Wurtman
1996). Elderly individuals with depression have higher intakes of saturated
fat and cholesterol than do those without depression (Payne et al. 2006). A
lack of energy may curtail healthy eating in depressed people; for example,
they may choose to eat fast food rather than spend the time preparing
healthier meals at home.
With regard to physical activity, the loss of energy and motivation that
may accompany depression naturally makes efforts to exercise extremely
difficult. At the other end of the spectrum, some people with depression,
bipolar disorder, or other mood disorders experience agitation, an increase
in energy, or excessive activity.
Alcoholism
Alcoholism is a condition with considerable impact on both mental and
physical health because it occurs at the intersection of nutritional imbalance
and mental disorders. People with alcoholism often consume inadequate
amounts of vitamins and minerals and may have an insufficient intake of
protein. Not only does ethanol displace healthier dietary components, but
behavioral manifestations of alcoholism may hinder consumption of a
nutritionally adequate diet. In addition, alcoholic individuals may develop
deficiencies of certain nutrients (e.g., folate and other B vitamins) due to
malabsorption (Halsted et al. 2002). Chronic alcohol misuse can lead to
severe vitamin depletion and the development of Wernicke-Korsakoff
syndrome in susceptible individuals (Thomson 2000).
In a study assessing the prevalence of alcohol consumption in older
adults, Wilson et al. (2014) found that 14.5% of older individuals consumed
more than the National Institute on Alcohol Abuse and Alcoholism’s
recommended limit. However, when health status was taken into account
using the Alcohol-Related Problems Survey, 53.3% (95% CI=0.50–0.57) of
older adult drinkers were categorized as having harmful or hazardous
alcohol consumption (Wilson et al. 2014). Alcohol can enhance or negate
medication effects and lead to adverse drug events, impaired psychomotor
function, and sedation in older adults; therefore, it is important that
clinicians also assess heath status while investigating alcohol consumption
(Moore et al. 2007; Wilson et al. 2014).
Drug-Nutrient Interactions
Use of medications, including polypharmacy, is higher among elderly
individuals than among younger adults. This greater use, combined with the
physiological changes of aging, contributes to drug-related problems
including drug-nutrient interactions (Knight-Klimas and Boullata 2004).
The poor nutritional state that is common in elderly individuals can
exacerbate drug-nutrient interactions. A serious example of such an
interaction is a hypertensive crisis resulting from consumption of tyramine-
containing foods during monoamine oxidase inhibitor (MAOI) therapy
(McCabe 1986). Although addressing the list of possible drug-nutrient
interactions that may occur with psychotropic medications is beyond the
scope of this chapter, a general approach is recommended: Clinicians
should 1) consider the patient’s nutritional status (including malnutrition,
nutrient deficiencies, and body weight) and current diet (including intake of
vitamin, mineral, and herbal dietary supplements), 2) evaluate the potential
for known drug-nutrient interactions for the medication being considered
(which can be obtained from a drug-nutrient interaction reference book or a
pharmacist), and 3) discontinue the offending agent and/or institute
appropriate dietary modifications.
References. (1) Vellas B, Villars H, Abellan G, et al: Overview of the MNA—its history and
challenges. J Nutr Health Aging 10(6):456–463, 2006; (2) Rubenstein LZ, Harker JO, Salvà A, et al:
Screening for undernutrition in geriatric practice: developing the short-form mini-nutritional
assessment (MNA-SF). J Gerontol A Biol Sci Med Sci 56(6):M366–M372, 2001; (3) Guigoz Y: The
Mini Nutritional Assessment (MNA) review of the literature—What does it tell us? J Nutr Health
Aging 10(6):466–485, 2006; (4) Kaiser MJ, Bauer JM, Ramsch C, et al: Validation of the Mini
Nutritional Assessment short-form (MNA-SF): a practical tool for identification of nutritional status.
J Nutr Health Aging 13(9):782–788, 2009.
MNA® is a registered trademark of Société des Produits Nestlé, S.A., Vevey, Switzerland.
© Nestlé, 1994, Revision 2009. N67200 12/99 10M. For more information: www.mna-elderly.com.
General Nutrition Recommendations
Men:
TDEE=864–(9.72×age)+
[PA×(14.2×weight+503×height)]
Women:
TDEE=387–(7.31×age)+
[PA×(10.9×weight+660.7×height)]
Energy requirements decline with age because of reductions in metabolic
rate, loss of lean body mass, and a diminution of energy expenditure for
physical activity. A progressive reduction in TDEE occurs with age,
assumed to be about 7 and 10 kcal per year for adult women and men,
respectively. Although food and calorie (energy) intakes also decrease with
age, before age 60 years there is rarely a detrimental effect, because the
reduced energy intake is more than offset by the aforementioned
decrements in requirements. In fact, being overweight and having excess
adiposity pose serious health risks in middle and early old age and persist as
late-life concerns for some individuals (Porter Starr et al. 2014; Villareal et
al. 2005). Typically, however, energy deficits and poor nutrient intakes are
the most likely nutritional concerns in late life. As people age from their
20s through their 80s, mean energy intakes are reduced by up to 1,200 kcal
in men and up to 800 kcal in women, according to the Third National
Health and Nutrition Examination Survey (Wakimoto and Block 2001). It is
well recognized that inadequate energy intakes and low body mass indices
(BMIs) are linked with frailty in elderly individuals (Markson 1997). Poor
food intake also jeopardizes the adequacy of micronutrients; food choices
may be poor when appetites falter.
TABLE 22–1. Dietary reference intakes for adults ages 51 years and
older
Men Women
Nutrient 51–70 years >70 years 51–70 years >70 years
Proteina (g) 56 56 46 46
Carbohydrate 130 130 130 130
(g)
Fiber (total)b 30 30 21 21
(g)
Fatc (g)
Linoleic 14 14 11 11
acidb
α-Linolenic 1.6 1.6 1.1 1.1
acidb
data. AIs are given in this table for essential fatty acids.
cAdequate Intakes (AIs) represent the recommended average daily intake
The current RDA for protein (4 kcal/g) is 0.80 g/kg/day and is the same
for adults of all ages. Protein intakes decrease with age; however, even
though most people’s protein intake usually exceeds the RDA, there are
indications that older adults may need more dietary protein than do younger
adults to support good health, promote recovery from illness, and maintain
functionality (Bauer et al. 2013). Protein insufficiency can be a particular
concern in some high-risk individuals, especially when experiencing the
stress of medical illness and when institutionalization occurs.
Fat is another macronutrient that provides energy and is in fact the major
fuel source for the body (providing 9 kcal/g). In addition, fat is needed for
the absorption of fat-soluble vitamins and other dietary components. As
discussed earlier in “Type of Fat and Intake of Fish,” dietary fats are
divided into four general categories: saturated, trans-unsaturated,
monounsaturated, and polyunsaturated, with the latter subdivided into
omega-6 and omega-3 fats. Only certain fatty acids must be consumed from
the diet (essential fatty acids)—namely, two of the long-chain
polyunsaturated fatty acids: linolenic (an omega-3) and linoleic (an omega-
6). Saturated fatty acids, monounsaturated fatty acids, and cholesterol can
be synthesized by the body. Trans fats are not needed by the body and are,
in fact, detrimental to health. Older adults consume substantially less than
the RDA of fat, as well as of cholesterol. The percentage of calories coming
from fat in the diet declines and reaches its lowest value in the oldest age
groups (Wakimoto and Block 2001).
Carbohydrates are categorized into starches and sugars
(monosaccharides and disaccharides) and are also an important energy
source for the body (providing 4 kcal/g). A minimum amount of
carbohydrate (130 g for adults) must be consumed daily to meet needs that
cannot be met by another fuel source (such as fat). Glucose is the only fuel
source used by erythrocytes and is the preferred energy source for the brain,
which accounts for 20% of the body’s resting metabolic rate (Siegel 1999).
In the absence of sufficient dietary carbohydrate, liver glycogen and
skeletal muscle are broken down to generate glucose.
In recent years, the popularity of low-carbohydrate diets for weight loss
(Yancy et al. 2004) and findings regarding the benefits of protein for satiety
have renewed interest in manipulations of macronutrient proportions in the
diet. In fact, a number of different distributions of macronutrients (e.g.,
Mediterranean diet pattern, vegetarian diet) other than the more traditional
low-fat diet can be used to achieve a healthy dietary pattern (Malik and Hu
2007).
Although alcohol does yield calories (7 kcal/g), recommendations about
alcohol for older adults are difficult to reconcile based on the available
scientific evidence. On the one hand, the intake of moderate amounts of
alcohol by older adults is linked with decreased risk of cardiovascular
disease (thought to be caused by decreased inflammation and increased
high-density lipoprotein levels) (Mukamal et al. 2006), risk of ischemic
stroke (except in APOE*E4-positive individuals) (Mukamal et al. 2005),
bone density of the hip (Mukamal et al. 2007), and even dementia
(Mukamal et al. 2003). On the other hand, the issue of alcohol misuse and
abuse among the elderly is a concern, particularly when any kind of
cognitive impairment is present. Therefore, it is difficult to apply these
research findings given the inadvisability of promoting ethanol
consumption in the elderly because of concerns about falls and alcohol
abuse (see earlier section “Alcoholism”). However, if a patient already
consumes light to moderate amounts of alcohol on a regular basis and is not
having problems, it may be acceptable for this level of consumption to
continue and it may be beneficial to suggest consumption of red wine, the
alcoholic beverage that contains the highest levels of beneficial
phytochemical compounds.
Benefits of Exercise
Physical activity is essential for optimal health throughout the life cycle,
providing protection against a diverse array of diseases and disorders linked
with aging, including cardiovascular disease, thromboembolic stroke,
diabetes mellitus, osteoporosis, certain cancers, anxiety, and depression
(Chodzko-Zajko et al. 2009; Nelson et al. 2007). Prevention of the risk of
falls and associated injuries is another key benefit. Physical activity
preserves muscle mass and strength as well as bone mineral density and has
been linked with improved mood and sleep patterns.
Initiating a fitness program to increase activity levels in a sedentary
individual requires an exercise assessment and subsequent individualization
of the exercise prescription to avoid injuries to the musculoskeletal or
cardiovascular system. Physical limitations must be identified, specific
exercises selected, and exertion ranges established. The American College
of Sports Medicine (2013) provides detailed guidance for fitness
assessments and exercise prescriptions. Additionally, the U.S. National
Institute on Aging has established Go4Life (http://go4life.nia.nih.gov), an
exercise and physical activity campaign targeted toward increasing exercise
in older adults. Ongoing reassessment will allow the program to be
upgraded in response to progress by the participant. It should be stressed
that even for individuals with physical limitations or chronic health
conditions, some level of physical activity is almost always beneficial.
• Balance activities. Many older adults are at risk of falls, and to reduce the
ensuing risk of injury, these adults should participate in activity that
focuses on maintaining or improving balance. The important components
of balance training are exercises that challenge balance (i.e., backward
walking, sideways walking) and lower-body strength exercises (i.e., chair
stands). T’ai Chi exercises are also a great way to increase balance and
reduce the risk of falls.
• Activities to increase flexibility. Flexibility is important in everyday
physical activity, and older adults should spend at least 10 minutes, 2 days
each week, on exercises that will maintain or increase flexibility.
• Developing and implementing an activity plan. Older adults seeking to
reach the recommended levels of physical activity should implement a
plan of gradual increase in exercise over time. It is acceptable to spend
time with activity that is lower than the recommended amount as long as
there is a stepwise increase toward sufficiency. Each recommended type
of activity, as well as how, when, and where such exercise should occur,
must be addressed in the plan, and older adults should self-monitor and
reevaluate their individual activity plans as physical ability and health
status vary.
Conclusion
Diet and physical activity are important determinants of both physical and
mental health. Although the specific mechanisms by which these lifestyle
parameters affect mental function are not fully understood, there is no
question that healthy dietary intakes and a physically active lifestyle
promote and help sustain optimal cognitive function. The guidelines
presented in this chapter can benefit all patients and should be a part of the
regular care plan. To assist patients in setting and achieving realistic
behavioral goals, the physician should partner with other professionals,
including registered dietitians, speech-language pathologists, dentists,
pharmacists, physical and occupational therapists, and other specialists, to
individualize diet and physical activity prescriptions.
Key Points
• Nutrition and physical activity are important determinants of overall
health and also play an important role in key elements of mental health
and cognitive status.
• Although a number of lifestyle factors have been implicated in the
etiology of mental disorders, the available scientific evidence does not
support a recommendation of specific nutrients or foods for the prevention
of dementia. However, adherence to a Mediterranean dietary pattern may
promote mental and brain health.
• Because late-life depression is known to promote vascular disease,
depressed individuals should be encouraged to consume a diet that will
reduce their vascular risk.
• Regular physical activity appears to benefit cognitive health and is
therapeutic for depression and dementia in terms of function and quality
of life.
• Mental disorders that can negatively affect nutritional and physical fitness
status include depression, dementia, and other mental illnesses; treatments
for these conditions may also impact nutrition and activity patterns.
• Older adults should consume a varied diet that includes high-quality
protein, fruits, and vegetables, and provides the recommended amounts of
all essential nutrients and meets but does not exceed energy requirements.
References
Acree LS, Longfors J, Fjeldstad AS, et al: Physical activity is related to
quality of life in older adults. Health Qual Life Outcomes 4:37, 2006
16813655
Adams PB, Lawson S, Sanigorski A, et al: Arachidonic acid to
eicosapentaenoic acid ratio in blood correlates positively with clinical
symptoms of depression. Lipids 31 (suppl):S157–S161, 1996 8729112
Age-Related Eye Disease Study Research Group: A randomized, placebo-
controlled, clinical trial of high-dose supplementation with vitamins C
and E, beta carotene, and zinc for age-related macular degeneration and
vision loss: AREDS report no 8. Arch Ophthalmol 119(10):1417–1436,
2001 11594942
Akbaraly NT, Faure H, Gourlet V, et al: Plasma carotenoid levels and
cognitive performance in an elderly population: results of the EVA
Study. J Gerontol A Biol Sci Med Sci 62(3):308–316, 2007 17389729
Almeida OP, Norman P, Hankey G, et al: Successful mental health aging:
results from a longitudinal study of older Australian men. Am J Geriatr
Psychiatry 14(1):27–35, 2006 16407579
Almeida OP, Calver J, Jamrozik K, et al: Obesity and metabolic syndrome
increase the risk of incident depression in older men: the health in men
study. Am J Geriatr Psychiatry 17(10):889–898, 2009 19910877
Alpert M, Silva RR, Pouget ER: Prediction of treatment response in
geriatric depression from baseline folate level: interaction with an SSRI
or a tricyclic antidepressant. J Clin Psychopharmacol 23(3):309–313,
2003 12826993
American College of Sports Medicine: ACSM’s Guidelines for Exercise
Testing and Prescription. Baltimore, MD, Lippincott Williams &
Wilkins, 2013
American Geriatrics Society Panel on Exercise and Osteoarthritis: Exercise
prescription for older adults with osteoarthritis pain: consensus practice
recommendations. A supplement to the AGS Clinical Practice
Guidelines on the management of chronic pain in older adults. J Am
Geriatr Soc 49(6):808–823, 2001 11480416
Andrès E, Loukili NH, Noel E, et al: Vitamin B12 (cobalamin) deficiency
in elderly patients. CMAJ 171(3):251–259, 2004 15289425
Angevaren M, Aufdemkampe G, Verhaar HJ, et al: Physical activity and
enhanced fitness to improve cognitive function in older people without
known cognitive impairment. Cochrane Database Syst Rev
(3):CD005381, 2008
Arntzen KA, Schirmer H, Wilsgaard T, et al: Moderate wine consumption is
associated with better cognitive test results: a 7 year follow up of 5033
subjects in the Tromsø Study. Acta Neurol Scand Suppl (190):23–29,
2010 20586731
Aselage M, Amella EJ, Rose SB, et al: Dementia-related mealtime
difficulties: assessment and management in the long-term care setting,
in Handbook of Clinical Nutrition and Aging. Edited by Bales CW,
Locher JL, Saltzman ES. New York, Springer, 2015, pp 287–301
Balanzá-Martínez V, Fries GR, Colpo GD, et al: Therapeutic use of omega-
3 fatty acids in bipolar disorder. Expert Rev Neurother 11(7):1029–
1047, 2011 21721919
Bales CW, Johnson MA: Nutrition for older adults, in Modern Nutrition in
Health and Disease. Edited by Ross C, Caballero B, Cousins R, et al.
Philadelphia, PA, Lippincott Williams & Wilkins, 2012, pp 744–756
Bardia A, Tleyjeh IM, Cerhan JR, et al: Efficacy of antioxidant
supplementation in reducing primary cancer incidence and mortality:
systematic review and meta-analysis. Mayo Clin Proc 83(1):23–34,
2008 18173999
Barrocas A, Bistrian BR, Blackburn GL, et al: Appropriate and effective
use of the NSI checklist and screens. An update on caring for the
elderly by preventing malnutrition. J Am Diet Assoc 95(6):647–648,
1995 7759737
Bauer JM, Kaiser MJ, Sieber CC: Evaluation of nutritional status in older
persons: nutritional screening and assessment. Curr Opin Clin Nutr
Metab Care 13(1):8–13, 2010 19851099
Bauer J, Biolo G, Cederholm T, et al: Evidence-based recommendations for
optimal dietary protein intake in older people: a position paper from the
PROTAGE Study Group. J Am Med Dir Assoc 14(8):542–559, 2013
23867520
Bell IR, Edman JS, Morrow FD, et al: B complex vitamin patterns in
geriatric and young adult inpatients with major depression. J Am
Geriatr Soc 39(3):252–257, 1991 2005338
Berke EM, Gottlieb LM, Moudon AV, et al: Protective association between
neighborhood walkability and depression in older men. J Am Geriatr
Soc 55(4):526–533, 2007 17397430
Bjelland I, Tell GS, Vollset SE, et al: Folate, vitamin B12, homocysteine,
and the MTHFR 677C->T polymorphism in anxiety and depression:
the Hordaland Homocysteine Study. Arch Gen Psychiatry 60(6):618–
626, 2003 12796225
Blaum CS, Fries BE, Fiatarone MA: Factors associated with low body mass
index and weight loss in nursing home residents. J Gerontol A Biol Sci
Med Sci 50(3):M162–M168, 1995 7743402
Blumenthal JA, Babyak MA, Moore KA, et al: Effects of exercise training
on older patients with major depression. Arch Intern Med
159(19):2349–2356, 1999 10547175
Bonner FJ Jr, Sinaki M, Grabois M, et al: Health professional’s guide to
rehabilitation of the patient with osteoporosis. Osteoporos Int 14 (suppl
2):S1–S22, 2003 12759719
Bridle C, Spanjers K, Patel S, et al: Effect of exercise on depression
severity in older people: systematic review and meta-analysis of
randomised controlled trials. Br J Psychiatry 201(3):180–185, 2012
22945926
Brynningsen PK, Damsgaard EM, Husted SE: Improved nutritional status
in elderly patients 6 months after stroke. J Nutr Health Aging 11(1):75–
79, 2007 17315085
Buhr G, Bales CW: Nutritional supplements for older adults: review and
recommendations—part I. J Nutr Elder 28(1):5–29, 2009 19234992
Buhr G, Bales CW: Nutritional supplements for older adults: review and
recommendations—part II. J Nutr Elder 29(1):42–71, 2010 20391042
Carlsson E, Ehrenberg A, Ehnfors M: Stroke and eating difficulties: long-
term experiences. J Clin Nurs 13(7):825–834, 2004 15361156
Centers for Disease Control and Prevention: Adult participation in aerobic
and muscle-strengthening physical activity—United States, 2011.
MMWR Morb Mortal Wkly Rep 62(17):326–330, 2013 23636025
Chernoff R: Micronutrient requirements in older women. Am J Clin Nutr
81(5): 1240S–1245S, 2005 15883458
Chodzko-Zajko WJ, Proctor DN, Fiatarone Singh MA, et al; American
College of Sports Medicine: American College of Sports Medicine
position stand. Exercise and physical activity for older adults. Med Sci
Sports Exerc 41(7):1510–1530, 2009 19516148
Christensen L, Somers S: Adequacy of the dietary intake of depressed
individuals. J Am Coll Nutr 13(6):597–600, 1994 7706593
Cimrová B, Budác S, Melicherová U, et al: Electrophysiological evidence
of the effect of natural polyphenols upon the human higher brain
functions. Neuroendocrinol Lett 32(4):464–468, 2011 21876511
Clarke R: Homocysteine, B vitamins, and the risk of dementia. Am J Clin
Nutr 85(2): 329–330, 2007 17284725
Clemens R, Kranz S, Mobley AR, et al: Filling America’s fiber intake gap:
summary of a roundtable to probe realistic solutions with a focus on
grain-based foods. J Nutr 142(7):1390S–1401S, 2012 22649260
Colcombe SJ, Erickson KI, Scalf PE, et al: Aerobic exercise training
increases brain volume in aging humans. J Gerontol A Biol Sci Med
Sci 61(11):1166–1170, 2006 17167157
Concannon LG, Grierson MJ, Harrast MA: Exercise in the older adult: from
the sedentary elderly to the masters athlete. PM R 4(11):833–839, 2012
23174546
Cuijpers P, van Straten A, Bohlmeijer E, et al: The effects of psychotherapy
for adult depression are overestimated: a meta-analysis of study quality
and effect size. Psychol Med 40(2):211–223, 2010 19490745
Detsky AS, Smalley PS, Chang J: The rational clinical examination. Is this
patient malnourished? JAMA 271(1):54–58, 1994 8258889
Donini LM, Savina C, Rosano A, et al: MNA predictive value in the
follow-up of geriatric patients. J Nutr Health Aging 7(5):282–293,
2003 12917741
Ebbing M, Bønaa KH, Nygård O, et al: Cancer incidence and mortality
after treatment with folic acid and vitamin B12. JAMA 302(19):2119–
2126, 2009 19920236
Eussen SJ, Ferry M, Hininger I, et al: Five year changes in mental health
and associations with vitamin B12/folate status of elderly Europeans. J
Nutr Health Aging 6(1):43–50, 2002 11813081
Eussen SJ, de Groot LC, Joosten LW, et al: Effect of oral vitamin B-12 with
or without folic acid on cognitive function in older people with mild
vitamin B-12 deficiency: a randomized, placebo-controlled trial. Am J
Clin Nutr 84(2):361–370, 2006 16895884
Ferry M, Sidobre B, Lambertin A, et al: The SOLINUT study: analysis of
the interaction between nutrition and loneliness in persons aged over 70
years. J Nutr Health Aging 9(4):261–268, 2005 15980927
Fiske A, Wetherell JL, Gatz M: Depression in older adults. Annu Rev Clin
Psychol 5: 363–389, 2009 19327033
Franklin BA: Preventing exercise-related cardiovascular events: is a
medical examination more urgent for physical activity or inactivity?
Circulation 129(10):1081–1084, 2014 24421369
Franklin B, Whaley M, Howley E: ACSM’s Guidelines for Exercise Testing
and Prescription, 6th Edition. Philadelphia, PA, Lippincott Williams &
Wilkins, 2000, pp 137–164
Frisardi V, Solfrizzi V, Imbimbo PB, et al: Towards disease-modifying
treatment of Alzheimer’s disease: drugs targeting beta-amyloid. Curr
Alzheimer Res 7(1):40–55, 2010 19939231
Gariepy G, Wang J, Lesage AD, Schmitz N: The longitudinal association
from obesity to depression: results from the 12-year National
Population Health Survey. Obesity (Silver Spring) 18(5):1033–1038,
2009 19816409
Gaziano JM, Sesso HD, Christen WG, et al: Multivitamins in the
prevention of cancer in men: the Physicians’ Health Study II
randomized controlled trial. JAMA 308(18):1871–1880, 2012
23162860
Ghadirian AM, Ananth J, Engelsmann F: Folic acid deficiency and
depression. Psychosomatics 21(11):926–929, 1980 7433596
Gregg EW, Yaffe K, Cauley JA, et al: Is diabetes associated with cognitive
impairment and cognitive decline among older women? Study of
Osteoporotic Fractures Research Group. Arch Intern Med 160(2):174–
180, 2000 10647755
Hakkarainen R, Partonen T, Haukka J, et al: Food and nutrient intake in
relation to mental wellbeing. Nutr J 3:14, 2004a 15363099
Hakkarainen R, Partonen T, Haukka J, et al: Is low dietary intake of omega-
3 fatty acids associated with depression? Am J Psychiatry 161(3):567–
569, 2004b 14992986
Halsted CH, Villanueva JA, Devlin AM, et al: Metabolic interactions of
alcohol and folate. J Nutr 132 (8 suppl):2367S–2372S, 2002 12163694
Hamer M, Lavoie KL, Bacon SL: Taking up physical activity in later life
and healthy ageing: the English Longitudinal Study of Ageing. Br J
Sports Med 48(3):239–243, 2014 24276781
Hartman CA, Manos TM, Winter C, et al: Effects of T’ai Chi training on
function and quality of life indicators in older adults with osteoarthritis.
J Am Geriatr Soc 48(12):1553–1559, 2000 11129742
Hibbeln JR: Fish consumption and major depression. Lancet
351(9110):1213, 1998 9643729
Hootman JM, Macera CA, Ainsworth BE, et al: Epidemiology of
musculoskeletal injuries among sedentary and physically active adults.
Med Sci Sports Exerc 34(5):838–844, 2002 11984303
Hunter R, Jones M, Jones TG, Matthews DM: Serum B12 and folate
concentrations in mental patients. Br J Psychiatry 113:1291–1295,
1967
Institute of Medicine: Dietary Reference Intakes for Calcium, Phosphorus,
Magnesium, Vitamin D, and Fluoride. Washington, DC, National
Academy Press, 1997
Institute of Medicine: Dietary Reference Intakes for Thiamin, Riboflavin,
Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and
Choline. Washington, DC, National Academy Press, 1998
Institute of Medicine: Dietary Reference Intakes for Vitamin C, E,
Selenium, and Carotenoids. Washington, DC, National Academy Press,
2000
Institute of Medicine: Dietary Reference Intakes for Energy, Carbohydrate,
Fiber, Fat, Fatty Acids, Cholesterol, Protein, and Amino Acids.
Washington, DC, National Academy Press, 2005
Institute of Medicine: Dietary Reference Intakes for Calcium and Vitamin
D. Washington, DC, National Academy Press, 2010
Kaiser MJ, Bauer JM, Ramsch C, et al: Validation of the Mini Nutritional
Assessment short-form (MNA-SF): a practical tool for identification of
nutritional status. J Nutr Health Aging 13(9):782–788, 2009 19812868
Kasper S, Anghelescu IG, Szegedi A, et al: Superior efficacy of St John’s
wort extract WS 5570 compared to placebo in patients with major
depression: a randomized, double-blind, placebo-controlled,
multicenter trial [ISRCTN77277298]. BMC Med 4:14, 2006 16796730
Kayser-Jones J: Preventable causes of dehydration: nursing home residents
are especially vulnerable. Am J Nurs 106(6):45, 2006 16728844
Kim JM, Stewart R, Kim SW, et al: Changes in folate, vitamin B12 and
homocysteine associated with incident dementia. J Neurol Neurosurg
Psychiatry 79(8):864–868, 2008a 18252751
Kim JM, Stewart R, Kim SW, et al: Predictive value of folate, vitamin B12
and homocysteine levels in late-life depression. Br J Psychiatry
192(4):268–274, 2008b 18378986
Knight-Klimas TC, Boullata JI: Drug-nutrient interactions in the elderly, in
Handbook of Drug-Nutrient Interactions, Edited by Boullata JI,
Armenti VT. Totowa, NJ, Humana Press, 2004, pp 363–410
Kolasa KM, Mitchell JP, Jobe AC: Food behaviors of southern rural
community-living elderly. Arch Fam Med 4(10):844–848, 1995
7551131
Kwakkel G, Kollen B, Twisk J: Impact of time on improvement of outcome
after stroke. Stroke 37(9):2348–2353, 2006 16931787
Lampinen P, Heikkinen RL, Kauppinen M, et al: Activity as a predictor of
mental well-being among older adults. Aging Ment Health 10(5):454–
466, 2006 16938681
Larson EB, Wang L, Bowen JD, et al: Exercise is associated with reduced
risk for incident dementia among persons 65 years of age and older.
Ann Intern Med 144(2):73–81, 2006 16418406
Lau FC, Shukitt-Hale B, Joseph JA: Nutritional intervention in brain aging:
reducing the effects of inflammation and oxidative stress. Subcell
Biochem 42:299–318, 2007 17612057
Li K, Kaaks R, Linseisen J, et al: Associations of dietary calcium intake and
calcium supplementation with myocardial infarction and stroke risk
and overall cardiovascular mortality in the Heidelberg cohort of the
European Prospective Investigation into Cancer and Nutrition study
(EPIC-Heidelberg). Heart 98(12): 920–925, 2012 22626900
Lindeman RD, Romero LJ, Koehler KM, et al: Serum vitamin B12, C and
folate concentrations in the New Mexico elder health survey:
correlations with cognitive and affective functions. J Am Coll Nutr
19(1):68–76, 2000a 10682878
Lindeman RD, Romero LJ, Liang HC, et al: Do elderly persons need to be
encouraged to drink more fluids? J Gerontol A Biol Sci Med Sci
55(7):M361–M365, 2000b 10898251
Locher JL, Robinson CO, Roth DL, et al: The effect of the presence of
others on caloric intake in homebound older adults. J Gerontol A Biol
Sci Med Sci 60(11):1475–1478, 2005 16339337
Lourida I, Soni M, Thompson-Coon J, et al: Mediterranean diet, cognitive
function, and dementia: a systematic review. Epidemiology 24(4):479–
489, 2013 23680940
Luchsinger JA, Tang MX, Miller J, et al: Relation of higher folate intake to
lower risk of Alzheimer disease in the elderly. Arch Neurol 64(1):86–
92, 2007 17210813
Luckey AE, Parsa CJ: Fluid and electrolytes in the aged. Arch Surg
138(10):1055–1060, 2003 14557120
Maes M, Christophe A, Delanghe J, et al: Lowered omega3 polyunsaturated
fatty acids in serum phospholipids and cholesteryl esters of depressed
patients. Psychiatry Res 85(3):275–291, 1999 10333380
Malik VS, Hu FB: Popular weight-loss diets: from evidence to practice. Nat
Clin Pract Cardiovasc Med 4(1):34–41, 2007 17180148
Malouf R, Areosa Sastre A: Vitamin B12 for cognition. Cochrane Database
Syst Rev (3):CD004326, 2003 12918012
Malouf R, Grimley Evans J: Folic acid with or without vitamin B12 for the
prevention and treatment of healthy elderly and demented people.
Cochrane Database Syst Rev (4):CD004514, 2008 18843658
Marano G, Traversi G, Nannarelli C, et al: Omega-3 fatty acids and
schizophrenia: evidences and recommendations. Clin Ter 164(6):e529–
e537, 2013 24424237
Marks BL, Madden DJ, Bucur B, et al: Role of aerobic fitness and aging on
cerebral white matter integrity. Ann NY Acad Sci 1097:171–174, 2007
17413020
Markson EW: Functional, social, and psychological disability as causes of
loss of weight and independence in older community-living people.
Clin Geriatr Med 13(4):639–652, 1997 9354746
Martineau J, Bauer JD, Isenring E, et al: Malnutrition determined by the
patient-generated subjective global assessment is associated with poor
outcomes in acute stroke patients. Clin Nutr 24(6):1073–1077, 2005
16213064
McCabe BJ: Dietary tyramine and other pressor amines in MAOI regimens:
a review. J Am Diet Assoc 86(8):1059–1064, 1986 3525654
Michaëlsson K, Melhus H, Warensjö Lemming E, et al: Long term calcium
intake and rates of all cause and cardiovascular mortality: community
based prospective longitudinal cohort study. BMJ 346:f228, 2013
23403980
Miller ER 3rd, Juraschek S, Pastor-Barriuso R, et al: Meta-analysis of folic
acid supplementation trials on risk of cardiovascular disease and risk
interaction with baseline homocysteine levels. Am J Cardiol 106(4):
517–527, 2010 20691310
Mitchell SL, Teno JM, Roy J, et al: Clinical and organizational factors
associated with feeding tube use among nursing home residents with
advanced cognitive impairment. JAMA 290(1):73–80, 2003 12837714
Moore AA, Whiteman EJ, Ward KT: Risks of combined alcohol/medication
use in older adults. Am J Geriatr Pharmacother 5(1):64–74, 2007
17608249
Morley JE: Anorexia in older persons: epidemiology and optimal treatment.
Drugs Aging 8(2):134–155, 1996 8845587
Morley J: Water, water everywhere and not a drop to drink. J Gerontol A
Biol Sci Med Sci 55(7):M359–M360, 2000 10898250
Morley JE: Decreased food intake with aging. J Gerontol A Biol Sci Med
Sci 56(Spec No 2):81–88, 2001 11730241
Morris MC, Evans DA, Tangney CC, et al: Fish consumption and cognitive
decline with age in a large community study. Arch Neurol
62(12):1849–1853, 2005 16216930
Moshfegh A, Goldman J, Ahuja J, et al: What We Eat in America,
NHANES 2005–2006: Usual Nutrient Intakes From Food and Water
Compared to 1997 Dietary Reference Intakes for Vitamin D, Calcium,
Phosphorus, and Magnesium. Beltsville, MD, U.S. Department of
Agriculture, Agricultural Research Service, 2009. Available at:
http://www.ars.usda.gov/SP2UserFiles/Place/12355000/pdf/0506/usual
_nutrient_intake_vitD_ca_phos_mg_2005-06.pdf. Accessed November
18, 2014.
Mozaffarian D, Rimm EB: Fish intake, contaminants, and human health:
evaluating the risks and the benefits. JAMA 296(15): 1885–1899, 2006
17047219
Mukamal KJ, Kuller LH, Fitzpatrick AL, et al: Prospective study of alcohol
consumption and risk of dementia in older adults. JAMA
289(11):1405–1413, 2003 12636463
Mukamal KJ, Chung H, Jenny NS, et al: Alcohol use and risk of ischemic
stroke among older adults: the cardiovascular health study. Stroke
36(9):1830–1834, 2005 16081863
Mukamal KJ, Chung H, Jenny NS, et al: Alcohol consumption and risk of
coronary heart disease in older adults: the Cardiovascular Health Study.
J Am Geriatr Soc 54(1):30–37, 2006 16420195
Mukamal KJ, Robbins JA, Cauley JA, et al: Alcohol consumption, bone
density, and hip fracture among older adults: the cardiovascular health
study. Osteoporos Int 18(5):593–602, 2007 17318666
National Osteoporosis Foundation: Clinician’s Guide to Prevention and
Treatment of Osteoporosis. Washington, DC, National Osteoporosis
Foundation, 2013, pp 1–55
Neafsey EJ, Collins MA: Moderate alcohol consumption and cognitive risk.
Neuropsychiatr Dis Treat 7:465–484, 2011 21857787
Nelson ME, Rejeski WJ, Blair SN, et al: Physical activity and public health
in older adults: recommendation from the American College of Sports
Medicine and the American Heart Association. Med Sci Sports Exerc
39(8):1435–1445, 2007 17762378
Ness AR, Gallacher JE, Bennett PD, et al: Advice to eat fish and mood: a
randomised controlled trial in men with angina. Nutr Neurosci 6(1):63–
65, 2003 12608739
Newman AB, Yanez D, Harris T, et al; Cardiovascular Study Research
Group: Weight change in old age and its association with mortality. J
Am Geriatr Soc 49(10):1309–1318, 2001 11890489
Niedert KC; American Dietetic Association: Position of the American
Dietetic Association: Liberalization of the diet prescription improves
quality of life for older adults in long-term care. J Am Diet Assoc
105(12):1955–1965, 2005 16402447
Omran ML, Morley JE: Assessment of protein energy malnutrition in older
persons, part II: laboratory evaluation. Nutrition 16(2):131–140, 2000
10696638
Osher Y, Bersudsky Y, Belmaker RH: Omega-3 eicosapentaenoic acid in
bipolar depression: report of a small open-label study. J Clin Psychiatry
66(6):726–729, 2005 15960565
Panza F, Frisardi V, Seripa D, et al: Alcohol consumption in mild cognitive
impairment and dementia: harmful or neuroprotective? Int J Geriatr
Psychiatry 27(12): 1218–1238, 2012 22396249
Papakostas GI, Shelton RC, Zajecka JM, et al: L-methylfolate as adjunctive
therapy for SSRI-resistant major depression: results of two
randomized, double-blind, parallel-sequential trials. Am J Psychiatry
169(12): 1267–1274, 2012 23212058
Parrott MD, Shatenstein B, Ferland G, et al: Relationship between diet
quality and cognition depends on socioeconomic position in healthy
older adults. J Nutr 143(11): 1767–1773, 2013 23986363
Paulionis L, Kane SL, Meckling KA: Vitamin status and cognitive function
in a long-term care population. BMC Geriatr 5:16, 2005 16351716
Payne ME, Hybels CF, Bales CW, et al: Vascular nutritional correlates of
late-life depression. Am J Geriatr Psychiatry 14(9): 787–795, 2006
16943175
Payne ME, Jamerson BD, Potocky CF, et al: Natural food folate and late-
life depression. J Nutr Elder 28(4):348–358, 2009 21184377
Penninx BW, Guralnik JM, Ferrucci L, et al: Vitamin B(12) deficiency and
depression in physically disabled older women: epidemiologic
evidence from the Women’s Health and Aging Study. Am J Psychiatry
157(5):715–721, 2000 10784463
Perry L, McLaren S: Nutritional support in acute stroke: the impact of
evidence-based guidelines. Clin Nutr 22(3):283–293, 2003 12765669
Pirlich M, Lochs H: Nutrition in the elderly. Best Pract Res Clin
Gastroenterol 15(6): 869–884, 2001 11866482
Porter Starr KN, McDonald SR, Bales CW: Obesity and physical frailty in
older adults: a scoping review of lifestyle intervention trials. J Am Med
Dir Assoc 15(4):240–250, 2014 24445063
Powell KE, Paluch AE, Blair SN: Physical activity for health: What kind?
How much? How intense? On top of what? Annu Rev Public Health
32:349–365, 2011 21128761
Przybelski RJ, Binkley NC: Is vitamin D important for preserving
cognition? A positive correlation of serum 25-hydroxyvitamin D
concentration with cognitive function. Arch Biochem Biophys
460(2):202–205, 2007 17258168
Psaltopoulou T, Sergentanis TN, Panagiotakos DB, et al: Mediterranean
diet, stroke, cognitive impairment, and depression: A meta-analysis.
Ann Neurol 74(4):580–591, 2013 23720230
Ramos MI, Allen LH, Haan MN, et al: Plasma folate concentrations are
associated with depressive symptoms in elderly Latina women despite
folic acid fortification. Am J Clin Nutr 80(4):1024–1028, 2004
15447915
Ravona-Springer R, Moshier E, Schmeidler J, et al: Changes in glycemic
control are associated with changes in cognition in non-diabetic elderly.
J Alzheimers Dis 30(2): 299–309, 2012 22426020
Reife CM: Involuntary weight loss. Med Clin North Am 79(2):299–313,
1995 7877392
Roberts RE, Deleger S, Strawbridge WJ, et al: Prospective association
between obesity and depression: evidence from the Alameda County
Study. Int J Obes Relat Metab Disord 27(4):514–521, 2003 12664085
Rock CL: Multivitamin-multimineral supplements: who uses them? Am J
Clin Nutr 85(1):277S–279S, 2007 17209209
Rolland Y, Pillard F, Klapouszczak A, et al: Exercise program for nursing
home residents with Alzheimer’s disease: a 1-year randomized,
controlled trial. J Am Geriatr Soc 55(2):158–165, 2007 17302650
Sachdev PS, Parslow RA, Lux O, et al: Relationship of homocysteine, folic
acid and vitamin B12 with depression in a middle-aged community
sample. Psychol Med 35(4):529–538, 2005 15856723
Sacks GS, Dearman K, Replogle WH, et al: Use of subjective global
assessment to identify nutrition-associated complications and death in
geriatric long-term care facility residents. J Am Coll Nutr 19(5):570–
577, 2000 11022870
Sánchez-Villegas A, Delgado-Rodríguez M, Alonso A, et al: Association of
the Mediterranean dietary pattern with the incidence of depression: the
Seguimiento Universidad de Navarra/University of Navarra follow-up
(SUN) cohort. Arch Gen Psychiatry 66(10):1090–1098, 2009
19805699
Scarmeas N, Stern Y, Tang MX, et al: Mediterranean diet and risk for
Alzheimer’s disease. Ann Neurol 59(6):912–921, 2006 16622828
Schmidt R, Hayn M, Fazekas F, et al: Magnetic resonance imaging white
matter hyperintensities in clinically normal elderly individuals.
Correlations with plasma concentrations of naturally occurring
antioxidants. Stroke 27(11):2043–2047, 1996 8898813
Siegel GJ: Basic Neurochemistry: Molecular, Cellular, and Medical
Aspects, 6th Edition. Philadelphia, PA, Lippincott-Raven, 1999
Silvers KM, Scott KM: Fish consumption and self-reported physical and
mental health status. Public Health Nutr 5(3):427–431, 2002 12003654
Skarupski KA, Tangney CC, Li H, et al: Mediterranean diet and depressive
symptoms among older adults over time. J Nutr Health Aging
17(5):441–445, 2013 23636545
Sofi F, Valecchi D, Bacci D, et al: Physical activity and risk of cognitive
decline: a meta-analysis of prospective studies. J Intern Med
269(1):107–117, 2011 20831630
Solfrizzi V, Colacicco AM, D’Introno A, et al: Dietary intake of unsaturated
fatty acids and age-related cognitive decline: a 8.5-year follow-up of
the Italian Longitudinal Study on Aging. Neurobiol Aging
27(11):1694–1704, 2006 16256248
Stewart JW, Harrison W, Quitkin F, et al: Low B6 levels in depressed
outpatients. Biol Psychiatry 19(4):613–616, 1984 6733177
Sullivan DH, Bopp MM, Roberson PK: Protein-energy undernutrition and
life-threatening complications among the hospitalized elderly. J Gen
Intern Med 17(12):923–932, 2002 12472928
Tanskanen A, Hibbeln JR, Hintikka J, et al: Fish consumption, depression,
and suicidality in a general population. Arch Gen Psychiatry
58(5):512–513, 2001 11343534
Taylor D, Meader N, Bird V, et al; pharmacology subgroup of the National
Institute for Health and Clinical Excellence Guideline Development
Group for Depression in Chronic Physical Health Problems:
Pharmacological interventions for people with depression and chronic
physical health problems: systematic review and meta-analyses of
safety and efficacy. Br J Psychiatry 198(3):179–188, 2011 21357876
Thombs BD, Bass EB, Ford DE, et al: Prevalence of depression in survivors
of acute myocardial infarction. J Gen Intern Med 21(1):30–38, 2006
16423120
Thomson AD: Mechanisms of vitamin deficiency in chronic alcohol
misusers and the development of the Wernicke-Korsakoff syndrome.
Alcohol Alcohol Suppl 35(1): 2–7, 2000 11304071
Tiemeier H, van Tuijl HR, Hofman A, et al: Vitamin B12, folate, and
homocysteine in depression: the Rotterdam Study. Am J Psychiatry
159(12):2099–2101, 2002 12450964
Tolmunen T, Hintikka J, Ruusunen A, et al: Dietary folate and the risk of
depression in Finnish middle-aged men. A prospective follow-up study.
Psychother Psychosom 73(6):334–339, 2004 15479987
Tolonen M, Schrijver J, Westermarck T, et al: Vitamin B6 status of Finnish
elderly. Comparison with Dutch younger adults and elderly. The effect
of supplementation. Int J Vitam Nutr Res 58(1):73–77, 1988 3384588
U.S. Department of Health and Human Services: Physical Activity
Guidelines for Americans. Atlanta, GA, National Center for Chronic
Disease and Health Promotion, 2008. Available at:
http://health.gov/paguidelines/guidelines/. Accessed December 6, 2014.
van de Rest O, Geleijnse JM, Kok FJ, et al: Effect of fish oil on cognitive
performance in older subjects: a randomized, controlled trial.
Neurology 71(6):430–438, 2008 18678826
Villareal DT, Apovian CM, Kushner RF, Klein S; American Society for
Nutrition; NAASO, The Obesity Society: Obesity in older adults:
technical review and position statement of the American Society for
Nutrition and NAASO, The Obesity Society. Am J Clin Nutr
82(5):923–934, 2005 16280421
Vreeland B: Bridging the gap between mental and physical health: a
multidisciplinary approach. J Clin Psychiatry 68 (suppl 4): 26–33, 2007
17539697
Wakimoto P, Block G: Dietary intake, dietary patterns, and changes with
age: an epidemiological perspective. J Gerontol A Biol Sci Med Sci
56(Spec No 2):65–80, 2001 11730239
Wellman NS: Prevention, prevention, prevention: nutrition for successful
aging. J Am Diet Assoc 107(5):741–743, 2007 17467368
West RK, Ravona-Springer R, Schmeidler J, et al: The association of
duration of type 2 diabetes with cognitive performance is modulated by
long-term glycemic control. Am J Geriatr Psychiatry 22(10):1055–
1059, 2014 24534521
Weyerer S, Schäufele M, Wiese B, et al; German AgeCoDe Study group
(German Study on Ageing, Cognition and Dementia in Primary Care
Patients): Current alcohol consumption and its relationship to incident
dementia: results from a 3-year follow-up study among primary care
attenders aged 75 years and older. Age Ageing 40(4):456–463, 2011
21367764
White H, Pieper C, Schmader K, et al: Weight change in Alzheimer’s
disease. J Am Geriatr Soc 44(3):265–272, 1996 8600194
White H, Pieper C, Schmader K: The association of weight change in
Alzheimer’s disease with severity of disease and mortality: a
longitudinal analysis. J Am Geriatr Soc 46(10):1223–1227, 1998
9777903
Williams MA, Haskell WL, Ades PA, et al; American Heart Association
Council on Clinical Cardiology; American Heart Association Council
on Nutrition, Physical Activity, and Metabolism: Resistance exercise in
individuals with and without cardiovascular disease: 2007 update: a
scientific statement from the American Heart Association Council on
Clinical Cardiology and Council on Nutrition, Physical Activity, and
Metabolism. Circulation 116(5):572–584, 2007 17638929
Wilson SR, Knowles SB, Huang Q, et al: The prevalence of harmful and
hazardous alcohol consumption in older U.S. adults: data from the
2005–2008 National Health and Nutrition Examination Survey
(NHANES). J Gen Intern Med 29(2):312–319, 2014 24101531
Wouters-Wesseling W, Wagenaar LW, Rozendaal M, et al: Effect of an
enriched drink on cognitive function in frail elderly persons. J Gerontol
A Biol Sci Med Sci 60(2):265–270, 2005 15814873
Wurtman RJ, Wurtman JJ: Brain Serotonin, Carbohydrate-craving, obesity
and depression. Adv Exp Med Biol 398:35–41, 1996 9045545
Yancy WSJr, Olsen MK, Guyton JR, et al: A low-carbohydrate, ketogenic
diet versus a low-fat diet to treat obesity and hyperlipidemia: a
randomized, controlled trial. Ann Intern Med 140(10):769–777, 2004
15148063
Depressive Disorders
Interpersonal Psychotherapy
Interpersonal psychotherapy (IPT) focuses on four common psychosocial
problems related to depression: loss and grief, interpersonal disputes and
conflicts, role transitions, and interpersonal skill deficits. IPT consists of an
assessment phase, treatment phase, maintenance, and relapse prevention
phase (Miller 2008). IPT for older adults is very similar to IPT for younger
adult populations. The initial assessment phase of IPT consists of collecting
an interpersonal inventory that helps the therapist and patient identify the
primary focus of treatment. Once the focus is identified, two important
techniques are used to help the patient move past the problem:
communication analysis and differentiating content affect and process affect
(Stuart and Noyes 2006). Communication analysis involves asking the
client to describe the last time he or she encountered the interpersonal
problem and discussing the specific details of that problem (e.g., what was
actually said in the encounter and how the client felt). After the causes of
the interpersonal problem are identified, the patient develops new
communication skills to express his or her needs and affect. These skills are
strengthened through in-session role-play and between-session practice of
these new communication skills. During this process, the therapist works
with the patient to identify feelings while discussing the incident in therapy
(process affect) and the feelings the client experiences outside of the
therapy session (content affect).
IPT in the general adult population is a very effective treatment;
however, the research on IPT in older adults is mixed, owing to a relatively
small data base and the fact that larger trials addressed prevention of
recurrence of depression in those at high risk for recurrence (Areán and
Cook 2002). IPT has been found to be very promising as an acute treatment
in older adults with depression (Miller et al. 2001, 2003; Reynolds et al.
1999). However, as a prevention intervention, the results are not as positive
(Carreira et al. 2008; Reynolds et al. 2010). The majority of studies on IPT
for late-life depression have been done in academic medicine; there has
only been one of primary care–based treatment for older adults (Post et al.
2008), and the specific effects of IPT have not been published (Bruce et al.
2004).
Relaxation Training
Relaxation training may be one of the most frequently used treatments for
anxiety in older adults. Work by DeBerry (1981–1982, 1982; DeBerry et al.
1989) showed that progressive muscle relaxation and meditation relaxation
techniques reduced anxiety symptoms more effectively than treatment
control conditions in older adults. Scogin et al. (1992) assessed the use of
progressive muscle relaxation and imaginal relaxation. General symptom
improvements were maintained at 1-month follow-up, and gains in
treatment responders were maintained at a 1-year follow-up assessment
(Rickard et al. 1994). Relaxation training has some advantages for treating
mild anxiety in older adults. The strategies can be taught in brief individual
or group sessions. Theoretically, the strategies can be delivered during a
regular visit to a primary care physician. As with many behavioral
strategies, relaxation training has the advantage of masquerading as skills
training for patients who might avoid traditional psychotherapy. Also,
patients with cognitive deficits, who may have difficulty with more
cognitive-based strategies, may benefit from purely behavioral strategies.
One caveat is that relaxation training may be counterproductive when used
in combination with exposure-based treatments, because intentional
relaxation during exposures may provide temporary relief but interfere with
long-term learning to tolerate anxiety (Abramowitz 2013).
Substance-Related Disorders
With the baby boom generation entering late life, a rise is expected in the
number of older adults who use alcohol and illicit substances because of the
higher rates of substance use in that age cohort. Since 2002 the rate of illicit
drug use more than doubled (from 1.9% to 4.1%) among adults ages 55–59
years (Substance Abuse and Mental Health Services Administration 2011).
Sadly, there has been very little focus on the development of treatments
specifically for older adults, despite the fact that a number of studies
indicate that substance use disorder treatment needs to be age specific for
the treatment to be acceptable to older adults (Schonfeld et al. 2000; Satre
et al. 2003, 2004).
Cognitive-Behavioral Treatment
Although some previous comprehensive cognitive-behavioral interventions
have shown promise for treating substance abuse in older adults, they are
also plagued with high dropout rates (Schonfeld et al. 2000). Schonfeld et
al. (2010) assessed the effectiveness of the Florida Brief Intervention and
Treatment for Elders (BRITE) project—a low-cost approach for older
adults at risk for illicit and nonillicit substance abuse and misuse.
Prescription medication misuse and alcohol abuse were most prevalent in
the sample. The treatment protocol involved a brief home-based
intervention of one to five sessions that included motivational interviewing,
education about relevant substances and consequences of substance use and
misuse, reasons to quit drinking, and medication management. Following
graduation from the brief intervention, participants completed a 16-session
cognitive-behavioral treatment. The BRITE approach resulted in a
significant reduction of depression and suicide risk, as well as alcohol and
prescription medication misuse (Schonfeld et al. 2010).
Personality Disorders
Cognitive-Behavioral Therapies
Personality disorders are enduring patterns of inner experience (e.g.,
cognition, affect, impulse control) and behavior (e.g., interpersonal
difficulties) that have an onset in adolescence or early adulthood, are stable
over time, deviate considerably from normal cultural expectations, and
cause distress or impairment in functioning. Meta-analyses have concluded
that the prevalence rate for personality disorders in older adults is between
10% and 20% (Abrams 1996; Abrams and Horowitz 1999), essentially
analogous to the 13% prevalence rate for younger age groups (Torgersen et
al. 2001). Overall, the emotionally constricted/risk-averse disorders in
Clusters A (paranoid and schizoid personality disorders) and C (obsessive-
compulsive, avoidant, and dependent personality disorders) are the most
commonly diagnosed in late life (Abrams 1996; Abrams and Horowitz
1999; Kenan et al. 2000; Morse and Lynch 2004), and there are also high
rates of diagnosis of personality disorder not otherwise specified (mapping
to the categories of other specified personality disorder or unspecified
personality disorder in DSM-5 [American Psychiatric Association 2013])
compared with individual personality disorder diagnoses (Abrams 1996;
Abrams and Horowitz 1999; Kenan et al. 2000). In addition, personality
disorder rates are even higher (approximately 30%) among depressed older
adult samples (Abrams 1996; Thompson et al. 1988).
Personality psychopathology has generally been associated with poorer
response to treatment among older adults, whether treated with
antidepressants or psychotherapy (Abrams et al. 1994; Lynch et al. 2007;
Thompson et al. 1988; Zweig 2008; but see Gum et al. 2007; also see
Gradman et al. 1999for a review). Depressed older adult patients with
comorbid personality disorder are four times more likely to experience
maintenance or reemergence of depressive symptoms than are those without
personality disorder diagnoses (Morse and Lynch 2004). Despite this, and
despite calls for greater attention to the development and improvement of
interventions for personality disorders in late life (e.g., Videler et al. 2012),
there are few controlled studies of the treatment of personality disorders in
older adults.
The only published randomized clinical trial specifically targeting
personality disorders in older adults was conducted by Lynch et al. (2007).
The study focused on providing standard DBT using both group and
individual sessions following Linehan’s (1993) manual. Participants were
depressed older adults who presented with at least one comorbid personality
disorder and who failed an 8-week SSRI trial. Compared with patients in a
medication-only condition, participants who received DBT plus medication
management reached the level of remission more quickly and showed
improvements in interpersonal sensitivity and aggression (Lynch et al.
2007). Adaptations of standard DBT focusing on the most common older
adult personality disorders (e.g., paranoid personality disorder, obsessive-
compulsive personality disorder) are in development, with treatment targets
that include reducing rigidity, cognitive inflexibility, emotional constriction,
and risk aversion (Lynch and Cheavens 2008).
Despite the promising nature of these findings, the bulk of empirical
evidence suggests that the presence of a personality disorder in an older
adult seriously compromises treatment. In addition, rates of personality
disorders among older adults may be only slightly lower than those in
younger age groups, and subsyndromal personality disorders may be more
prevalent in older populations relative to younger ones (Abrams and
Bromberg 2006). It appears that psychotherapy interventions likely will be
enhanced when they target the unique behavioral, cognitive, and
interpersonal dynamics associated with older-adult personality disorders.
Conclusion
It is evident that psychotherapy offers significant promise for the treatment
of psychopathology in elderly persons and at times may be the treatment of
choice in terms of both efficacy and patient preference. We encourage
practitioners to select treatments that have been tested with randomized
clinical trials rather than basing their choices on theoretical preference or
ease of application. The use of treatments without this type of empirical
support can slow or reduce recovery.
Future research should continue to examine the beneficial effects of
strategies combining medication and psychotherapy. In addition, research
examining the mechanisms of change and issues associated with treatment
response by disorder and type of therapy remain to be more fully
developed. Finally, continued research is needed to focus on populations
with treatment-resistant illnesses such as personality disorders, substance
dependence, and comorbid disorders.
Key Points
• Psychotherapy is a good option for treating mental disorders in older
adults who have trouble tolerating medications, who prefer
psychotherapy over medication treatment, or who have conditions for
which psychotherapy is the most effective treatment.
• Modifications of traditional therapies may be necessary to compensate
for age-related problems with vision, hearing, mobility, and cognition.
• Effective treatments for depression include several different individual
and group cognitive-behavioral therapies, interpersonal psychotherapy,
and short-term psychodynamic therapy.
• The most common anxiety disorder among older adults is generalized
anxiety disorder. Behavioral treatments such as relaxation training and
cognitive-behavioral therapy are the most effective treatments for this
disorder.
• Patients with personality pathology have poorer response to treatment of
other comorbid conditions such as depression and anxiety. Few
treatments targeting personality disorder in older adults have been tested.
One promising and empirically validated treatment is a modification of
dialectical behavior therapy for older adults.
• Although cognitive impairments may limit the effectiveness of
psychotherapy, modifications such as the inclusion of a caregiver in the
therapy process or adjunctive cognitive remediation techniques can
improve outcomes.
References
Abramowitz JS: The practice of exposure therapy: relevance of cognitive-
behavioral theory and extinction theory. Behav Ther 44(4):548–558,
2013 24094780
Abrams RC: Personality disorders in the elderly. Int J Geriatr Psychiatry
11:759–763, 1996
Abrams RC, Bromberg CE: Personality disorders in the elderly: a flagging
field of inquiry. Int J Geriatr Psychiatry 21(11):1013–1017, 2006
17061248
Abrams RC, Horowitz SV: Personality disorders after age 50: a meta-
analytic review of the literature, in Personality Disorders in Older
Adults: Emerging Issues in Diagnosis and Treatment. Edited by
Rosowsky E, Abrams RC. Mahwah, NJ, Lawrence Erlbaum, 1999, pp
55–68
Abrams RC, Rosendahl E, Card C, et al: Personality disorder correlates of
late and early onset depression. J Am Geriatr Soc 42(7):727–731, 1994
8014347
American Psychiatric Association: Diagnostic and Statistical Manual of
Mental Disorders, 5th Edition. Washington, DC, American Psychiatric
Association, 2013
Alexander CL, Arnkoff DB, Glass CR: Bringing psychotherapy to primary
care: innovations and challenges. Clinical Psychology: Science and
Practice 17(3):191–214, 2010
Anguera JA, Boccanfuso J, Rintoul JL, et al: Video game training enhances
cognitive control in older adults. Nature 501(7465): 97–101, 2013
24005416
Areán PA, Cook BL: Psychotherapy and combined
psychotherapy/pharmacotherapy for late life depression. Biol
Psychiatry 52(3):293–303, 2002 12182934
Areán PA, Perri MG, Nezu AM, et al: Comparative effectiveness of social
problem-solving therapy and reminiscence therapy as treatments for
depression in older adults. J Consult Clin Psychol 61(6):1003–1010,
1993 8113478
Areán P, Hegel M, Vannoy S, et al: Effectiveness of problem-solving
therapy for older, primary care patients with depression: results from
the IMPACT project. Gerontologist 48(3):311–323, 2008 18591356
Areán PA, Mackin S, Vargas-Dwyer E, et al: Treating depression in
disabled, low-income elderly: a conceptual model and
recommendations for care. Int J Geriatr Psychiatry 25(8):765–769,
2010a 20602424
Areán PA, Raue P, Mackin RS, et al: Problem-solving therapy and
supportive therapy in older adults with major depression and executive
dysfunction. Am J Psychiatry 167(11):1391–1398, 2010b 20516155
Armento ME, Stanley MA, Marsh L, et al: Cognitive behavioral therapy for
depression and anxiety in Parkinson’s disease: a clinical review. J
Parkinsons Dis 2(2):135–151, 2012 23939438
Ayers CR, Sorrell JT, Thorp SR, et al: Evidence-based psychological
treatments for late-life anxiety. Psychol Aging 22(1):8–17, 2007
17385978
Babor TF, McRee BG, Kassebaum PA, et al: Screening, Brief Intervention,
and Referral to Treatment (SBIRT): toward a public health approach to
the management of substance abuse. Subst Abus 28(3):7–30, 2007
18077300
Barrowclough C, King P, Colville J, et al: A randomized trial of the
effectiveness of cognitive-behavioral therapy and supportive
counseling for anxiety symptoms in older adults. J Consult Clin
Psychol 69(5):756–762, 2001 11680552
Barry KL, Blow FC, Oslin DW: Substance abuse in older adults: review
and recommendations for education and practice in medical settings.
Subst Abus 23 (2 suppl):105–131, 2002
Bartels SJ, Coakley EH, Zubritsky C, et al; PRISM-E Investigators:
Improving access to geriatric mental health services: a randomized trial
comparing treatment engagement with integrated versus enhanced
referral care for depression, anxiety, and at-risk alcohol use. Am J
Psychiatry 161(8):1455–1462, 2004 15285973
Bland P: Group CBT is a cost-effective option for persistent back pain.
Practitioner 254(1728):7, 2010 20486479
Blazer D, George LK, Hughes D: The epidemiology of anxiety disorders:
an age comparison, in Anxiety in the Elderly: Treatment and Research.
Edited by Salzman C, Lebowitz BD. New York, Springer, 1991, pp 17–
30
Bowman D, Scogin F, Lyrene B: The efficacy of self-examination therapy
and cognitive bibliotherapy in the treatment of mild to moderate
depression. Psychother Res 5:131–140, 1995
Brown JS, Sellwood K, Beecham JK, et al: Outcome, costs and patient
engagement for group and individual CBT for depression: a naturalistic
clinical study. Behav Cogn Psychother 39(3):355–358, 2011 21406135
Bruce ML, Ten Have TR, Reynolds CF 3rd, et al: Reducing suicidal
ideation and depressive symptoms in depressed older primary care
patients: a randomized controlled trial. JAMA 291(9):1081–1091, 2004
14996777
Carreira K, Miller MD, Frank E, et al: A controlled evaluation of monthly
maintenance interpersonal psychotherapy in late-life depression with
varying levels of cognitive function. Int J Geriatr Psychiatry
23(11):1110–1113, 2008 18457338
Chen H, Coakley EH, Cheal K, et al: Satisfaction with mental health
services in older primary care patients. Am J Geriatr Psychiatry
14(4):371–379, 2006 16582046
Choi NG, Hegel MT, Marti CN, et al: Telehealth problem-solving therapy
for depressed low-income homebound older adults. Am J Geriatr
Psychiatry 22(3):263–271, 2013 23567376
Conner KO, Copeland VC, Grote NK, et al: Barriers to treatment and
culturally endorsed coping strategies among depressed African-
American older adults. Aging Ment Health 14(8):971–983, 2010
21069603
Covin R, Ouimet AJ, Seeds PM, et al: A meta-analysis of CBT for
pathological worry among clients with GAD. J Anxiety Disord
22(1):108–116, 2008 17321717
Cuijpers P, van Straten A, Warmerdam L: Problem solving therapies for
depression: a meta-analysis. Eur Psychiatry 22(1):9–15, 2007
17194572
Cuijpers P, van Straten A, Warmerdam L, et al: Psychological treatment of
depression: a meta-analytic database of randomized studies. BMC
Psychiatry 8:36, 2008 18485191
Deacon BJ, Abramowitz JS: Cognitive and behavioral treatments for
anxiety disorders: a review of meta-analytic findings. J Clin Psychol
60(4):429–441, 2004 15022272
DeBerry S: An evaluation of progressive muscle relaxation on stress related
symptoms in a geriatric population. Int J Aging Hum Dev 14(4):255–
269, 1981–1982 7345035
DeBerry S: The effects of meditation-relaxation on anxiety and depression
in a geriatric population. Psychotherapy 19:512–521, 1982
DeBerry S, Davis S, Reinhard KE: A comparison of meditation-relaxation
and cognitive/behavioral techniques for reducing anxiety and
depression in a geriatric population. J Geriatr Psychiatry 22(2):231–
247, 1989 2701175
Fleming MF, Manwell LB, Barry KL, et al: Brief physician advice for
alcohol problems in older adults: a randomized community-based trial.
J Fam Pract 48(5):378–384, 1999 10334615
Foa EB, Kozak MJ: Emotional processing of fear: exposure to corrective
information. Psychol Bull 99(1):20–35, 1986 2871574
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state”: a practical
method for grading the cognitive state of patients for the clinician. J
Psychiatr Res 12(3):189–198, 1975 1202204
Gerdner LA, Buckwalter KC, Reed D: Impact of a psychoeducational
intervention on caregiver response to behavioral problems. Nurs Res
51(6):363–374, 2002 12464756
Gorenstein EE, Kleber MS, Mohlman J, et al: Cognitive-behavioral therapy
for management of anxiety and medication taper in older adults. Am J
Geriatr Psychiatry 13(10):901–909, 2005 16223969
Gould RL, Coulson MC, Howard RJ: Cognitive behavioral therapy for
depression in older people: a meta-analysis and meta-regression of
randomized controlled trials. J Am Geriatr Soc 60(10):1817–1830,
2012 23003115
Gradman TJ, Thompson LW, Gallagher-Thompson D: Personality disorders
and treatment outcome, in Personality Disorders in Older Adults:
Emerging Issues in Diagnosis and Treatment. Edited by Rosowsky E,
Abrams RC. Mahwah, NJ, Erlbaum, 1999, pp 69–94
Gum AM, Areán PA, Hunkeler E, et al: Depression treatment preferences in
older primary care patients. Gerontologist 46(1): 14–22, 2006
16452280
Gum AM, Areán PA, Bostrom A: Low-income depressed older adults with
psychiatric comorbidity: secondary analyses of response to
psychotherapy and case management. Int J Geriatr Psychiatry
22(2):124–130, 2007 17096464
Gum AM, Iser L, Petkus A: Behavioral health service utilization and
preferences of older adults receiving home-based aging services. Am J
Geriatr Psychiatry 18(6):491–501, 2010 21217560
Hall GR, Buckwalter KC: Progressively lowered stress threshold: a
conceptual model for care of adults with Alzheimer’s disease. Arch
Psychiatr Nurs 1(6):399–406, 1987 3426250
Hariri AR, Bookheimer SY, Mazziotta JC: Modulating emotional responses:
effects of a neocortical network on the limbic system. Neuroreport
11(1):43–48, 2000 10683827
Hayslip B, Paggi K, Poole M, et al: The impact of mental aerobics training
on memory impaired older adults. Clinical Gerontologist 32(4):389–
394, 2009
Hinrichsen GA: Why multicultural issues matter for practitioners working
with older adults. Professional Psychology: Research and Practice
37(1):29–35, 2006
Huang HL, Shyu YIL, Chen MC, et al: A pilot study on a home-based
caregiver training program for improving caregiver self-efficacy and
decreasing the behavioral problems of elders with dementia in Taiwan.
Int J Geriatr Psychiatry 18(4):337–345, 2003 12673611
Karlin BE, Trockel M, Brown GK, et al: Comparison of the effectiveness of
cognitive behavioral therapy for depression among older versus
younger veterans: results of a national evaluation. J Gerontol B Psychol
Sci Soc Sci 70(1):3–12, 2015 24218096
Kenan MM, Kendjelic EM, Molinari VA, et al: Age-related differences in
the frequency of personality disorders among inpatient veterans. Int J
Geriatr Psychiatry 15(9):831–837, 2000 10984730
Kiosses DN, Areán PA, Teri L, et al: Home-delivered problem adaptation
therapy (PATH) for depressed, cognitively impaired, disabled elders: a
preliminary study. Am J Geriatr Psychiatry 18(11):988–998, 2010
20808092
Kiosses DN, Teri L, Velligan DI, Alexopoulos GS: A home-delivered
intervention for depressed, cognitively impaired, disabled elders. Int J
Geriatr Psychiatry 26(3):256–262, 2011 20665555
Knight BG, Poon CY: Contextual adult life span theory for adapting
psychotherapy with older adults. Journal of Rational-Emotive &
Cognitive-Behavior Therapy 26(4):232–249, 2008
Lebowitz BD, Pearson JL, Schneider LS, et al: Diagnosis and treatment of
depression in late life. Consensus statement update. JAMA
278(14):1186–1190, 1997 9326481
Linehan MM: Cognitive-Behavioral Treatment of Borderline Personality
Disorder. New York, Guilford, 1993
Livingston G, Johnston K, Katona C, et al; Old Age Task Force of the
World Federation of Biological Psychiatry: Systematic review of
psychological approaches to the management of neuropsychiatric
symptoms of dementia. Am J Psychiatry 162(11):1996–2021, 2005
16263837
Luoma JB, Hayes SC, Walser RD: Learning ACT: An Acceptance and
Commitment Therapy skills-training manual for therapists. Oakland,
CA, New Harbinger, 2007
Lynch TR, Cheavens JS: Dialectical behavior therapy for comorbid
personality disorders. J Clin Psychol 64(2):154–167, 2008 18186120
Lynch TR, Cheavens JS, Cukrowicz KC, et al: Treatment of older adults
with co-morbid personality disorder and depression: a dialectical
behavior therapy approach. Int J Geriatr Psychiatry 22(2):131–143,
2007 17096462
Manning KJ, Alexopoulos GS, Banerjee S, et al: Executive functioning
complaints and escitalopram treatment response in late-life depression.
Am J Geriatr Psychiatry Nov 27, 2013 [Epub ahead of print] 24388222
Marquett RM, Thompson LW, Reiser RP, et al: Psychosocial predictors of
treatment response to cognitive-behavior therapy for late-life
depression: an exploratory study. Aging Ment Health 17(7):830–838,
2013 23631698
McLaughlin DP, McFarland K: A randomized trial of a group based
cognitive behavior therapy program for older adults with epilepsy: the
impact on seizure frequency, depression and psychosocial well-being. J
Behav Med 34(3):201–207, 2011 20927577
Miller MD: Using interpersonal therapy (IPT) with older adults today and
tomorrow: a review of the literature and new developments. Curr
Psychiatry Rep 10(1):16–22, 2008 18269890
Miller MD, Reynolds CF 3rd: Expanding the usefulness of Interpersonal
Psychotherapy (IPT) for depressed elders with co-morbid cognitive
impairment. Int J Geriatr Psychiatry 22(2):101–105, 2007 17096459
Miller MD, Cornes C, Frank E, et al: Interpersonal psychotherapy for late-
life depression: past, present, and future. J Psychother Pract Res
10(4):231–238, 2001 11696649
Miller MD, Frank E, Cornes C, et al: The value of maintenance
interpersonal psychotherapy (IPT) in older adults with different IPT
foci. Am J Geriatr Psychiatry 11(1):97–102, 2003 12527545
Mohlman J: More power to the executive? A preliminary test of CBT plus
executive skills training for treatment of late-life GAD. Cognitive and
Behavioral Practice 15:306–316, 2008
Mohlman J, Gorman JM: The role of executive functioning in CBT: a pilot
study with anxious older adults. Behav Res Ther 43(4):447–465, 2005
15701356
Mohlman J, Gorenstein EE, Kleber M, et al: Standard and enhanced
cognitive-behavior therapy for late-life generalized anxiety disorder:
two pilot investigations. Am J Geriatr Psychiatry 11(1):24–32, 2003
12527537
Morse JQ, Lynch TR: A preliminary investigation of self-reported
personality disorders in late life: prevalence, predictors of depressive
severity, and clinical correlates. Aging Ment Health 8(4):307–315,
2004 15370047
Moscovitch DA, Antony MM, Swinson RP: Exposure-based treatments for
anxiety disorders, in Oxford Handbook of Anxiety and Related
Disorders. Edited by Antony MM, Stein MB. New York, Oxford
University Press, 2009, pp 461–475
Moss-Morris R, Dennison L, Landau S, et al: A randomized controlled trial
of cognitive behavioral therapy (CBT) for adjusting to multiple
sclerosis (the saMS trial): does CBT work and for whom does it work?
J Consult Clin Psychol 81(2):251–262, 2013 22730954
Naylor EV, Antonuccio DO, Litt M, et al: Bibliotherapy as a treatment for
depression in primary care. J Clin Psychol Med Settings 17(3):258–
271, 2010 20803165
Nezu AM, Perri MG: Social problem-solving therapy for unipolar
depression: an initial dismantling investigation. J Consult Clin Psychol
57(3):408–413, 1989 2738213
Onder G, Zanetti O, Giacobini E, et al: Reality orientation therapy
combined with cholinesterase inhibitors in Alzheimer’s disease:
randomised controlled trial. Br J Psychiatry 187:450–455, 2005
16260821
Oslin DW, Grantham S, Coakley E, et al: PRISM-E: comparison of
integrated care and enhanced specialty referral in managing at-risk
alcohol use. Psychiatr Serv 57(7):954–958, 2006 16816279
Peng XD, Huang CQ, Chen LJ, et al: Cognitive behavioural therapy and
reminiscence techniques for the treatment of depression in the elderly:
a systematic review. J Int Med Res 37(4):975–982, 2009 19761679
Pimontel MA, Culang-Reinlieb ME, Morimoto SS, et al: Executive
dysfunction and treatment response in late-life depression. Int J Geriatr
Psychiatry 27(9):893–899, 2012 22009869
Pinquart M, Duberstein PR, Lyness JM: Effects of psychotherapy and other
behavioral interventions on clinically depressed older adults: a meta-
analysis. Aging Ment Health 11(6):645–657, 2007 18074252
Post EP, Miller MD, Schulberg HC: Using interpersonal psychotherapy
(IPT) to treat depression in older primary care patients. Geriatrics
63(3):18–28, 2008 18312024
Preschl B, Maercker A, Wagner B, et al: Life-review therapy with computer
supplements for depression in the elderly: a randomized controlled
trial. Aging Ment Health 16(8):964–974, 2012 22788983
Quayhagen MP, Quayhagen M, Corbeil RR, et al: A dyadic remediation
program for care recipients with dementia. Nurs Res 44(3): 153–159,
1995 7761291
Quayhagen MP, Quayhagen M, Corbeil RR, et al: Coping with dementia:
evaluation of four nonpharmacologic interventions. Int Psychogeriatr
12(2):249–265, 2000 10937544
Reynolds CF3rd, Frank E, Perel JM, et al: Nortriptyline and interpersonal
psychotherapy as maintenance therapies for recurrent major
depression: a randomized controlled trial in patients older than 59
years. JAMA 281(1):39–45, 1999 9892449
Reynolds CF3rd, Dew MA, Martire LM, et al: Treating depression to
remission in older adults: a controlled evaluation of combined
escitalopram with interpersonal psychotherapy versus escitalopram
with depression care management. Int J Geriatr Psychiatry
25(11):1134–1141, 2010 20957693
Rickard HC, Scogin F, Keith S: A one-year follow-up of relaxation training
for elders with subjective anxiety. Gerontologist 34(1):121–122, 1994
8150300
Rokke PD, Scogin F: Depression treatment preferences in younger and
older adults. Journal of Clinical Geropsychology 1:243–257, 1995
Rovner BW, Casten RJ, Hegel MT, et al: Minimal depression and vision
function in age-related macular degeneration. Ophthalmology
113(10):1743–1747, 2006 16893569
Rovner BW, Casten RJ, Hegel MT, et al: Preventing depression in age-
related macular degeneration. Arch Gen Psychiatry 64(8):886–892,
2007 17679633
Satre DD, Mertens J, Areán PA, et al: Contrasting outcomes of older versus
middle-aged and younger adult chemical dependency patients in a
managed care program. J Stud Alcohol 64(4):520–530, 2003 12921194
Satre DD, Mertens JR, Areán PA, et al: Five-year alcohol and drug
treatment outcomes of older adults versus middle-aged and younger
adults in a managed care program. Addiction 99(10):1286–1297, 2004
15369567
Schonfeld L, Dupree LW, Dickson-Euhrmann E, et al: Cognitive-behavioral
treatment of older veterans with substance abuse problems. J Geriatr
Psychiatry Neurol 13(3): 124–129, 2000 11001134
Schonfeld L, King-Kallimanis BL, Duchene DM, et al: Screening and brief
intervention for substance misuse among older adults: the Florida
BRITE project. Am J Public Health 100(1):108–114, 2010 19443821
Scogin F, Jamison C, Gochneaur K: Comparative efficacy of cognitive and
behavioral bibliotherapy for mildly and moderately depressed older
adults. J Consult Clin Psychol 57(3):403–407, 1989 2738212
Scogin F, Rickard HC, Keith S, et al: Progressive and imaginal relaxation
training for elderly persons with subjective anxiety. Psychol Aging
7(3):419–424, 1992 1388863
Serfaty MA, Haworth D, Blanchard M, et al: Clinical effectiveness of
individual cognitive behavioral therapy for depressed older people in
primary care: a randomized controlled trial. Arch Gen Psychiatry
66(12):1332–1340, 2009 19996038
Serrano Selva JP, Latorre Postigo JM, Ros Segura L, et al: Life review
therapy using autobiographical retrieval practice for older adults with
clinical depression. Psicothema 24(2):224–229, 2012 22420349
Snodgrass J: Toward holistic care: integrating spirituality and cognitive
behavioral therapy for older adults. Journal of Religion, Spirituality &
Aging 21(3):219–236, 2009
Spector A, Orrell M, Davies S, et al: Can reality orientation be
rehabilitated? Development and piloting of an evidence-based
programme of cognition-based therapies for people with dementia.
Neuropsychol Rehabil 11:377–379, 2001
Spector A, Thorgrimsen L, Woods B, et al: Efficacy of an evidence-based
cognitive stimulation therapy programme for people with dementia:
randomised controlled trial. Br J Psychiatry 183:248–254, 2003
12948999
Stanley MA, Novy DM: Cognitive-behavior therapy for generalized anxiety
in late life: an evaluative overview. J Anxiety Disord 14(2):191–207,
2000 10864385
Stanley MA, Beck JG, Glassco JD: Treatment of generalized anxiety in
older adults: a preliminary comparison of cognitive-behavioral and
supportive approaches. Behav Ther 27:565–581, 1996
Stanley MA, Beck JG, Novy DM, et al: Cognitive-behavioral treatment of
late-life generalized anxiety disorder. J Consult Clin Psychol
71(2):309–319, 2003a 12699025
Stanley MA, Hopko DR, Diefenbach GJ, et al: Cognitive-behavior therapy
for late-life generalized anxiety disorder in primary care: preliminary
findings. Am J Geriatr Psychiatry 11:92–96, 2003b 12527544
Stanley MA, Diefenbach GJ, Hopko DR: Cognitive behavioral treatment
for older adults with generalized anxiety disorder. A therapist manual
for primary care settings. Behav Modif 28(1):73–117, 2004 14710708
Stanley MA, Wilson NL, Novy DM, et al: Cognitive behavior therapy for
generalized anxiety disorder among older adults in primary care: a
randomized clinical trial. JAMA 301(14):1460–1467, 2009 19351943
Stuart S, Noyes R Jr: Interpersonal psychotherapy for somatizing patients.
Psychother Psychosom 75(4):209–219, 2006 16785770
Substance Abuse and Mental Health Services Administration (SAMHSA):
Results from the 2010 National Survey on Drug Use and Health:
Summary of National Findings (HHS Publ No SMA-11-4658; NSDUH
Series H-41). 2011. Available at:
http://www.samhsa.gov/data/nsduh/2k10nsduh/2k10results.htm.
Accessed June 10, 2014.
Tasman W, Rovner B: Age-related macular degeneration: treating the whole
patient. Arch Ophthalmol 122(4):648–649, 2004 15078685
Teri L, Gallagher-Thompson D: Cognitive-behavioral interventions for
treatment of depression in Alzheimer’s patients. Gerontologist
31(3):413–416, 1991 1879719
Teri L, McKenzie G, LaFazia D: Psychosocial treatment of depression in
older adults with dementia. Clinical Psychology: Science and Practice
12:303–316, 2005
Thompson LW, Gallagher D, Czirr R: Personality disorder and outcome in
the treatment of late-life depression. J Geriatr Psychiatry 21(2):133–
153, 1988 3216093
Torgersen S, Kringlen E, Cramer V: The prevalence of personality disorders
in a community sample. Arch Gen Psychiatry 58(6):590–596, 2001
11386989
Unützer J, Katon W, Williams JW Jr, et al: Improving primary care for
depression in late life: the design of a multicenter randomized trial.
Med Care 39(8):785–799, 2001 11468498
van’t Hof E, Stein DJ, Marks I, et al: The effectiveness of problem solving
therapy in deprived South African communities: results from a pilot
study. BMC Psychiatry 11:156, 2011 21961801
Videler AC, van Royen RJJ, van Alphen SPJ: Schema therapy with older
adults: call for evidence. Int Psychogeriatr 24(7):1186–1187, 2012
22340711
Walker DA, Clarke M: Cognitive behavioural psychotherapy: a comparison
between younger and older adults in two inner city mental health
teams. Aging Ment Health 5(2):197–199, 2001 11511068
Wang PS, Berglund P, Olfson M, et al: Failure and delay in initial treatment
contact after first onset of mental disorders in the National Comorbidity
Survey Replication. Arch Gen Psychiatry 62(6):603–613, 2005
15939838
Weissman MM, Myers JK, Thompson WD: Depression and its treatment in
a US urban community—1975–1976. Arch Gen Psychiatry 38(4):417–
421, 1981 6111302
Wetherell JL, Gatz M, Craske MG: Treatment of generalized anxiety
disorder in older adults. J Consult Clin Psychol 71(1):31–40, 2003
12602423
Wetherell JL, Hopko DR, Diefenbach GJ, et al: Cognitive-behavioral
therapy for late-life generalized anxiety disorder: who gets better?
Behav Ther 36:147–156, 2005
Wetherell JL, Afari N, Ayers CR, et al: Acceptance and Commitment
Therapy for generalized anxiety disorder in older adults: a preliminary
report. Behav Ther 42(1):127–134, 2011 21292059
Wetherell JL, Petkus AJ, White KS, et al: Antidepressant medication
augmented with cognitive-behavioral therapy for generalized anxiety
disorder in older adults. Am J Psychiatry 170(7):782–789, 2013
23680817
Wilkinson P, Alder N, Juszczak E, et al: A pilot randomised controlled trial
of a brief cognitive behavioural group intervention to reduce recurrence
rates in late life depression. Int J Geriatr Psychiatry 24(1):68–75, 2009
18615497
Wolitzky-Taylor KB, Castriotta N, Lenze EJ, et al: Anxiety disorders in
older adults: a comprehensive review. Depress Anxiety 27(2):190–211,
2010 20099273
Yang JA, Garis J, Jackson C, et al: Providing psychotherapy to older adults
in home: benefits, challenges, and decision-making guidelines. Clinical
Gerontologist 32(4):333–346, 2009
Young MM, Stevens A, Porath-Waller A, et al: Effectiveness of brief
interventions as part of the screening, brief intervention and referral to
treatment (SBIRT) model for reducing the non-medical use of
psychoactive substances: a systematic review protocol. Syst Rev 1:22,
2012 22587894
Zweig RA: Personality disorder in older adults: assessment challenges and
strategies. Professional Psychology: Research and Practice 39(3):298–
305, 2008
Suggested Readings
Barrowclough C, King P, Colville J, et al: A randomized trial of the
effectiveness of cognitive-behavioral therapy and supportive
counseling for anxiety symptoms in older adults. J Consult Clin
Psychol 69:756–762, 2001
Bartels SJ, Coakley EH, Zubritsky C, et al: Improving access to geriatric
mental health services: a randomized trial comparing treatment
engagement with integrated versus enhanced referral care for
depression, anxiety, and at-risk alcohol use. Am J Psychiatry
161:1455–1462, 2004
Bruce ML, Ten Have TR, Reynolds CF, et al: Reducing suicidal ideation
and depressive symptoms in depressed older primary care patients.
JAMA 291:1081–1091, 2004
Lebowitz BD, Pearson JL, Schneider LS, et al: Diagnosis and treatment of
depression in late life: consensus statement update. JAMA 278:1186–
1190, 1997
Livingston G, Johnston K, Katona C, et al: Systematic review of
psychological approaches to the management of neuropsychiatric
symptoms of dementia. Am J Psychiatry 162:1996–2021, 2005
Lynch TR, Cheavens JS, Cukrowicz KC, et al: Treatment of older adults
with co-morbid personality disorder and depression: a dialectical
behavior therapy approach. Int J Geriatr Psychiatry 22:131–143, 2007
Mohlman J, Gorman JM: The role of executive functioning in CBT: a pilot
study with anxious older adults. Behav Res Ther 43: 447–465, 2004
Oslin DW, Grantham S, Coakley E, et al: PRISM-E: comparison of
integrated care and enhanced specialty referral in managing at-risk
alcohol use. Psychiatr Serv 57: 954–958, 2006
Thompson LW, Coon DW, Gallagher-Thompson D, et al: Comparison of
desipramine and cognitive/behavioral therapy in the treatment of
elderly outpatients with mild-to-moderate depression. Am J Geriatr
Psychiatry 9:225–240, 2001
Unützer JM, Katon WM, Williams JWJ, et al: Improving primary care for
depression in late life: the design of a multicenter randomized trial.
Med Care 39:785–799, 2001
We gratefully acknowledge the contributions of Thomas R. Lynch, Ph.D.,
and Dawn E. Epstein to previous editions of this chapter. Moria J. Smoski’s
effort toward this chapter was supported by grant K23 MH087754 from the
National Institute of Mental Health (NIMH). Patricia A. Areán’s effort
toward this chapter was supported by grant K24 MH074717 from NIMH.
CHAPTER 24
• Can the family member with dementia use the telephone to call for help
from a family member or to call 911? Will the person respond
inappropriately to telemarketers or to mail or e-mail scams? Have
mysterious packages or bills for unusual items begun appearing? Does the
person make repetitive calls every few minutes to the police or to the same
family member at work or at home? Are any suspicious new friends
accompanying the patient to the bank?
• Can the family member with dementia get to the store or to his or her
regular activities? Does the patient overbuy or underbuy certain items?
• Can the individual handle money and pay bills or, if not, is he or she
willing to let others do this for him or her or have bills forwarded or paid
online?
• Can the patient take medicine appropriately, on time, and in correct doses?
Does he or she self-medicate or risk overdoses of unnecessary
medications?
• Is the family member bathing, changing clothes, and dressing
appropriately for the weather?
• Is the person leaving the house after dark or traveling in dangerous areas
alone? Does the patient let strangers in or buy from or contribute to
questionable causes based on visits to his or her home?
• Is the person having problems positioning his or her body to use a toilet,
or is he or she urinating in wastebaskets or outdoors?
• Is the patient falling or getting lost by wandering outside a safe area?
• Are there significant changes in the family member’s appetite, weight,
sleep, appearance, or eating habits?
• Is discreet surveillance by neighbors, friends, or family readily available?
Conclusion
Clinical work with families of older adults is about helping them adapt to
change, uncertainty, variability, and loss. Much of psychiatric treatment of
families helps them modify expectations for new dependency while
learning to forgive themselves and others for inevitable doubts and
mistakes. Interprofessional partnerships and teamwork with the Alzheimer’s
Association, with health care or primary care coordination or care transition
(medical home) initiatives, or with nurses or social workers offer the most
effective and efficient models for psychiatric services to families of older
adults. There is often as much need for “timed and dosed” patient and
family education as there is need for treatment of psychotic, depressive,
anxious, or other psychiatric symptoms or syndromes of the elder or family
members. Families expect psychiatrists to provide active treatment and
monitoring of psychiatric symptoms, reassurance, interpretation of
information, and referrals. In addition, it is always helpful to acknowledge
losses and contributions to care by individual family members, to encourage
caregiver self-care, to offer authoritative absolution for inevitable mistakes,
and to offer decisional support, especially in regard to transitions in care,
including a move to palliative or hospice care.
Key Points
• Psychiatric treatment of families helps them modify expectations for
their family member’s progressive cognitive decline and learn to forgive
themselves and others for inevitable doubts and mistakes.
• Interprofessional consultation, partnerships, and teamwork with the
Alzheimer’s Association and with health, aging, and social services offer
the most effective and efficient models for psychiatric services to
families of older adults.
• Psychiatrists should provide active treatment and monitoring of family
caregivers’ psychiatric symptoms, outline reasonable expectations, and
offer families information about outcomes of treatment.
• It is helpful to acknowledge family caregivers’ losses and their
contributions to care. It is also important to encourage their own self-
care and to offer them expert decisional support during transitions in
care.
References
Adelman RD, Tmanova LL, Delgado D, et al: Caregiver burden: a clinical
review. JAMA 311(10):1052–1060, 2014 24618967
Bass DM, Judge KS, Snow AL, et al: Caregiver outcomes of partners in
dementia care: effect of a care coordination program for veterans with
dementia and their family members and friends. J Am Geriatr Soc
61(8):1377–1386, 2013 23869899
Batsch NL, Mittelman MS: International World Alzheimer Report 2012:
Overcoming the Stigma of Dementia. 2012. Available at:
www.alz.org/documents_custom/world_report_2012_final.pdfwww.alz
.org/documents_custom/world_report_2012_final.pdf. Accessed June
11, 2014.
Belle SH, Burgio L, Burns R, et al; Resources for Enhancing Alzheimer’s
Caregiver Health (REACH) II Investigators: Enhancing the quality of
life of dementia caregivers from different ethnic or racial groups: a
randomized, controlled trial. Ann Intern Med 145(10):727–738, 2006
17116917
Boustani MA, Sachs GA, Alder CA, et al: Implementing innovative models
of dementia care: The Healthy Aging Brain Center. Aging Ment Health
15(1):13–22, 2011 21271387
Burgio L, Stevens A, Guy D, et al: Impact of two psychosocial
interventions on white and African American family caregivers of
individuals with dementia. Gerontologist 43(4):568–579, 2003
12937335
Callahan CM, Arling G, Tu W, et al: Transitions in care for older adults
with and without dementia. J Am Geriatr Soc 60(5):813–820, 2012
22587849
Coon DW, Thompson L, Steffen A, et al: Anger and depression
management: psychoeducational skill training interventions for women
caregivers of a relative with dementia. Gerontologist 43(5):678–689,
2003 14570964
Derence K: Dementia-specific respite: the key to effective caregiver
support. N C Med J 66(1):48–51, 2005 15786679
Eisdorfer C, Czaja SJ, Loewenstein DA, et al: The effect of a family
therapy and technology-based intervention on caregiver depression.
Gerontologist 43(4):521–531, 2003 12937331
Fortinsky RH, Delaney C, Harel O, et al: Results and lessons learned from a
nurse practitioner-guided dementia care intervention for primary care
patients and their family caregivers. Res Gerontol Nurs 7(3):126–137,
2014
Fulmer T, Paveza G, VandeWeerd C, et al: Dyadic vulnerability and risk
profiling for elder neglect. Gerontologist 45(4):525–534, 2005
16051915
Gallagher-Thompson D, Coon DW: Evidence-based psychological
treatments for distress in family caregivers of older adults. Psychol
Aging 22(1):37–51, 2007 17385981
Gaugler JE, Mittelman MS, Hepburn K, et al: Predictors of change in
caregiver burden and depressive symptoms following nursing home
admission. Psychol Aging 24(2):385–296, 2009 19485656
Gillick MR: The critical role of caregivers in achieving patient-centered
care. JAMA 310(6):575–576, 2013 23867885
Gitlin LN, Reever K, Dennis MP, et al: Enhancing quality of life of families
who use adult day services: Short- and long-term effects of the adult
day services plus program. Gerontologist 46(5):630–639, 2006
17050754
Gitlin LN, Kales KH, Lyketsos CG: Nonpharmacologic management of
behavioral symptoms in dementia. JAMA 308(19): 2020–2029, 2012
23168825
Gwyther LP: Family issues in dementia: finding a new normal. Neurol Clin
18(4):993–1010, 2000 11072271
Gwyther LP: Family care and Alzheimer’s disease: what do we know?
What can we do? N C Med J 66(1):37–40, 42, 2005 15786677
Hepburn K, Lewis M, Tornatore J, et al: The Savvy Caregiver program: the
demonstrated effectiveness of a transportable dementia caregiver
psychoeducation program. J Gerontol Nurs 33(3):30–36, 2007
17378189
Hughes SL, Giobbie-Hurder A, Weaver FM, et al: Relationship between
caregiver burden and health-related quality of life. Gerontologist
39(5):534–545, 1999 10568078
Hurd MD, Martorell P, Delavande A, et al: Monetary costs of dementia in
the United States. N Engl J Med 368(14):1326–1334, 2013 23550670
King AC, Brassington G: Enhancing physical and psychological
functioning in older family caregivers: the role of regular physical
activity. Ann Behav Med 19(2):91–100, 1997 9603683
Langa KM, Chernew ME, Kabeto MU, et al: National estimates of the
quantity and cost of informal caregiving for the elderly with dementia.
J Gen Intern Med 16:770–776, 2001 11722692
Livingston G, Barber J, Rapaport P, et al: Clinical effectiveness of a manual
based coping strategy programme (START, STrAtegies for RelaTives)
in promoting the mental health of carers of family members with
dementia: pragmatic randomised controlled trial. BMJ 347:f6276, 2013
24162942
Logsdon RG, Pike KC, McCurry SM, et al: Early-stage memory loss
support groups: outcomes from a randomized controlled clinical trial. J
Gerontol B Psychol Sci Soc Sci 65(6):691–697, 2010 20693265
Lund D, Utz R, Casarta M, et al: Examining what caregivers do during
respite time to make respite more effective. J Appl Gerontol 28(1):109–
131, 2009
Lyketsos CG, Steinberg M, Tschanz JT, et al: Mental and behavioral
disturbances in dementia: findings from the Cache County Study on
Memory in Aging. Am J Psychiatry 157(5):708–714, 2000 10784462
Lynch CA, Moran M, Lawlor BA: Firearms and dementia: a smoking gun?
Int J Geriatr Psychiatry 23(1):1–6, 2008 18081001
Maslow K: Translating Innovation to Impact: Evidence-Based Interventions
to Support People With Alzheimer’s Disease and Their Caregivers at
Home and in the Community. A White Paper. Washington, DC,
Alliance for Aging Research, Administration on Aging, MetLife
Foundation, September 2012. Available at:
http://www.aoa.gov/AoA_Programs/HPW/Alz_Grants/docs/Translating
InnovationtoImpactAlzheimersDisease.pdf. Accessed October 31,
2014.
McCurry SM, Gibbons LE, Logsdon RG, et al: Nighttime insomnia
treatment and education for Alzheimer’s disease: a randomized,
controlled trial. J Am Geriatr Soc 53(5):793–802, 2005 15877554
McCurry SM, Pike KC, Vitiello MV, et al: Increasing walking and bright
light exposure to improve sleep in community-dwelling persons with
Alzheimer’s disease: results of a randomized, controlled trial. J Am
Geriatr Soc 59(8):1393–1402, 2011 21797835
Mittelman MS, Ferris SH, Shulman E, et al: A family intervention to delay
nursing home placement of patients with Alzheimer disease. A
randomized controlled trial. JAMA 276(21):1725–1731, 1996 8940320
Mittelman MS, Roth DL, Haley WE, et al: Effects of a caregiver
intervention on negative caregiver appraisals of behavior problems in
patients with Alzheimer’s disease: results of a randomized trial. J
Gerontol B Psychol Sci Soc Sci 59(1):27–34, 2004 14722336
Mittelman MS, Roth DL, Clay OJ, et al: Preserving health of Alzheimer
caregivers: impact of a spouse caregiver intervention. Am J Geriatr
Psychiatry 15(9):780–789, 2007 17804831
Olin JT, Schneider LS, Katz IR, et al: Provisional diagnostic criteria for
depression of Alzheimer disease. Am J Geriatr Psychiatry 10(2):125–
128, 2002 11925273
Pillemer K, Suitor JJ: “It takes one to help one”: effects of similar others on
the well-being of caregivers. J Gerontol B Psychol Sci Soc Sci
51(5):S250–S257, 1996 8809010
Pinquart M, Sörensen S: Ethnic differences in stressors, resources, and
psychological outcomes of family caregiving: a meta-analysis.
Gerontologist 45(1):90–106, 2005 15695420
Pinquart M, Sörensen S: Gender differences in caregiver stressors, social
resources, and health: an updated meta-analysis. J Gerontol B Psychol
Sci Soc Sci 61:P33–P45, 2006a
Pinquart M, Sörensen S: Helping caregivers of persons with dementia:
which interventions work and how large are the effects? Int
Psychogeriatr 18:577–595, 2006b 16686964
Rabins PV, Lyketsos C, Steele C: Practical Dementia Care. Oxford, UK,
Oxford University Press, 2006
Reinhard SC, Levine C, Samis S: Home alone: family caregivers providing
complex chronic care. AARP Public Policy Institute. 2012. Available
at: http://www.aarp.org/home-family/caregiving/info-10-2012/home-
alone-family-caregivers-providing-complex-chronic-care.html.
Accessed June 13, 2014.
Roth DL, Haley WE, Hovater M, et al: Family caregiving and all-cause
mortality: findings from a population-based propensity-matched
analysis. Am J Epidemiol 178(10):1571–1578, 2013 24091890
Semiatin AM, O’Connor MK: The relationship between self-efficacy and
positive aspects of caregiving in Alzheimer’s disease caregivers. Aging
Ment Health 16(6):683–688, 2012 22360626
Sun F, Durkin DW, Hilgeman MM, et al: Predicting desire for institutional
placement among racially diverse dementia family caregivers: the role
of quality of care. Gerontologist 53(3):418–429, 2013 22961466
Teri L, Logsdon RG, Uomoto J, et al: Behavioral treatment of depression in
dementia patients: a controlled clinical trial. J Gerontol B Psychol Sci
Soc Sci 52(4):159–166, 1997 9224439
Teri L, Gibbons LE, McCurry SM, et al: Exercise plus behavioral
management in patients with Alzheimer disease: a randomized
controlled trial. JAMA 290(15):2015–2022, 2003 14559955
Tierney MC, Charles J, Naglie G, et al: Risk factors for harm in cognitively
impaired seniors who live alone: a prospective study. J Am Geriatr Soc
52(9):1435–1441, 2004 15341543
Tractenberg RE, Weiner MF, Thal LJ: Estimating the prevalence of
agitation in community-dwelling persons with Alzheimer’s disease. J
Neuropsychiatry Clin Neurosci 14(1):11–18, 2002 11884649
Tschanz JT, Piercy K, Corcoran CD, et al: Caregiver coping strategies
predict cognitive and functional decline in dementia: the cache county
dementia progression study. Am J Geriatr Psychiatry 21(1):57–66,
2013 23290203
U.S. Department of Veterans Affairs: Caregiver Self-Assessment
Worksheet. 2014. Available at:
http://va.gov/GERIATRICS/Guide/LongTermCare/Caregiver_Self_Ass
essment.pdf#zoom=100. Accessed June 11, 2014.
Weimer DL, Sager MA: Early identification and treatment of Alzheimer’s
disease: social and fiscal outcomes. Alzheimers Dement 5(3):215–226,
2009 19362885
Werner P, Mittelman MS, Goldstein D, et al: Family stigma and caregiver
burden in Alzheimer’s disease. Gerontologist 52(1): 89–97, 2012
22048807
Whitlatch CJ, Judge K, Zarit SH, et al: Dyadic intervention for family
caregivers and care receivers in early-stage dementia. Gerontologist
46(5):688–694, 2006 17050761
Yaffe K, Fox P, Newcomer R, et al: Patient and caregiver characteristics and
nursing home placement in patients with dementia. JAMA
287(16):2090–2097, 2002 11966383
Zarit SH, Whetzel CA, Kim K, et al: Daily stressors and adult day service
use by family caregivers: effects on depressive symptoms, positive
mood, and dehydroepiandrosterone-sulfate. Am J Geriatr Psychiatry
22(12):1592–1602, 2014 24566240
Suggested Readings
Adelman RD, Tmanova LL, Delgado D, et al: Caregiver burden: a clinical
review. JAMA 311(10):1052–1060, 2014
Belle SH, Burgio L, Burns R, et al; Resources for Enhancing Alzheimer’s
Caregiver Health (REACH) II Investigators: Enhancing the quality of
life of dementia caregivers from different ethnic or racial groups: a
randomized, controlled trial. Ann Intern Med 145(10):727–738, 2006
Gallagher-Thompson D, Coon DW: Evidence-based psychological
treatments for distress in family caregivers of older adults. Psychol
Aging 22(1):37–51, 2007
Mittelman MS, Roth DL, Clay OJ, Haley WE: Preserving health of
Alzheimer caregivers: impact of a spouse caregiver intervention. Am J
Geriatr Psychiatry 15(9):780–789, 2007
Rabins PV, Lyketsos C, Steele C: Practical Dementia Care. Oxford, UK,
Oxford University Press, 2006
CHAPTER 25
Depression
Among community-dwelling elders in the United States and Europe, depression increases the risk
of nursing home admission (Ahmed et al. 2007; Harris and Cooper 2006; Onder et al. 2007), and
this association remains after controlling for age, physical illness, and functional status (Harris
2007). Among nursing homes residents, depressive disorders represent the second most common
psychiatric diagnosis, after dementia. Most studies in U.S. nursing homes show depression
prevalence rates of 15%–50%, depending on the population studied and instruments used, and on
whether major depression or depressive symptoms are being reported (Baker and Miller 1991;
Chandler and Chandler 1988; Hyer and Blazer 1982; Katz et al. 1989; Kaup et al. 2007; Lesher
1986; Levin et al. 2007; McCusker et al. 2014; Parmelee et al. 1989; Rovner et al. 1986, 1990a,
1991; Tariot et al. 1993; Teeter et al. 1976). Studies from other countries have shown similar rates
(Ames 1990, 1991; Ames et al. 1988; Barca et al. 2010; Chahine et al. 2007; Harrison et al. 1990;
Horiguchi and Inami 1991; Jongenelis et al. 2004; Mann et al. 1984; Rozzini et al. 1996; Snowdon
1986; Snowdon and Donnelly 1986; Spagnoli et al. 1986; Trichard et al. 1982). Approximately
6%–10% of all nursing home residents, and 20%–25% of those who are cognitively intact, meet
criteria for a diagnosis of major depression; the latter figure is an order of magnitude greater than
rates among community-dwelling elderly persons (Blazer and Williams 1980; Kramer et al. 1985).
The prevalence of less severe but clinically significant (e.g., minor or subsyndromal)
depression among residents in long-term-care settings is even higher. In one study, Parmelee et al.
(1992) reported that the 1-year incidence of major depression was 9.4% and that patients with
preexisting minor depression were at increased risk; the incidence of minor depression among
those who were euthymic at baseline was 7.4%. Similarly, McCusker et al. (2014) found that
baseline depression score was an independent risk factor for incident depression. These data show
that minor or subsyndromal depression in nursing home residents appears to be a risk factor for
major depression and might represent an opportunity for preventive treatment in this population.
Depression among nursing home residents tends to be persistent. Although there may be
moderate decreases in self-rated depression in the initial 2 weeks to 6 months after nursing home
admission (Engle and Graney 1993; Smalbrugge et al. 2006), Ames et al. (1988) found that only
17% of patients with diagnosable depressive disorders had recovered after an average of 3.6 years
of follow-up. Smalbrugge et al. (2006) found persistence of symptoms in two-thirds of residents at
6-month follow-up, although rates were significantly higher among those with more severe
symptoms at baseline.
Evidence for morbidity associated with depression comes from studies that showed an increase
in pain complaints among residents with depression (McCusker et al. 2014; Parmelee et al. 1991),
an association between depression and biochemical markers of subnutrition (Katz et al. 1993), and
independent associations of delirium and diabetes with depression (McCusker et al. 2014).
Depression in nursing home residents, both with and without dementia, is associated with
disability (Kaup et al. 2007). Among individuals admitted to nursing homes for post–acute care
rehabilitation, those with depression have poorer functional outcomes (Webber et al. 2005). In
addition to its association with morbidity and disability, depression in nursing home populations
has been found to be associated with an increase in mortality rate, with effect sizes ranging from
1.6 to 3 (Ashby et al. 1991; Katz et al. 1989; Parmelee et al. 1992; Rovner et al. 1991; Sutcliffe et
al. 2007). Whereas Rovner et al. (1991) reported that the increased mortality rate remained
apparent after controlling for the patients’ medical diagnoses and level of disability, Parmelee et
al. (1992) found that the effect could be attributed to the interrelationships among depression,
disability, and physical illness.
In addition to the medical comorbidity, disability, and mortality that characterize major
depression among nursing home residents, there is evidence for heterogeneity in these patients
that may reflect the existence of clinically relevant subtypes of depression. A treatment study by
Katz et al. (1990) demonstrated that measures of self-care deficits and serum levels of albumin
were highly intercorrelated and that both predicted a lack of response to treatment with
nortriptyline. Therefore, although this study demonstrated that major depression is a specific,
treatable disorder—even in long-term-care patients with medical comorbidity—there is also
evidence in this setting for a treatment-relevant subtype of depression characterized by high levels
of disability and low levels of serum albumin. This latter condition may be related to the
syndrome of frailty that is well described in the geriatric medical literature (Malmstrom et al.
2014). As described later in this chapter (see “Pharmacotherapy”), evidence also indicates that
depression among nursing home residents with dementia may differ from depression among
residents who are depressed and cognitively intact. Several studies of nursing home residents have
demonstrated poorer response to treatment with noradrenergic and serotonergic antidepressant
drugs in very old residents with comorbid cognitive impairment or dementia (Magai et al. 2000;
Oslin et al. 2000; Rosen et al. 2000; Streim et al. 2000; Trappler and Cohen 1996, 1998). These
findings provide evidence for another treatment-relevant subtype of depression, although this
condition may not be specific to the nursing home setting. Similarly poor antidepressant treatment
responses have been amply demonstrated in very old outpatients whose depressive symptoms are
associated with vascular risk factors and executive dysfunction (Sneed et al. 2007, 2008).
Psychotherapy
Evidence for the efficacy of psychotherapy in other settings suggests that it may be of value for
treating mental disorders of aging in patients whose cognitive abilities allow them to participate.
However, a search of the PsycINFO, PubMed, and Cochrane databases for the terms
psychotherapy and nursing homes from 1987 to the time of writing this chapter identified only a
limited number of controlled studies of the effectiveness of specific psychotherapeutic modalities,
individual or group, for nursing home residents (Table 25–1). Bharucha et al. (2006) identified and
reviewed 18 controlled studies of “talk” psychotherapy for depression and psychological well-
being in nursing home populations, and found that a majority showed at least short-term benefits
on measures of mood, hopelessness, perceived control, self-esteem, or other psychological
variables. However, the authors noted that interpretation of the findings of many of these studies
was limited by small sample sizes, variable study entry criteria, short duration of trials,
heterogeneous outcome assessment methods, and lack of detail on intervention methods.
Controlled research on psychotherapeutic interventions has included studies of task-oriented
versus insight-oriented therapy (Moran and Gatz 1987); reality orientation (Baines et al. 1987);
reminiscence groups (Baines et al. 1987; Chao et al. 2006; Goldwasser et al. 1987; Haslam et al.
2010; McMurdo and Rennie 1993; Orten et al. 1989; Politis et al. 2004; Rattenbury and Stones
1989; Youssef 1990); exercise, activity, and progressive relaxation groups (Bensink et al. 1992;
McMurdo and Rennie 1993); supportive group psychotherapy (Goldwasser et al. 1987;
Szczepanska-Gieracha et al. 2014; Williams-Barnard and Lindell 1992); validation therapy (Tondi
et al. 2007; Toseland et al. 1997); cognitive or cognitive-behavioral group therapies (Abraham et
al. 1992; Zerhusen et al. 1991); focused visual imagery therapy (Abraham et al. 1997); and a
psychosocial activity intervention (Beck et al. 2002). Except in the investigations by Abraham et
al. (1992, 1997), patients were not selected on the basis of specific psychiatric symptoms or
syndromes but rather on the basis of age, cognitive status, or mobility.
Some of these studies reported improvements on measures of communication, behavior,
cognitive performance, mood, social withdrawal, physical function, somatic preoccupation, self-
esteem, perceived locus of control, quality of life, and life satisfaction. Case reports and
demonstration projects by experienced clinicians also documented the value of psychotherapy for
treating depressed nursing home residents (Leszcz et al. 1985; Ollech 2006; Sadavoy 1991).
Overall, there is a paucity of research on the outcomes of well-described psychotherapies among
nursing home residents who have well-characterized psychiatric disorders. Nevertheless, the
available evidence from nursing home research, considered together with outcomes of
psychotherapy for older adults in other clinical settings, suggests that psychotherapy should be
regarded as an important component of mental health treatment for the more cognitively intact
nursing home residents with depression.
Pharmacotherapy
Pharmacological treatments are commonly used in nursing homes for dementia and its associated
psychological and behavioral symptoms, and for depression. More comprehensive reviews of the
evidence for pharmacological treatment of neuropsychiatric symptoms of dementia specifically in
the nursing home setting are provided by Bharani and Snowden (2005) and Seitz et al. (2013).
Randomized, placebo-controlled clinical trials of psychotropic drugs conducted specifically in
nursing home populations are summarized in Table 25–2. Some of the earlier studies provided
evidence for the efficacy of antipsychotic drugs in managing agitation and related symptoms in
nursing home residents with dementia, but the effect sizes were often modest, and high placebo
response rates were common (Barnes et al. 1982; Schneider et al. 1990; Sunderland and Silver
1988). Subsequently, several multicenter, randomized, double-blind, placebo-controlled clinical
trials demonstrated efficacy of some of the atypical antipsychotic agents for the treatment of
psychotic symptoms and agitated behavior in nursing home residents with dementia. These
include published studies of risperidone (Brodaty et al. 2003; Katz et al. 1999), olanzapine
(Meehan et al. 2002; Street et al. 2000), quetiapine (Zhong et al. 2007), and aripiprazole (Mintzer
et al. 2007, Rappaport et al. 2009). Secondary analyses of data from the nursing home trials of
risperidone showed that the medication had antipsychotic effects and also had independent effects
on aggression or agitation. Other studies of atypical antipsychotic drugs in nursing home residents
with dementia failed to show statistically significant benefits on the a priori designated primary
outcome measures related to psychosis or behavioral disturbances (De Deyn et al. 1999, 2005;
Mintzer et al. 2006; Streim et al. 2008), although some of these studies found possible benefits on
secondary behavioral measures. Widespread interest in studying atypical antipsychotics for
treatment of elderly nursing home residents was partly attributable to the expectation that these
agents would be better tolerated than conventional antipsychotics in this population. For example,
early follow-up studies had suggested that risperidone may cause less tardive dyskinesia than do
typical antipsychotic agents (Jeste et al. 2000).
These controlled clinical trials have examined only the acute effects of treatment, typically for
6–12 weeks of treatment, and little is known about the effectiveness of treatment for longer
periods. However, there is evidence to suggest that the need for and benefit from antipsychotic
drug treatment changes over the course of months in nursing home patients with dementia. Several
double-blind, placebo-controlled studies of antipsychotic drug discontinuation demonstrated that
the majority of patients who had been receiving longer-term treatment could be withdrawn from
these agents without reemergence of psychosis or agitated behaviors (Bridges-Parlet et al. 1997;
Cohen-Mansfield et al. 1999; Ruths et al. 2004). These data are consistent with findings from
older discontinuation studies (Barton and Hurst 1966; Risse et al. 1987). Therefore, it is important
to periodically reevaluate the need for continuing antipsychotic drug treatment.
Since 2003, analyses of safety data from randomized controlled studies of atypical
antipsychotic drugs in elderly patients with dementia, including the aforementioned nursing home
studies, have revealed significantly increased risks of cerebrovascular adverse events and
mortality in this population. Although elevated risks were not found in every study, pooled
analyses showed that the rate of cerebrovascular adverse events (including stroke and transient
ischemic attacks) is greater than with placebo (U.S. Food and Drug Administration 2003). Most of
the affected individuals had known cerebrovascular risk factors prior to starting drug treatment.
These findings led to regulatory warnings in the United States, Canada, and the United Kingdom
regarding the safety of these drugs in elderly patients with dementia.
The U.S. Food and Drug Administration (FDA) also warns that elderly patients with dementia-
related psychosis who are treated with atypical antipsychotics have a risk of death of about 1.7
times greater than those treated with placebo (4.5% vs. 2.6%) and provides a reminder that
atypical antipsychotics do not have FDA approval for the treatment of patients with dementia-
related psychosis (U.S. Food and Drug Administration 2005). Consistent with this FDA warning,
a meta-analysis by Schneider et al. (2005), which examined results of 15 randomized controlled
trials, many of which were conducted using nursing home patients, found that the risk of mortality
was 3.5% for elderly patients treated with atypical antipsychotics versus 2.3% for patients treated
with placebo. Although no placebo-controlled trials of ziprasidone or clozapine are known to have
been conducted in elderly patients with dementia, it is reasonable to view the increased mortality
as a class effect. Wang et al. (2005) found a significantly higher adjusted risk of death for elderly
patients taking conventional antipsychotics compared with those taking atypical antipsychotic
medications, regardless of whether the patients had dementia or resided in a nursing home. The
authors suggested that conventional antipsychotic medications are at least as likely as atypical
agents to increase the risk of death in older adults. Subsequent comparisons with community-
dwelling populations revealed higher mortality rates among nursing home residents with dementia
who were newly started on antipsychotic drugs (Gill et al. 2007; Rochon et al. 2008).
TABLE 25–2. Randomized, placebo-controlled studies of the efficacy of psychotropic
medications in elderly nursing home residents
Study Medication and dosage Sample Efficacy measures Results and comments
(mg/day)
Beber 1965 Oxazepam (10–80) vs. N=100; mean age 79 Anxiety and Improvement in all
placebo years; nonpsychotic
with chronic brain
tension; parameters was
syndrome (n=28), depression, significantly
mixed lethargy, greater in
anxiety/depression
(n=26), anxiety autonomic oxazepam-treated
(n=43), or depression reactions; group than in
(n=3) irritability, placebo-treated
insomnia, group; 44 subjects
agitation, received
phobic concomitant
reactions treatment with
other drugs,
including
neuroleptics,
antidepressants,
hypnotics,
antiparkinsonian
agents, and
analgesics.
Barnes et al. Thioridazine N=53; mean age BPRS; SCAG; Total scores and
1982 (mean 62.5) vs. 83 years; NOSIE; CGI global ratings
loxapine (mean dementia and showed modest
10.5) vs. three or more efficacy with
placebo behavioral thioridazine and
symptoms loxapine, but
were not
statistically better
than placebo; high
placebo response
rate; significant
improvement in
anxiety,
excitement,
emotional lability,
and
uncooperativeness
in active
treatment groups,
but no significant
differences in
overall efficacy
between
thioridazine and
loxapine;
significant
improvement on
BPRS and SCAG
only in subjects
with high-severity
baseline scores.
Stotsky 1984 Thioridazine N=237 nursing Modified Ham- Thioridazine was
(10–200) vs. home patients; A; modified well tolerated,
diazepam (20– mean age 80 NOSIE; with few side
40) vs. placebo years; all global effects.
nonpsychotic evaluations Thioridazine-
with cognitive treated group
impairment, improved
emotional significantly more
lability, ADL than placebo
dysfunction group on all Ham-
and agitation, A items (74% vs.
anxiety, 42%) and global
depressed evaluations.
mood, or sleep Thioridazine-
disturbance treated group
(also studied improved
were 273 significantly more
patients on than diazepam
geriatric wards group on NOSIE
of state and global
hospitals) ratings. Insomnia
responded better
to diazepam, but
there was more
overall
improvement on
Ham-A rating
with thioridazine.
Dehlin et al. Alaproclate N=40; mean age Intellectual No difference in
1985 (400) 82 years; function; efficacy was
(serotonin primary motor found between
reuptake degenerative, function alaproclate and
inhibitor) vs. multi-infarct, (ADL); placebo. Severity
placebo or mixed emotional of dementia
dementia; not function ranged from mild
selected on (including to severe.
basis of depressive Behavioral
affective or symptoms); problems were
behavioral clinical global not described.
symptoms evaluation
Katz et al. Nortriptyline N=30 residents Ham-D*; Significant
1990 (mean 65.25) of nursing Geriatric improvement
vs. placebo home or Depression occurred in
congregate Scale; CGI patients treated
housing; mean with nortriptyline
age 84 years; compared with
major placebo on Ham-
depression D and CGI but
(Ham-D scores not on Geriatric
≥ 18) Depression Scale.
Location (in
nursing home vs.
congregate
housing) was not
significantly
related to
response. Trend
toward decreased
nortriptyline
response in
nursing home was
related to higher
levels of disability
and lower serum
albumin in
nursing home
patients.
Nyth and Citalopram (10– N=98; mean age GBS; CGI; Compared with
Gottfries 30; mean 25) 77.6 years; MADRS control subjects,
1990 vs. placebo primary Alzheimer’s
degenerative disease patients in
(Alzheimer’s citalopram group
disease) or had significant
multi-infarct reduction in
dementia irritability and
(vascular depressed mood
dementia) on GBS. From
baseline to week
4, Alzheimer’s
disease patients in
citalopram group
improved
significantly on
MADRS and on
GBS emotional
blunting,
confusion,
irritability,
anxiety, fear-
panic, depressed
mood, and
restlessness. No
treatment benefits
were found in
vascular dementia
patients. Placebo
group worsened
on CGI. Few
adverse effects
occurred, with no
drug-placebo
differences.
Finkel et al. Thiothixene N=33; mean age CMAI; MMSE; Thiothixene-treated
1995 (0.25–18; 85 years; GDS; ADLs group had
mean 4.6) vs. dementia with significantly
placebo agitated or greater reduction
aggressive in agitation than
behavior placebo group
after 11 weeks of
treatment and 6
weeks after
crossover from
placebo; no
between-group
differences were
seen on MMSE,
GDS, and ADLs.
Tariot et al. Carbamazepine N=51; dementia BPRS; CGI; Carbamazepine-
1998 (modal dose with agitation agitation; treated group had
300; mean aggression; significantly
serum level 5.3 cognition; greater
μg/mL) vs. functional improvement on
placebo status; staff BPRS compared
time with placebo
group at 6 weeks.
Global
improvement
occurred in 77%
of patients taking
carbamazepine
and in 22% of
those taking
placebo.
Secondary
analyses showed
that improvement
was attributable to
decreased
agitation and
aggression.
Nurses reported
perception of
decreased time
required to
manage agitation
in carbamazepine
group.
Significantly
more adverse
events were seen
with
carbamazepine
(59%) than with
placebo (29%).
De Deyn et al. Risperidone N=344; BEHAVE-AD*; No significant
1999 (0.5–4; mean Alzheimer’s, CMAI; CGI; difference in
1.1) vs. vascular, and MMSE response rates
haloperidol mixed were seen on
(0.5–4; mean dementia BEHAVE-AD
1.2) vs. (MMSE mean between
placebo; 12 score 8.7) risperidone and
weeks placebo,
BEHAVE-AD
total and
aggression
subscale and
CMAI aggression
item scores were
lower and CGI
was significantly
greater with
risperidone than
with placebo; post
hoc analysis
showed
significantly
greater reduction
in BEHAVE-AD
aggression scores
with risperidone
than with
haloperidol.
Somnolence was
greater with
risperidone
(12.2%) than with
placebo (4.4%).
Extrapyramidal
symptoms were
significantly
greater with
haloperidol (22%)
than with
risperidone (15%)
or placebo (11%).
Slight but
significant decline
was seen in
MMSE scores in
haloperidol group.
Response defined
as ≥ 30%
reduction in
BEHAVE-AD
score
Katz et al. Risperidone N=625; mean BEHAVE-AD*; Significantly
1999 (0.5–2) vs. age 82.7 years; CMAI; CGI; greater reductions
placebo; 12 psychotic and MMSE; occurred in
weeks behavioral FAST; PSMS BEHAVE-AD
symptoms and total scores and in
Alzheimer’s Aggressiveness
disease, and Psychosis
vascular subscale scores
dementia, or with risperidone 1
mixed mg and 2 mg than
Alzheimer’s with placebo.
disease and Response rates
vascular were significantly
dementia greater with
(MMSE mean risperidone 1 mg
score 6.6) (45%) and 2 mg
(50%) than with
placebo (33%).
More
extrapyramidal
symptoms and
somnolence were
seen with 2 mg
than with 1 mg of
risperidone.
Optimal dosage
for nursing home
patients with
severe dementia
appears to be 1
mg.
Response defined
as ≥ 50%
reduction in
BEHAVE-AD
score
Magai et al. Sertraline vs. N=31; all women Depression; Both groups were
2000 placebo with late-stage facial affect improved at 8
Alzheimer’s weeks; sertraline
disease and had no significant
depression benefits over
placebo. “Knit-
brow” facial
response
approached
significance for
treatment × time
effect.
Street et al. Olanzapine (5, N=206; mean NPI-NH core* Olanzapine 5 mg
2000 10, 15) vs. age 83.8 years; and total and 10 mg
placebo; 6 Alzheimer’s (including produced
weeks disease with Occupational significant
psychotic Disruptiveness improvement in
and/or scores); summary
behavioral BPRS; MMSE measures of
symptoms agitation,
(MMSE mean aggression, and
score 6.9) psychosis.
Olanzapine 5 mg
significantly
reduced
disruptive effects
on caregivers
compared with
placebo.
Olanzapine was
associated with
somnolence and
gait disturbance
but not with
increased
extrapyramidal
symptoms, central
anticholinergic
effects, or
cognitive
impairment
compared with
placebo; 18% of
placebo group and
44% of
olanzapine group
dropouts were due
to adverse events.
Olin et al. Carbamazepine N=21; BPRS*; CGI-C; Both groups
2001 (mean 388) vs. Alzheimer’s Ham-D improved on CGI-
placebo; 6 dementia C (56% on
weeks (MMSE mean carbamazepine,
score 6.0) 58% on placebo),
but no significant
differences were
seen on BPRS,
CGI, or Ham-D.
Adverse events
were mild and
similar in drug
and placebo.
Porsteinsson Divalproex N=56; mean age BPRS*; OAS; Divalproex group
et al. 2001 (mean 826; 85 years; BRSD; showed
mean serum probable or CMAI; CGI-C significant
level 45.4 possible improvement on
μg/mL) vs. Alzheimer’s BPRS at 6 weeks
placebo; 6 disease, compared with
weeks vascular placebo only in a
dementia, or secondary
mixed analysis after
dementia with adjustment for
agitation several
(MMSE mean covariates; 68%
score 6.8) of divalproex
group and 52% of
placebo group had
reduced agitation
on CGI (NS).
Significantly
more side effects
(generally mild)
were reported in
divalproex than in
placebo group
(68% vs. 33%),
but sedation was
39% with
divalproex vs.
11% with
placebo.
Tariot et al. Donepezil (5– N=208; mean NPI-NH; CDR- Both groups
2001a 10) vs. placebo age 85.7 years; SB; MMSE; improved on NPI-
probable or PSMS NH, with no
possible significant
Alzheimer’s difference
disease or between
Alzheimer’s donepezil and
disease with placebo.
cerebrovascular Significantly
disease greater
(MMSE mean improvement was
score 14.4) seen with
donepezil than
with placebo on
CDR-SB at week
24 and on MMSE
at weeks 8, 16,
and 20, but not on
PSMS.
Improvement was
not influenced by
advanced age. No
difference in
adverse events
was found for
drug vs. placebo.
Tariot et al. Divalproex N=172; BRMS*; BPRS; Study was
2001b (mean 1,000) Alzheimer’s CMAI; CGI-C discontinued early
vs. placebo; 6 disease, due to
weeks vascular or significantly
mixed greater rate of
dementia with adverse events
symptoms of with divalproex,
mania (MMSE especially
mean score somnolence.
7.4)
Burrows et al. Paroxetine (10– N=24; age 80 CSD; Ham-D No significant
2002 30) vs. placebo years and benefit was found
older; with paroxetine
for treatment of
nonmajor nonmajor
depression depression.
Meehan et al. Olanzapine (2.5– N=204; recruited PANSS-EC*, Significantly
2002 5 IM single from nursing ACES greater mean
dose) vs. home and reduction in
lorazepam vs. hospital sites; PANSS-EC scores
placebo; 24 Alzheimer’s, was seen 2 hours
hours vascular, and postdose for
mixed olanzapine 2.5
Alzheimer’s- and 5 mg vs.
vascular placebo; no
dementia difference was
(MMSE mean found between
score 11.8) olanzapine and
lorazepam;
adverse events
were not
significantly
different across
groups.
Brodaty et al. Risperidone N=345; mean CMAI Compared with
2003 (0.5–2; mean age 83 years; aggression*; placebo group,
0.95) vs. Alzheimer’s, CMAI non- risperidone group
placebo; 12 vascular, or aggression had significant
weeks mixed subscales; improvement on
dementia, with BEHAVE- CMAI total
aggressive AD; CGI-S; aggression and
behavior CGI-C nonaggressive
(MMSE mean agitation scores,
score 5.5) on BEHAVE-AD
total scores and
psychosis
subscale, and on
CGI severity and
change scores.
Risperidone
group had more
somnolence, falls,
urinary tract
infections, and
cerebrovascular
adverse events,
but no drug-
placebo difference
was found in
extrapyramidal
side effects.
De Deyn et al. Olanzapine (1, N=652; NPI-NH No significant
2005 2.5, 5, or 7.5) Alzheimer’s psychosis difference was
vs. placebo; 10 dementia with subscale*; seen between any
weeks psychosis CGI-C*; NPI dose of
(MMSE mean total, olanzapine and
score 13.7) individual placebo on
item, and primary
occupational outcomes. More
disruptiveness weight gain
scores occurred with
olanzapine, but no
difference was
found in
anticholinergic or
extrapyramidal
adverse effects or
dropout rates due
to adverse effects.
Tariot et al. Divalproex (800) N=153; probable BPRS agitation The trial was
2005 vs. placebo; 6 or possible factor*; BPRS completed by
weeks Alzheimer’s total, CGI-C, 72% of enrolled
disease with CMAI subjects. No
agitation significant drug-
placebo
differences were
found on primary
or secondary
outcome
measures.
Mintzer et al. Risperidone (1 N=473; BEHAVE-AD Both groups
2006 or 1.5; mean Alzheimer’s psychosis improved, with no
1.03) vs. dementia with subscale*; significant
placebo; 8 psychosis CGI-C* differences on
weeks (MMSE mean primary outcome
score 12.4) measures.
Subgroup analysis
showed patients
with MMSE < 10
had significantly
greater
improvement on
CGI-C with
risperidone. No
difference in
discontinuation
rates for
risperidone and
placebo groups
were found, but
risperidone group
had higher rates
of somnolence
(16.2% vs. 4.6%)
and death (3.8%
and 2.5%).
Winblad et al. Donepezil (10) N=248; severe SIB*; ADCS- Significant
2006 vs. placebo; 26 Alzheimer’s ADL severe*; improvement in
weeks disease MMSE; NPI; SIB scores and
(MMSE score CGI-I less decline in
range 1–10) ADL function
were seen in
donepezil group
at 6 months. CGI-
I significant for
donepezil in
completer
analysis only. No
significant
behavioral
benefits were
seen. Rates of
adverse events
were comparable
in drug and
placebo groups,
but more patients
in donepezil
group
discontinued
treatment because
of adverse events.
Mintzer et al. Aripiprazole (2, N=487; mean NPI-NH Aripiprazole 10 mg
2007 5, or 10 fixed age 82.5 years; psychosis group had
dose) vs. Alzheimer’s subscale*; significantly
placebo; 10 dementia with NPI-NH total; greater
weeks psychosis CGI-S; CGI-I; improvement than
(MMSE mean BPRS placebo group on
score 12.4) psychosis, NPI-NH
core and total psychosis scores
score; CMAI; and response
response rates, BPRS core
defined as ≥ and total scores,
50% decrease and CMAI scores.
in NPI score Aripiprazole 5 mg
group showed
significantly
greater
improvement only
on BPRS and
CMAI.
Aripiprazole 2 mg
was not
efficacious. There
was a dose-
dependent
occurrence of
cerebrovascular
adverse events in
the aripiprazole
groups. Death
rates in the
placebo, 2, 5, and
10 mg groups
were 3%, 3%,
2%, and 7%,
respectively,
although the
differences were
not statistically
significant.
Zhong et al. Quetiapine (100 N=333; PANSS-EC*; No significant
2007 or 200) vs. Alzheimer’s CGI-C; NPI- differences were
placebo; 10 and mixed NH; CMAI found between
weeks dementia quetiapine and
placebo by LOCF
analysis of
PANSS-EC, NPI-
NH, or CMAI
scores at
endpoint.
However,
quetiapine 200
mg (but not 100
mg) was
significantly
better than
placebo on CGI-C
scores and CGI-C
response rates
using both LOCF
and observed case
(OC) analyses and
on PANSS-EC
using OC
analysis. No
differences were
seen in incidence
of postural
hypotension, falls,
and
cerebrovascular
adverse events.
There were more
deaths with
quetiapine,
although rates
were not
statistically
different from
those with
placebo.
Response defined
as ≥ 40%
reduction in
PANSS-EC, or
much or very
much improved
on CGI-C
Streim et al. Aripiprazole (2, N=256; mean NPI-NH No significant
2008 5, 10, or 15 age 83 years; psychosis difference was
flexible dose Alzheimer’s subscale*; found between
titration) vs. dementia with CGI-S*; NPI- aripiprazole and
placebo; 10 psychosis NH total; placebo on NPI-
weeks (MMSE mean BPRS NH psychosis or
score 13.6) psychosis, CGI-S at
core, and total endpoint.
scores; CMAI; However,
CSDD; NPI significantly
caregiver greater
distress; improvement
ADCS ADL occurred in the
scores; CGI-I aripiprazole group
on secondary
outcomes: NPI
and BPRS total
scores, CMAI,
CGI-I, CSDD.
More somnolence
was seen with
aripiprazole
(14%) than with
placebo (4%);
other adverse
effects were
similar for
aripiprazole and
placebo, with low
rate of
extrapyramidal
side effects in
both groups.
Response defined
as ≥ 50%
decrease in NPI
scores
Rappaport et Aripiprazole (5– N=129; mean PANSS-EC; Greater
al. 2009 15 IM) vs. age 80 years; ACES improvements
placebo (IM); Alzheimer’s, were seen on
24 hours vascular, mixed PANSS-EC scores
dementia with with aripiprazole
agitation 10 and 15 mg
(MMSE mean than with placebo;
score 13.3) double the
incidence of
adverse events
were seen with
aripiprazole
(50%–60%) than
with placebo
(32%).
Sommer et al. Oxcarbazepine N=103; mean NPI-NH No significant
2009 (300–900) vs. age 83.5 years; agitation and between-group
placebo; 8 Alzheimer’s, aggression differences were
weeks vascular, or subscores; found on any
mixed NPI caregiver outcome
dementia with burden scale; measures.
agitation and BARS
aggression
Note. ACES=Agitation-Calmness Evaluation Scale (Copyright ©1998, Eli Lilly & Company; all rights reserved); ADCS-
ADL=Alzheimer’s Disease Cooperative Study–Activities of Daily Living Inventory (Galasko et al. 1997); ADL=activities of daily
living; BARS=Brief Agitation Rating Scale (Finkel et al. 1993); BEHAVE-AD=Behavioral Pathology in Alzheimer’s Disease
(Reisberg et al. 1996); BPRS=Brief Psychiatric Rating Scale (Overall and Gorham 1962); BRMS=Bech-Rafaelsen Mania Scale
(Bech et al. 1979); BRSD=Behavior Rating Scale for Dementia (by the Consortium to Establish a Registry for Dementia [Mack et
al. 1999]); CDR-SB=Clinical Dementia Rating (Nursing Home Version)–Sum of the Boxes (Hughes et al. 2000); CGI=Clinical
Global Impression Scale (Guy 1976); CGI-C=Clinical Global Impression of Change (Guy 1976; Schneider et al. 1997); CGI-
I=Clinical Global Impression of Improvement (Guy 1976); CGI-S=Clinical Global Impression of Severity (Guy 1976);
CMAI=Cohen-Mansfield Agitation Inventory (Cohen-Mansfield et al. 1989); CSDD=Cornell Scale for Depression in Dementia
(Alexopoulos et al. 1988); FAST=Functional Assessment Staging (Reisberg 1988); GBS=Gottfries-Brane-Steen Geriatric Rating
Scale (Gottfries et al. 1982); GDS=Global Deterioration Scale (Reisberg et al. 1988); Geriatric Depression Scale (Yesavage et al.
1982); Ham-A=Hamilton Anxiety Scale (Hamilton 1959); Ham-D=Hamilton Rating Scale for Depression (Hamilton 1960);
IM=intramuscular; LOCF=last observation carried forward; MADRS=Montgomery-Åsberg Depression Rating Scale (Montgomery
and Åsberg 1979); MMSE=Mini-Mental State Examination (Folstein et al. 1975); NOSIE=Nurses’ Observation Scale for Inpatient
Evaluation (Honigfeld et al. 1966); NPI-NH=Neuropsychiatric Inventory–Nursing Home Version (Wood et al. 2000);
NS=nonsignificant; OAS=Overt Aggression Scale (Yudofsky et al. 1986); PANSS-EC=Positive and Negative Symptom Scale–
Excited Component (Kay et al. 1987); PSMS=Physical Self-Maintenance Scale (Lawton and Brody 1969); SCAG=Sandoz Clinical
Assessment–Geriatric (Shader et al. 1974); SIB=self-injurious behavior.
In light of the concerns about risks of antipsychotic drugs in elderly nursing home residents
with dementia, experts in the field have suggested that nonpharmacological approaches should be
considered first when treating noncognitive behavioral symptoms (Blazer 2013). For those nursing
home patients whose behavioral symptoms do not respond to nonpharmacological interventions,
the decision to use an atypical antipsychotic should be based on a careful assessment of each
individual’s risk-benefit profile.
Six randomized clinical trials evaluated the efficacy of mood-stabilizing anticonvulsant drugs
for the treatment of agitation and aggression in nursing home residents. The first was a study of
carbamazepine that showed it to be effective for agitation and aggression but not for psychotic
symptoms such as delusions and hallucinations (Tariot et al. 1998). In this study, nursing reports
indicated that less staff time was required for patient care in the group treated with carbamazepine.
Another trial of carbamazepine found high rates of improvements in both drug- and placebo-
treated patients, with nonsignificant between-group differences (Olin et al. 2001). A subsequent
study of oxcarbazepine failed to show efficacy for treatment of agitation and aggression (Sommer
et al. 2009). Several placebo-controlled studies evaluated divalproex with few encouraging results;
one trial was discontinued before completion because of adverse effects among the drug treatment
group. Overall, these studies failed to provide evidence for efficacy in reducing agitated behavior
(Porsteinsson et al. 2001; Tariot et al. 2001b, 2005). It is noteworthy that the tolerability of
divalproex was limited by somnolence, weakness, and diminished oral intake in this population of
elderly nursing home subjects with dementia.
Acetylcholinesterase inhibitors have been shown to delay the decline in cognitive function in
patients with mild to severe Alzheimer’s disease. Although few studies have been conducted
specifically in nursing home samples, the 2004 NNHS found that of the 49.1% of survey
participants with dementia, 30% were receiving cholinesterase inhibitors (Seitz et al. 2009). One
randomized clinical trial of donepezil in nursing home residents showed effects on cognitive
performance that were comparable with those observed in less impaired outpatients (Tariot et al.
2001a). A subsequent study demonstrated that donepezil improves cognition and preserves
function in Alzheimer’s disease patients with severe dementia who are residing in nursing homes
(Winblad et al. 2006). These studies also examined the effects of donepezil on behavioral
disturbances, as a secondary outcome measure, and did not find significant benefits. Ballard et al.
(2005) compared the effects of quetiapine, rivastigmine, and placebo on agitation and cognition in
nursing home patients with dementia and found that neither drug is effective for the treatment of
agitation and that quetiapine is associated with significantly greater cognitive decline. One study
by Tariot et al. (2004) reported significant improvement on Neuropsychiatric Inventory scores in
patients for whom memantine rather than placebo was added to stable doses of donepezil, but
memantine has not yet been studied prospectively in the nursing home setting. Survival analyses
in an observational study suggest that the addition of memantine to a cholinesterase inhibitor may
delay the time to nursing home placement (Lopez et al. 2009).
There have been only five randomized controlled clinical trials evaluating the effects of
antidepressants in nursing home residents. The first study found no differences in intellectual,
emotional, or functional status in residents with dementia who were treated with alaproclate
(Dehlin et al. 1985). The second study, which was also placebo controlled, showed a positive
response to nortriptyline for treatment of major depression in a long-term-care population with
high levels of medical comorbidity (Katz et al. 1990). In the third study, patients were randomized
to receive regular or low-dose nortriptyline, and significant plasma level–response relationships
were demonstrated in cognitively intact patients (Streim et al. 2000). These findings confirmed the
validity of the diagnosis of depression in nursing home residents in the context of significant
medical comorbidity and disability. However, in patients with dementia, the plasma level–
response relationship was significantly different, suggesting that the depression occurring in
dementia might be a treatment-relevant subtype of depression or a distinct disorder. The fourth, a
controlled antidepressant trial in nursing home residents with late-stage Alzheimer’s disease,
showed no significant benefits of sertraline over placebo (Magai et al. 2000). In the fifth, a
randomized placebo-controlled trial of paroxetine in very old residents with non-major depression,
no significant benefit was found for drug over placebo (Burrows et al. 2002).
Available open-label studies of the efficacy of selective serotonin reuptake inhibitors (SSRIs)
in nursing home residents with depression have had mixed results, some consistent with the
findings of Magai et al. (2000), suggesting that SSRIs may be less effective for depression in
patients with dementia than in those who are cognitively intact (Oslin et al. 2000; Rosen et al.
2000; Trappler and Cohen 1996, 1998). Findings from nonrandomized open-label antidepressant
trials in depressed nursing home residents are summarized in a review by Boyce et al. (2012).
Although the SSRIs might be expected to be well tolerated by frail elderly nursing home
patients because of their side-effect profiles, there is evidence that these drugs can cause serious
adverse events in this population. Thapa et al. (1998) demonstrated that the use of SSRIs was
associated with a nearly twofold increase in the risk of falls among nursing home residents,
comparable with the risk found with tricyclic antidepressant drugs. Investigators in the United
Kingdom reported that antidepressant use was associated with better physical functioning but also
with greater frequency of falls among residential care patients (Arthur et al. 2002). A randomized,
double-blind comparison trial found that venlafaxine was less well tolerated than sertraline in frail
nursing home patients without conferring more treatment benefits, as might be expected from an
agent with mixed serotonergic and noradrenergic effects (Oslin et al. 2003).
Physical Restraints
The 1977 NNHS showed that 25% of 1.3 million U.S. nursing home residents were restrained by
geriatric chairs, cuffs, belts, or similar devices, primarily in an attempt to control behavioral
symptoms (National Center for Health Statistics 1979). Other early surveys demonstrated rates of
restraint as high as 85%. Patient factors predicting the use of restraints, in addition to agitation and
behavior problems, include age, cognitive impairment, risk of injuries to self (e.g., from falls) or
others (e.g., from combative behavior), physical frailty, the presence of monitoring or treatment
devices, and the need to promote body alignment. Institutional and systemic factors associated
with restraint use include pressure to avoid litigation, staff attitudes, insufficient staffing, and the
availability of restraint devices. Potential adverse effects include an increased risk of falls and
other injuries (Capezuti et al. 1996) as well as functional decline, skin breakdown, physiological
effects of immobilization stress, disorganized behavior, and demoralization. Although mechanical
restraints have frequently been used in attempts to control agitation, they do not in fact decrease
behavioral disturbances (Werner et al. 1989), and cross-national studies indicated that nursing
home residents can be managed without such measures (Cape 1983; Evans and Strumpf 1989;
Innes and Turman 1983).
Conclusion
The high prevalence of mental health problems among nursing home residents underscores the
importance of psychiatric care in nursing homes. Psychiatry in U.S. nursing homes has evolved
substantially over several decades, with changes driven by a combination of factors, including
scientific knowledge, availability of new treatments, evolving federal regulations, measurement-
based care, public dissemination of quality measures, and recognition of the need for integrated
approaches to the delivery of mental health services.
Key Points
• According to the 2004 National Nursing Home Survey, 4% of Americans ages 65 years and
older—1.5 million people—resided in 16,100 long-term-care facilities.
• Epidemiological studies have consistently shown that 80%–94% of nursing home residents
have diagnosable psychiatric disorders. These prevalence data suggest that nursing homes are
de facto neuropsychiatric institutions, although they were not originally intended for this
purpose.
• Since the implementation of federal nursing home regulations in the early 1990s, the rate of
antipsychotic medication use has declined, and the use of antidepressants has increased.
However, approximately half of nursing home residents who are receiving antidepressant
medications continue to have symptoms of depression.
• Since 2003, analyses of safety data from randomized controlled studies of atypical
antipsychotic drugs in elderly patients with dementia, including nursing home studies, have
revealed significantly increased risks of cerebrovascular adverse events and mortality in this
population. This finding had led to initiatives by the Centers for Medicare and Medicaid to
improve the quality of dementia care in nursing homes, including the goal of reducing
antipsychotic drug use.
• Although mechanical restraints have frequently been used in attempts to control agitation, they
do not decrease behavioral disturbances, and cross-national studies have indicated that it is
possible to manage nursing home residents more safely without such measures.
• The social environment within which care is provided can have a significant effect on nursing
home residents; environmental design should be viewed as a component of mental health care.
References
Abraham IL, Neundorfer MM, Currie LJ: Effects of group interventions on cognition and
depression in nursing home residents. Nurs Res 41(4):196–202, 1992 1383947
Abraham IL, Onega LL, Reel SJ, et al: Effects of cognitive group interventions on depressed frail
nursing home residents, in Depression in Long Term and Residential Care: Advances in
Research and Treatment. Edited by Rubinstein RL, Lawton MP. New York, Springer, 1997,
pp 154–168
Ahmed A, Lefante CM, Alam N: Depression and nursing home admission among hospitalized
older adults with coronary artery disease: a propensity score analysis. Am J Geriatr Cardiol
16(2):76–83, 2007 17380615
Alessi CA, Yoon EJ, Schnelle JF, et al: A randomized trial of a combined physical activity and
environmental intervention in nursing home residents: do sleep and agitation improve? J Am
Geriatr Soc 47(7): 784–791, 1999 10404920
Alexopoulos GS, Abrams RC, Young RC, Shamoian CA: Cornell Scale for Depression in
Dementia. Biol Psychiatry 23(3):271–284, 1988 3337862
American Association for Geriatric Psychiatry, American Geriatrics Society, American Psychiatric
Association: Psychotherapeutic medications in the nursing home. J Am Geriatr Soc 40:946–
949, 1992 1512392
American Geriatrics Society, American Association for Geriatric Psychiatry: The American
Geriatrics Society and American Association for Geriatric Psychiatry recommendations for
policies in support of quality mental health care in U.S. nursing homes. J Am Geriatr Soc
51(9):1299–1304, 2003 12919244
American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, 3rd
Edition. Washington, DC, American Psychiatric Association, 1980
American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, 3rd
Edition, Revised. Washington, DC, American Psychiatric Association, 1987
American Society of Consultant Pharmacists: Fact Sheet. Alexandria, VA, American Society of
Consultant Pharmacists, September 2000
Ames D: Depression among elderly residents of local-authority residential homes: its nature and
the efficacy of intervention. Br J Psychiatry 156:667–675, 1990 2095943
Ames D: Epidemiological studies of depression among the elderly in residential and nursing
homes. Int J Geriatr Psychiatry 6(6):347–354, 1991
Ames D, Ashby D, Mann AH, Graham N: Psychiatric illness in elderly residents of Part III homes
in one London Borough: prognosis and review. Age Ageing 17(4):249–256, 1988 3262986
Arthur A, Matthews R, Jagger C, et al: Factors associated with antidepressant treatment in
residential care: changes between 1990 and 1997. Int J Geriatr Psychiatry 17(1):54–60, 2002
11802231
Aschbrenner KA, Cai S, Grabowski DC, et al: Medical comorbidity and functional status among
adults with major mental illness newly admitted to nursing homes. Psychiatr Serv
62(9):1098–1100, 2011a 21885592
Aschbrenner K, Grabowski DC, Cai S, et al: Nursing home admissions and long-stay conversions
among persons with and without serious mental illness. J Aging Soc Policy 23(3):286–304,
2011b 21740203
Ashby D, Ames D, West CR, et al: Psychiatric morbidity as predictor of mortality for residents of
local authority homes for the elderly. Int J Geriatr Psychiatry 6(8):567–575, 1991
Avorn J, Langer E: Induced disability in nursing home patients: a controlled trial. J Am Geriatr
Soc 30(6):397–400, 1982 7077022
Avorn J, Dreyer P, Connelly K, et al: Use of psychoactive medication and the quality of care in
rest homes. Findings and policy implications of a statewide study. N Engl J Med 320(4):227–
232, 1989 2911307
Avorn J, Soumerai SB, Everitt DE, et al: A randomized trial of a program to reduce the use of
psychoactive drugs in nursing homes. N Engl J Med 327(3):168–173, 1992 1608408
Baines S, Saxby P, Ehlert K: Reality orientation and reminiscence therapy. A controlled cross-over
study of elderly confused people. Br J Psychiatry 151:222–231, 1987 3318991
Baker FM, Miller CL: Screening a skilled nursing home population for depression. J Geriatr
Psychiatry Neurol 4(4):218–221, 1991 1789910
Ballard CG, Margallo-Lana M, Fossey J, et al: A 1-year follow-up study of behavioral and
psychological symptoms in dementia among people in care environments. J Clin Psychiatry
62(8):631–636, 2001 11561936
Ballard CG, Margallo-Lana M, Juszczak E, et al: Quetiapine and rivastigmine and cognitive
decline in Alzheimer’s disease: randomized double blind placebo controlled trial. BMJ
330(7496):874, 2005 15722369
Banziger G, Roush S: Nursing homes for the birds: a control-relevant intervention with bird
feeders. Gerontologist 23:527–531, 1983
Barca ML, Engedal K, Laks J, et al: A 12 months follow-up study of depression among nursing-
home patients in Norway. J Affect Disord 120(1–3):141–148, 2010 19467560
Barnes RF, Raskind MA: DSM-III criteria and the clinical diagnosis of dementia: a nursing home
study. J Gerontol 36(1):20–27, 1981 7451830
Barnes R, Veith R, Okimoto J, et al: Efficacy of antipsychotic medications in behaviorally
disturbed dementia patients. Am J Psychiatry 139(9):1170–1174, 1982 7114310
Bartels SJ, Horn SD, Smout RJ, et al: Agitation and depression in frail nursing home elderly
patients with dementia: treatment characteristics and service use. Am J Geriatr Psychiatry
11(2):231–238, 2003 12611753
Barton R, Hurst L: Unnecessary use of tranquilizers in elderly patients. Br J Psychiatry
112(491):989–990, 1966 5339369
Beber CR: Management of behavior in the institutionalized aged. Dis Nerv Syst 26(9): 591–595,
1965 5319033
Bech P, Bolwig TG, Kramp P, et al: The Bech-Rafaelsen Mania Scale and the Hamilton
Depression Scale. Acta Psychiatr Scand 59(4):420–430, 1979 433633
Beck AT, Ward CH, Mendelson M, et al: An inventory for measuring depression. Arch Gen
Psychiatry 4:561–571, 1961 13688369
Beck CK, Vogelpohl TS, Rasin JH, et al: Effects of behavioral interventions on disruptive
behavior and affect in demented nursing home residents. Nurs Res 51(4):219–228, 2002
12131234
Beers M, Avorn J, Soumerai SB, et al: Psychoactive medication use in intermediate-care facility
residents. JAMA 260(20):3016–3020, 1988 2903260
Bensink GW, Godbey KL, Marshall MJ, et al: Institutionalized elderly: relaxation, locus of
control, self-esteem. J Gerontol Nurs 18(4):30–36, 1992
Berg A, Sadowski K, Beyrodt M, et al: Snoezelen, structured reminiscence therapy and 10-
minutes activation in long term care residents with dementia (WISDE): study protocol of a
cluster randomized controlled trial. BMC Geriatr 10:5, 2010 20113526
Berrios GE, Brook P: Delusions and the psychopathology of the elderly with dementia. Acta
Psychiatr Scand 72(3):296–301, 1985 4072730
Bharani N, Snowden M: Evidence-based interventions for nursing home residents with dementia-
related behavioral symptoms. Psychiatr Clin North Am 28(4):985–1005, x, 2005 16325737
Bharucha AJ, Dew MA, Miller MD, et al: Psychotherapy in long-term care: a review. J Am Med
Dir Assoc 7(9):568–580, 2006 17095422
Blazer DG: Antipsychotics and dementia: where do we go from here? J Am Med Dir Assoc
14(4):307–308, 2013 23415842
Blazer D, Williams CD: Epidemiology of dysphoria and depression in an elderly population. Am J
Psychiatry 137(4):439–444, 1980 7361929
Borson S, Liptzin B, Nininger J, et al: Psychiatry and the nursing home. Am J Psychiatry
144(11):1412–1418, 1987 3314537
Borson S, Loebel JP, Kitchell M, et al: Psychiatric assessments of nursing home residents under
OBRA-87: should PASARR be reformed? Pre-Admission Screening and Annual Review. J
Am Geriatr Soc 45(10): 1173–1181, 1997 9329477
Boyce RD, Hanlon JT, Karp JF, et al: A review of the effectiveness of antidepressant medications
for depressed nursing home residents. J Am Med Dir Assoc 13(4):326–331, 2012 22019084
Bridges-Parlet S, Knopman D, Steffes S: Withdrawal of neuroleptic medications from
institutionalized dementia patients: results of a double-blind, baseline-treatment-controlled
pilot study. J Geriatr Psychiatry Neurol 10(3):119–126, 1997 9322135
Briesacher BA, Limcangco MR, Simoni-Wastila L, et al: The quality of antipsychotic drug
prescribing in nursing homes. Arch Intern Med 165(11):1280–1285, 2005 15956008
Brodaty H, Ames D, Snowdon J, et al: A randomized placebo-controlled trial of risperidone for
the treatment of aggression, agitation, and psychosis of dementia. J Clin Psychiatry
64(2):134–143, 2003 12633121
Brown MN, Lapane KL, Luisi AF: The management of depression in older nursing home
residents. J Am Geriatr Soc 50(1): 69–76, 2002 12028249
Buck JA: Psychotropic drug practice in nursing homes. J Am Geriatr Soc 36(5):409–418, 1988
3283198
Buijck BI, Zuidema SU, Spruit-van Eijk M, et al: Neuropsychiatric symptoms in geriatric patients
admitted to skilled nursing facilities in nursing homes for rehabilitation after stroke: a
longitudinal multicenter study. Int J Geriatr Psychiatry 27(7):734–741, 2012 21932248
Burns BJ, Taube CA: Mental health services in general medical care and in nursing homes, in
Mental Health Policy for Older Americans: Protecting Minds at Risk. Edited by Fogel BS,
Furino A, Gottlieb GL. Washington, DC, American Psychiatric Press, 1990, pp 63–84
Burns BJ, Larson DB, Goldstrom ID, et al: Mental disorder among nursing home patients:
preliminary findings from the National Nursing Home Survey Pretest. Int J Geriatr Psychiatry
3(1):27–35, 1988
Burrows AB, Salzman C, Satlin A, et al: A randomized, placebo-controlled trial of paroxetine in
nursing home residents with non-major depression. Depress Anxiety 15(3):102–110, 2002
12001178
Buttar AB, Mhyre J, Fries BE, et al: Six-month cognitive improvement in nursing home residents
with severe cognitive impairment. J Geriatr Psychiatry Neurol 16(2): 100–108, 2003
12801160
Cape RD: Freedom from restraint. Gerontologist 23:217, 1983
Capezuti E, Evans L, Strumpf N, et al: Physical restraint use and falls in nursing home residents. J
Am Geriatr Soc 44(6):627–633, 1996 8642150
Castle NG, Shea D: Institutional factors of nursing homes that predict the provision of mental
health services. J Ment Health Adm 24(1):44–54, 1997 9033155
Castle NG, Fogel B, Mor V: Risk factors for physical restraint use in nursing homes: pre- and
post-implementation of the Nursing Home Reform Act. Gerontologist 37(6):737–747, 1997
9432990
Centers for Medicare and Medicaid Services: Minimum Data Set 3.0. 2006. Available at:
http://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-
Instruments/NursingHomeQualityInits/downloads/MDS30Draft.pdf. Accessed June 12, 2014.
Centers for Medicare and Medicaid Services: MDS 3.0 RAI Manual. 2013. Available at:
http://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-
Instruments/NursingHomeQualityInits/MDS30RAIManual.html. Accessed June 12, 2014.
Centers for Medicare and Medicaid Services: State Operations Manual, Appendix PP: Guidance to
Surveyors for Long Term Care Facilities. Baltimore, MD, Centers for Medicare and
Medicaid, Rev 107, 04-04-14. Baltimore, MD, Centers for Medicare and Medicaid. 2014.
Available at: https://www.cms.gov/Regulations-and-
Guidance/Guidance/Manuals/downloads/som107ap_pp_guidelines_ltcf.pdf. Accessed June
12, 2014.
Chahine LM, Bijlsma A, Hospers AP, et al: Dementia and depression among nursing home
residents in Lebanon: a pilot study. Int J Geriatr Psychiatry 22(4):283–285, 2007 16977675
Chandler JD, Chandler JE: The prevalence of neuropsychiatric disorders in a nursing home
population. J Geriatr Psychiatry Neurol 1(2):71–76, 1988 3266998
Chao SY, Liu HY, Wu CY, et al: The effects of group reminiscence therapy on depression, self
esteem, and life satisfaction of elderly nursing home residents. J Nurs Res 14(1):36–45, 2006
16547904
Cody M, Beck C, Svarstad BL: Challenges to the use of nonpharmacologic interventions in
nursing homes. Psychiatr Serv 53(11):1402–1406, 2002 12407267
Cohen-Mansfield J: Agitated behaviors in the elderly. II. Preliminary results in the cognitively
deteriorated. J Am Geriatr Soc 34(10):722–727, 1986 3760436
Cohen-Mansfield J: Nonpharmacologic interventions for inappropriate behaviors in dementia: a
review, summary, and critique. Am J Geriatr Psychiatry 9(4):361–381, 2001 11739063
Cohen-Mansfield J, Billig N: Agitated behaviors in the elderly. I. A conceptual review. J Am
Geriatr Soc 34(10):711–721, 1986 3531296
Cohen-Mansfield J, Libin A: Verbal and physical non-aggressive agitated behaviors in elderly
persons with dementia: robustness of syndromes. J Psychiatr Res 39(3): 325–332, 2005
15725431
Cohen-Mansfield J, Marx MS, Rosenthal AS: A description of agitation in a nursing home. J
Gerontol 44(3):M77–M84, 1989 2715584
Cohen-Mansfield J, Lipson S, Werner P, et al: Withdrawal of haloperidol, thioridazine, and
lorazepam in the nursing home: a controlled, double-blind study. Arch Intern Med
159(15):1733–1740, 1999 10448776
Cohen-Mansfield J, Thein K, Marx MS, et al: Efficacy of nonpharmacologic interventions for
agitation in advanced dementia: a randomized, placebo-controlled trial. J Clin Psychiatry
73(9):1255–1261, 2012 23059151
Colenda CC, Streim JE, Greene JA, et al: The impact of OBRA ’87 on psychiatric services in
nursing homes. Joint testimony of the American Psychiatric Association and the American
Association for Geriatric Psychiatry. Am J Geriatr Psychiatry 7(1): 12–17, 1999 9919316
Custer RL, Davis JE, Gee SC: Psychiatric drug usage in VA nursing home care units. Psychiatr
Annals 14(4):285–292, 1984
Datto CJ, Oslin DW, Streim JE, et al: Pharmacologic treatment of depression in nursing home
residents: a mental health services perspective. J Geriatr Psychiatry Neurol 15(3):141–146,
2002 12230084
Decker FH: Nursing Home, 1977–1999: What Has Changed, What Has Not? Hyattsville, MD,
National Center for Health Statistics, 2005
De Deyn PP, Rabheru K, Rasmussen A, et al: A randomized trial of risperidone, placebo, and
haloperidol for behavioral symptoms of dementia. Neurology 53(5):946–955, 1999 10496251
De Deyn PP, Katz IR, Brodaty H, et al: Management of agitation, aggression, and psychosis
associated with dementia: a pooled analysis including three randomized, placebo-controlled
double-blind trials in nursing home residents treated with risperidone. Clin Neurol Neurosurg
107(6): 497–508, 2005 15922506
Dehlin O, Hedenrud B, Jansson P, et al: A double-blind comparison of alaproclate and placebo in
the treatment of patients with senile dementia. Acta Psychiatr Scand 71(2):190–196, 1985
3883697
DeLeo D, Stella AG, Spagnoli A: Prescription of psychotropic drugs in geriatric institutions. Int J
Geriatr Psychiatry 4:11–16, 1989
Deudon A, Maubourguet N, Gervais X, et al: Non-pharmacological management of behavioural
symptoms in nursing homes. Int J Geriatr Psychiatry 24(12):1386–1395, 2009 19370714
Dey AN: Characteristics of elderly nursing home residents: data from the 1995 National Nursing
Home Survey. Adv Data 289(289):1–8, 1997 10182808
Dwyer M, Byrne GJ: Disruptive vocalization and depression in older nursing home residents. Int
Psychogeriatr 12(4):463–471, 2000 11263713
Elon R, Pawlson LG: The impact of OBRA on medical practice within nursing facilities. J Am
Geriatr Soc 40(9):958–963, 1992 1512394
Engle VF, Graney MJ: Stability and improvement of health after nursing home admission. J
Gerontol 48(1):S17–S23, 1993 8418151
Evans LK, Strumpf NE: Tying down the elderly: a review of the literature on physical restraint. J
Am Geriatr Soc 37(1):65–74, 198, 1989 2642501
Evans LK, Strumpf NE, Allen-Taylor SL, et al: A clinical trial to reduce restraints in nursing
homes. J Am Geriatr Soc 45(6):675–681, 1997 9180659
Fenton J, Raskin A, Gruber-Baldini AL, et al: Some predictors of psychiatric consultation in
nursing home residents. Am J Geriatr Psychiatry 12(3):297–304, 2004 15126231
Finkel SI, Lyons JS, Anderson RL: A brief agitation rating scale (BARS) for nursing home
elderly. J Am Geriatr Soc 41(1):50–52, 1993 8418123
Finkel SI, Lyons JS, Anderson RL, et al: A randomized, placebo-controlled trial of thiothixene in
agitated, demented nursing home patients. Int J Geriatr Psychiatry 10:129–136, 1995
Folstein MF, Folstein SE, McHugh PR: “Mini-mental state.” A practical method for grading the
cognitive state of patients for the clinician. J Psychiatr Res 12(3):189–198, 1975 1202204
Fossey J, Ballard C, Juszczak E, et al: Effect of enhanced psychosocial care on antipsychotic use
in nursing home residents with severe dementia: cluster randomised trial. BMJ
332(7544):756–761, 2006 16543297
Fullarton J, Volicer L: Reductions of antipsychotic and hypnotic medications in Namaste Care. J
Am Med Dir Assoc 14(9): 708–709, 2013 23890761
Fullerton CA, McGuire TG, Feng Z, et al: Trends in mental health admissions to nursing homes,
1999–2005. Psychiatr Serv 60(7):965–971, 2009 19564228
Galasko D, Bennett D, Sano M, et al: An inventory to assess activities of daily living for clinical
trials in Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis
Assoc Disord 11 (suppl 2):S33–S39, 1997 9236950
German PS, Shapiro S, Kramer M: Nursing home study of eastern Baltimore epidemiologic
catchment area, in Mental Illness in Nursing Homes: Agenda for Research. Edited by Harper
MS, Lebowitz BD. Rockville, MD, National Institute of Mental Health, 1986, pp 21–40
Gill SS, Bronskill SE, Normand SL, et al: Antipsychotic drug use and mortality in older adults
with dementia. Ann Intern Med 146(11):775–786, 2007 17548409
Goldwasser AN, Auerbach SM, Harkins SW: Cognitive, affective, and behavioral effects of
reminiscence group therapy on demented elderly. Int J Aging Hum Dev 25(3):209–222, 1987
3429043
Gottfries CG, Bråne G, Gullberg B, et al: A new rating scale for dementia syndromes. Arch
Gerontol Geriatr 1(4):311–330, 1982 7186327
Grabowski DC, Aschbrenner KA, Feng Z, et al: Mental illness in nursing homes: variations across
States. Health Aff (Millwood) 28(3):689–700, 2009 19414877
Grabowski DC, Aschbrenner KA, Rome VF, et al: Quality of mental health care for nursing home
residents: a literature review. Med Care Res Rev 67(6):627–656, 2010 20223943
Graessel E, Stemmer R, Eichenseer B, et al: Non-pharmacological, multicomponent group therapy
in patients with degenerative dementia: a 12-month randomized, controlled trial. BMC Med
9:129, 2011 22133165
Grant LA, Kane RA, Stark AJ: Beyond labels: nursing home care for Alzheimer’s disease in and
out of special care units. J Am Geriatr Soc 43(5):569–576, 1995 7730542
Guy W (ed): ECDEU Assessment Manual for Psychopharmacology, Revised (DHEW Publ No
ADM-76–388). Rockville, MD, U.S. Department of Health, Education and Welfare, 1976
Hamilton M: The assessment of anxiety states by rating. Br J Med Psychol 32(1):50–55, 1959
13638508
Hamilton M: A rating scale for depression. J Neurol Neurosurg Psychiatry 23:56–62, 1960
14399272
Hanlon JT, Handler SM, Castle NG: Antidepressant prescribing in U.S. nursing homes between
1996 and 2006 and its relationship to staffing patterns and use of other psychotropic
medications. J Am Med Dir Assoc 11(5):320–324, 2010 20511098
Hansen SS, Patterson MA, Wilson RW: Family involvement on a dementia unit: the Resident
Enrichment and Activity Program. Gerontologist 28(4):508–510, 1988 3224863
Harris Y: Depression as a risk factor for nursing home admission among older individuals. J Am
Med Dir Assoc 8(1):14–20, 2007 17210498
Harris Y, Cooper JK: Depressive symptoms in older people predict nursing home admission. J Am
Geriatr Soc 54(4):593–597, 2006 16686868
Harrison R, Savla N, Kafetz K: Dementia, depression and physical disability in a London
borough: a survey of elderly people in and out of residential care and implications for future
developments. Age Ageing 19(2):97–103, 1990 2140005
Haslam C, Haslam SA, Jetten J, et al: The social treatment: the benefits of group interventions in
residential care settings. Psychol Aging 25(1):157–167, 2010 20230136
Hawes C, Mor V, Phillips CD, et al: The OBRA-87 nursing home regulations and implementation
of the Resident Assessment Instrument: effects on process quality. J Am Geriatr Soc
45(8):977–985, 1997 9256852
Health Care Financing Administration: Medicare and Medicaid: requirements for long-term care
facilities—HCFA. Final rule. Fed Regist 56:48865–48921, 1991
Health Care Financing Administration: Medicare and Medicaid programs; preadmission screening
and annual resident review—HCFA. Final rule with comment period. Fed Regist
57(230):56450–56514, 1992a 10122884
Health Care Financing Administration: Medicare and Medicaid: resident assessment in long-term
care facilities—HCFA. Final rule. Fed Regist 57:61614–61733, 1992b
Health Care Financing Administration: State Operations Manual: Provider Certification
(Transmittal No 250). Washington, DC, Health Care Financing Administration, 1992c
Health Care Financing Administration: Report to Congress: Study of Private Accreditation
(Deeming) of Nursing Homes, Regulatory Incentives and Non-Regulatory Incentives, and
Effectiveness of the Survey and Certification System. Washington, DC, Health Care
Financing Administration, 1998. Available at:
https://archive.org/details/reporttocongress00unit_13. Accessed June 12, 2014.
Heeren O, Borin L, Raskin A, et al: Association of depression with agitation in elderly nursing
home residents. J Geriatr Psychiatry Neurol 16(1):4–7, 2003 12641365
Heston LL, Garrard J, Makris L, et al: Inadequate treatment of depressed nursing home elderly. J
Am Geriatr Soc 40(11): 1117–1122, 1992 1401696
Hoegh M, Ibrahim AK, Chibnall J, et al: Prevalence of Parkinson disease and Parkinson disease
dementia in community nursing homes. Am J Geriatr Psychiatry 21(6):529–535, 2013
23567411
Holmes D, Teresi J, Weiner A, et al: Impacts associated with special care units in long-term care
facilities. Gerontologist 30(2): 178–183, 1990 2189792
Honigfeld G, Gillis RD, Klett CJ: NOSIE-30: a treatment-sensitive ward behavior scale. Psychol
Rep 19(1):180–182, 1966 5942082
Horiguchi J, Inami Y: A survey of the living conditions and psychological states of elderly people
admitted to nursing homes in Japan. Acta Psychiatr Scand 83(5):338–341, 1991 1853725
Hughes CM, Lapane KL, Mor V: Influence of facility characteristics on use of antipsychotic
medications in nursing homes. Med Care 38(12):1164–1173, 2000 11186295
Hyer L, Blazer DG: Depressive symptoms: impact and problems in long term care facilities.
International Journal of Behavioral Gerontology 1:33–44, 1982
Innes EM, Turman WG: Evaluation of patient falls. QRB Qual Rev Bull 9(2):30–35, 1983
6403903
Institute of Medicine Committee on Nursing Home Regulation: Improving the Quality of Care in
Nursing Homes. Washington, DC, National Academy Press, 1986
Jeste DV, Okamoto A, Napolitano J, et al: Low incidence of persistent tardive dyskinesia in
elderly patients with dementia treated with risperidone. Am J Psychiatry 157(7): 1150–1155,
2000 10873925
Jones AL, Dwyer LL, Bercovitz AR, et al: The National Nursing Home Survey: 2004 overview.
Vital Health Stat 13 (167):3–6, 2009
Jongenelis K, Pot AM, Eisses AM, et al: Prevalence and risk indicators of depression in elderly
nursing home patients: the AGED study. J Affect Disord 83(2–3):135–142, 2004 15555706
Kales HC, Gitlin LN, Lyketsos CG; Detroit Expert Panel on the Assessment and Management of
the Neuropsychiatric Symptoms of Dementia: Management of neuropsychiatric symptoms of
dementia in clinic settings: recommendations from a multidisciplinary expert panel. J Am
Geriatr Soc 62(4):762–769, 2014 24635665
Karkare SU, Bhattacharjee S, Kamble P, et al: Prevalence and predictors of antidepressant
prescribing in nursing home residents in the United States. Am J Geriatr Pharmacother
9(2):109–119, 2011 21565710
Karlin BE, Visnic S, Shealy McGee J, et al: Results from the multisite implementation of STAR-
VA: a multicomponent psychosocial intervention for managing challenging dementia-related
behaviors of veterans. Psychol Serv 11(2):200–208, 2014 23937081
Katz IR, Lesher E, Kleban M, et al: Clinical features of depression in the nursing home. Int
Psychogeriatr 1(1):5–15, 1989 2518789
Katz IR, Simpson GM, Curlik SM, et al: Pharmacologic treatment of major depression for elderly
patients in residential care settings. J Clin Psychiatry 51 (suppl):41–47, discussion 48, 1990
2195013
Katz IR, Parmelee P, Brubaker K: Toxic and metabolic encephalopathies in long-term care
patients. Int Psychogeriatr 3(2):337–347, 1991 1811786
Katz IR, Beaston-Wimmer P, Parmelee P, et al: Failure to thrive in the elderly: exploration of the
concept and delineation of psychiatric components. J Geriatr Psychiatry Neurol 6(3):161–169,
1993 8397760
Katz IR, Jeste DV, Mintzer JE, et al; Risperidone Study Group: Comparison of risperidone and
placebo for psychosis and behavioral disturbances associated with dementia: a randomized,
double-blind trial. J Clin Psychiatry 60(2):107–115, 1999 10084637
Kaup BA, Loreck D, Gruber-Baldini AL, et al: Depression and its relationship to function and
medical status, by dementia status, in nursing home admissions. Am J Geriatr Psychiatry
15(5):438–442, 2007 17463194
Kay SR, Fiszbein A, Opler LA: The positive and negative syndrome scale (PANSS) for
schizophrenia. Schizophr Bull 13(2): 261–276, 1987 3616518
Kiely DK, Jones RN, Bergmann MA, et al: Association between delirium resolution and
functional recovery among newly admitted postacute facility patients. J Gerontol A Biol Sci
Med Sci 61(2):204–208, 2006 16510867
Kolanowski AM, Litaker M, Buettner L: Efficacy of theory-based activities for behavioral
symptoms of dementia. Nurs Res 54(4):219–228, 2005 16027564
Kramer M, German PS, Anthony JC, et al: Patterns of mental disorders among the elderly
residents of eastern Baltimore. J Am Geriatr Soc 33(4):236–245, 1985 3989184
Krantz DS, Schulz PR: Personal control and health: some applications to crises of middle and old
age. Advances in Environmental Psychology 2:23–57, 1980
Krauss NA, Altman BM: Characteristics of Nursing Home Residents, 1996. MEPS Research
Findings No 5 (AHCPR Publ No 99-0006). Rockville, MD, Agency for Health Care Policy
and Research, 1998
Kutner N, Mistretta E, Barnhart H, et al: Family members’ perceptions of quality of life change in
dementia SCU residents. J Appl Gerontol 18:423–439, 1999
Lai CK, Yeung JH, Mok V, et al: Special care units for dementia individuals with behavioural
problems. Cochrane Database Syst Rev 2009 Oct 7;(4):CD006470. 19821370
Langer EJ, Rodin J: The effects of choice and enhanced personal responsibility for the aged: a
field experiment in an institutional setting. J Pers Soc Psychol 34(2):191–198, 1976 1011073
Lantz MS, Giambanco V, Buchalter EN: A ten-year review of the effect of OBRA-87 on
psychotropic prescribing practices in an academic nursing home. Psychiatr Serv 47(9):951–
955, 1996 8875659
Lawrence V, Fossey J, Ballard C, et al: Improving quality of life for people with dementia in care
homes: making psychosocial interventions work. Br J Psychiatry 201(5): 344–351, 2012
23118034
Lawton MP, Brody EM: Assessment of older people: self-maintaining and instrumental activities
of daily living. Gerontologist 9(3):179–186, 1969 5349366
Lawton MP, Van Haitsma K, Klapper J, et al: A stimulation-retreat special care unit for elders with
dementing illness. Int Psychogeriatr 10(4):379–395, 1998 9924833
Lenze EJ, Host HH, Hildebrand MW, et al: Enhanced medical rehabilitation increases therapy
intensity and engagement and improves functional outcomes in postacute rehabilitation of
older adults: a randomized-controlled trial. J Am Med Dir Assoc 13(8):708–712, 2012
22863663
Lesher E: Validation of the Geriatric Depression Scale among nursing home residents. Clin
Gerontol 4:21–28, 1986
Leszcz M, Feigenbaum E, Sadavoy J, et al: A men’s group: psychotherapy of elderly men. Int J
Group Psychother 35(2):177–196, 1985 4008130
Levin CA, Wei W, Akincigil A, et al: Prevalence and treatment of diagnosed depression among
elderly nursing home residents in Ohio. J Am Med Dir Assoc 8(9):585–594, 2007 17998115
Lin MK, Kramer AM: The Quality Indicator Survey: background, implementation, and
widespread change. J Aging Soc Policy 25(1):10–29, 2013 23256556
Linkins KW, Lucca AM, Housman M, et al: Use of PASRR programs to assess serious mental
illness and service access in nursing homes. Psychiatr Serv 57(3):325–332, 2006 16524989
Livingston G, Johnston K, Katona C, et al; Old Age Task Force of the World Federation of
Biological Psychiatry: Systematic review of psychological approaches to the management of
neuropsychiatric symptoms of dementia. Am J Psychiatry 162(11): 1996–2021, 2005
16263837
Llorente MD, Olsen EJ, Leyva O, et al: Use of antipsychotic drugs in nursing homes: current
compliance with OBRA regulations. J Am Geriatr Soc 46(2):198–201, 1998 9475449
Loebel JP, Borson S, Hyde T, et al: Relationships between requests for psychiatric consultations
and psychiatric diagnoses in long-term-care facilities. Am J Psychiatry 148(7):898–903, 1991
2053630
Lopez OL, Becker JT, Wahed AS, et al: Long-term effects of the concomitant use of memantine
with cholinesterase inhibition in Alzheimer disease. J Neurol Neurosurg Psychiatry
80(6):600–607, 2009 19204022
Low LF, Brodaty H, Goodenough B, et al: The Sydney Multisite Intervention of LaughterBosses
and ElderClowns (SMILE) study: cluster randomized trial of humour therapy in nursing
homes. BMJ Open 3(1), 2013 23315520
Lyketsos CG, Lindell Veiel L, Baker A, et al: A randomized, controlled trial of bright light therapy
for agitated behaviors in dementia patients residing in long-term care. Int J Geriatr Psychiatry
14(7):520–525, 1999 10440971
Magai C, Kennedy G, Cohen CI, et al: A controlled clinical trial of sertraline in the treatment of
depression in nursing home patients with late-stage Alzheimer’s disease. Am J Geriatr
Psychiatry 8(1):66–74, 2000 10648297
Mack JL, Patterson MB, Tariot PN: Behavior rating scale for dementia: development of test scales
and presentation of data for 555 individuals with Alzheimer’s disease. J Geriatr Psychiatry
Neurol 12(4):211–223, 1999 10616870
Malmstrom TK, Miller DK, Morley JE: A comparison of four frailty models. J Am Geriatr Soc
62(4):721–726, 2014 24635726
Mann AH, Graham N, Ashby D: Psychiatric illness in residential homes for the elderly: a survey
in one London borough. Age Ageing 13(5):257–265, 1984 6496236
McCarthy JF, Blow FC, Kales HC: Disruptive behaviors in Veterans Affairs nursing home
residents: how different are residents with serious mental illness? J Am Geriatr Soc
52(12):2031–2038, 2004 15571538
McCurren C: Assessment for depression among nursing home elders: evaluation of the MDS
mood assessment. Geriatr Nurs 23(2):103–108, 2002 11956523
McCusker J, Cole MG, Voyer P, et al: Observer-rated depression in long-term care: frequency and
risk factors. Arch Gerontol Geriatr 58(3):332–338, 2014 24345307
McMurdo MET, Rennie L: A controlled trial of exercise by residents of old people’s homes. Age
Ageing 22(1):11–15, 1993 8438659
Meador KG, Taylor JA, Thapa PB, et al: Predictors of antipsychotic withdrawal or dose reduction
in a randomized controlled trial of provider education. J Am Geriatr Soc 45(2):207–210, 1997
9033521
Medicare.gov: The Official U.S. Government Site for Medicare, Nursing Home Compare. 2014a.
Available at: http://www.medicare.gov/nursinghomecompare/search.html?
AspxAutoDetectCookie-Support=1. Accessed January 5, 2015.
Medicare.gov: Quality Improvement Organizations. 2014b. Available at:
http://www.cms.gov/Medicare/Quality-Initiatives-Patient-Assessment-
Instruments/QualityImprovementOrgs/index.html?redirect=/qualityimprovementorgs/.
Accessed June 11, 2014.
Meehan KM, Wang H, David SR, et al: Comparison of rapidly acting intramuscular olanzapine,
lorazepam, and placebo: a double-blind, randomized study in acutely agitated patients with
dementia. Neuropsychopharmacology 26(4):494–504, 2002 11927174
Mintzer J, Greenspan A, Caers I, et al: Risperidone in the treatment of psychosis of Alzheimer
disease: results from a prospective clinical trial. Am J Geriatr Psychiatry 14(3):280–291, 2006
16505133
Mintzer JE, Tune LE, Breder CD, et al: Aripiprazole for the treatment of psychoses in
institutionalized patients with Alzheimer dementia: a multicenter, randomized, double-blind,
placebo-controlled assessment of three fixed doses. Am J Geriatr Psychiatry 15(11):918–931,
2007 17974864
Montgomery SA, Åsberg M: A new depression scale designed to be sensitive to change. Br J
Psychiatry 134:382–389, 1979 444788
Moran JA, Gatz M: Group therapies for nursing home adults: an evaluation of two treatment
approaches. Gerontologist 27(5):588–591, 1987 3678897
Morris JN, Hawes C, Fries BE, et al: Designing the national resident assessment instrument for
nursing homes. Gerontologist 30(3):293–307, 1990 2354790
Murphy E: The use of psychotropic drugs in long-term care (editorial). Int J Geriatr Psychiatry
4:1–2, 1989
National Center for Health Statistics: The National Nursing Home Survey (DHEW Publ No PHS-
79-1794). Hyattsville, MD, National Center for Health Statistics, 1979
National Center for Health Statistics: The National Nursing Home Survey. Hyattsville, MD,
National Center for Health Statistics. 2004. Available at:
http://www.cdc.gov/nchs/nnhs/about_nnhs.htm. Accessed June 11, 2014.
Nobili A, Piana I, Balossi L, et al: Alzheimer special care units compared with traditional nursing
home for dementia care: are there differences at admission and in clinical outcomes?
Alzheimer Dis Assoc Disord 22(4):352–361, 2008 18978601
Nursing Home Quality Indicators Development Group: Facility Guide for the Nursing Home
Quality Indicators. National Data System. September 28, 1999. Available at:
http://www.qtso.com/downloads/mds/facman.pdf. Accessed June 11, 2014.
Nyth AL, Gottfries CG: The clinical efficacy of citalopram in treatment of emotional disturbances
in dementia disorders: a Nordic multicentre study. Br J Psychiatry 157: 894–901, 1990
1705151
Office of Inspector General: Medicare Payments for Psychiatric Services in Nursing Homes: A
Follow-Up (Publ No OEI-02-99-00140). Washington, DC, U.S. Department of Health and
Human Services. 2001a. Available at: http://oig.hhs.gov/oei/reports/oei-02-99-00140.pdf.
Accessed June 11, 2014.
Office of Inspector General: Nursing Home Resident Assessment, Quality of Care (Publ No OEI-
02-99-00040). Washington, DC, U.S. Department of Health and Human Services. 2001b.
Available at: http://oig.hhs.gov/oei/reports/oei-02-99-00040.pdf. Accessed June 11, 2014.
Office of Inspector General: Psychotropic Drug Use in Nursing Homes (Publ No OEI-02-00-
00490). Washington, DC, U.S. Department of Health and Human Services. 2001c. Available
at: http://oig.hhs.gov/oei/reports/oei-02-00-00490.pdf. Accessed June 11, 2014.
Office of Technology Assessment: Special Care Units for People With Alzheimer’s and Other
Dementias: Consumer Education, Research, Regulatory, and Reimbursement Issues (OTA-H-
543). Washington, DC, U.S. Government Printing Office, August 1992
Ohta RJ, Ohta BM: Special units for Alzheimer’s disease patients: a critical look. Gerontologist
28(6):803–808, 1988 3150963
Olazarán J, Reisberg B, Clare L, et al: Nonpharmacological therapies in Alzheimer’s disease: a
systematic review of efficacy. Dement Geriatr Cogn Disord 30(2):161–178, 2010 20838046
Olin JT, Fox LS, Pawluczyk S, et al: A pilot randomized trial of carbamazepine for behavioral
symptoms in treatment-resistant outpatients with Alzheimer disease. Am J Geriatr Psychiatry
9(4):400–405, 2001 11739066
Ollech D: An analyst’s experience working in a skilled nursing facility: a case study. Am J
Psychoanal 66(4):381–390, 2006 17115282
Omnibus Budget Reconciliation Act of 1987, Pub L No 100-203. Subtitle C: Nursing home
reform.
Onder G, Liperoti R, Soldato M, et al: Depression and risk of nursing home admission among
older adults in home care in Europe: results from the Aged in Home Care (AdHOC) study. J
Clin Psychiatry 68(9): 1392–1398, 2007 17915978
Orten JD, Allen M, Cook J: Reminiscence groups with confused nursing center residents: an
experimental study. Soc Work Health Care 14(1):73–86, 1989 2781444
Oslin DW, Streim JE, Katz IR, et al: Heuristic comparison of sertraline with nortriptyline for the
treatment of depression in frail elderly patients. Am J Geriatr Psychiatry 8(2):141–149, 2000
10804075
Oslin DW, Ten Have TR, Streim JE, et al: Probing the safety of medications in the frail elderly:
evidence from a randomized clinical trial of sertraline and venlafaxine in depressed nursing
home residents. J Clin Psychiatry 64(8):875–882, 2003 12927001
Overall JE, Gorham DR: The Brief Psychiatric Rating Scale. Psychol Rep 10:799–812, 1962
Parmelee PA, Katz IR, Lawton MP: Depression among institutionalized aged: assessment and
prevalence estimation. J Gerontol 44(1):M22–M29, 1989 2783434
Parmelee PA, Katz IR, Lawton MP: The relation of pain to depression among institutionalized
aged. J Gerontol 46(1):15–21, 1991 1986040
Parmelee PA, Katz IR, Lawton MP: Incidence of depression in long-term care settings. J Gerontol
47(6):M189–M196, 1992 1430853
Phillips CD, Sloane PD, Hawes C, et al: Effects of residence in Alzheimer disease special care
units on functional outcomes. JAMA 278(16):1340–1344, 1997 9343465
Phillips CD, Spry KM, Sloane PD, Hawes C: Use of physical restraints and psychotropic
medications in Alzheimer special care units in nursing homes. Am J Public Health 90(1):92–
96, 2000 10630143
Politis AM, Vozzella S, Mayer LS, et al: A randomized, controlled, clinical trial of activity therapy
for apathy in patients with dementia residing in long-term care. Int J Geriatr Psychiatry
19(11):1087–1094, 2004 15481065
Porsteinsson AP, Tariot PN, Erb R, et al: Placebo-controlled study of divalproex sodium for
agitation in dementia. Am J Geriatr Psychiatry 9(1):58–66, 2001 11156753
Rappaport SA, Marcus RN, Manos G, et al: A randomized, double-blind, placebo-controlled
tolerability study of intramuscular aripiprazole in acutely agitated patients with Alzheimer’s,
vascular, or mixed dementia. J Am Med Dir Assoc 10(1):21–27, 2009 19111849
Rattenbury C, Stones MJ: A controlled evaluation of reminiscence and current topics discussion
groups in a nursing home context. Gerontologist 29(6):768–771, 1989 2620839
Ray WA, Federspiel CF, Schaffner W: A study of antipsychotic drug use in nursing homes:
epidemiologic evidence suggesting misuse. Am J Public Health 70(5):485–491, 1980
6103676
Ray WA, Taylor JA, Meador KG, et al: Reducing antipsychotic drug use in nursing homes. A
controlled trial of provider education. Arch Intern Med 153(6):713–721, 1993 8447709
Reichman WE, Conn DK: Nursing home psychiatry: is it time for a reappraisal? Am J Geriatr
Psychiatry 18(12):1049–1053, 2010
Reichman WE, Coyne AC, Borson S, et al: Psychiatric consultation in the nursing home: a survey
of six states. Am J Geriatr Psychiatry 6(4):320–327, 1998 9793580
Reisberg B: Functional assessment staging (FAST). Psychopharmacol Bull 24(4):653–659, 1988
3249767
Reisberg B, Ferris SH, de Leon MJ, Crook T: Global Deterioration Scale (GDS).
Psychopharmacol Bull 24(4):661–663, 1988 3249768
Reisberg B, Auer SR, Monteiro IM: Behavioral pathology in Alzheimer’s disease (BEHAVE-AD)
rating scale. Int Psychogeriatr 8 (suppl 3):301–308, discussion 351–354, 1996 9154579
Rhoades J, Krauss N: Nursing Home Trends, 1987 and 1996. MEPS Chartbook No 3 (AHCPR
Publ No 99-0032). Rockville, MD, Agency for Health Care Policy and Research, 1999
Risse SC, Cubberley L, Lampe TH, et al: Acute effects of neuroleptic withdrawal in elderly
dementia patients. J Geriatr Drug Ther 2:65–77, 1987
Rochon PA, Normand SL, Gomes T, et al: Antipsychotic therapy and short-term serious events in
older adults with dementia. Arch Intern Med 168(10):1090–1096, 2008 18504337
Rollow W, Lied TR, McGann P, et al: Assessment of the Medicare quality improvement
organization program. Ann Intern Med 145(5):342–353, 2006 16908911
Rosen J, Mulsant BH, Pollock BG: Sertraline in the treatment of minor depression in nursing
home residents: a pilot study. Int J Geriatr Psychiatry 15(2):177–180, 2000 10679849
Rovner BW, Kafonek S, Filipp L, et al: Prevalence of mental illness in a community nursing
home. Am J Psychiatry 143(11): 1446–1449, 1986 3777239
Rovner BW, German PS, Broadhead J, et al: The prevalence and management of dementia and
other psychiatric disorders in nursing homes. Int Psychogeriatr 2(1):13–24, 1990a 2101294
Rovner BW, Lucas-Blaustein J, Folstein MF, et al: Stability over one year in patients admitted to a
nursing home dementia unit. Int J Geriatr Psychiatry 5:77–82, 1990b
Rovner BW, German PS, Brant LJ, et al: Depression and mortality in nursing homes. JAMA
265(8):993–996, 1991 1992213
Rovner BW, Edelman BA, Cox MP, et al: The impact of antipsychotic drug regulations on
psychotropic prescribing practices in nursing homes. Am J Psychiatry 149(10): 1390–1392,
1992 1530076
Rovner BW, Steele CD, Shmuely Y, et al: A randomized trial of dementia care in nursing homes. J
Am Geriatr Soc 44(1):7–13, 1996 8537594
Rozzini R, Boffelli S, Franzoni S: Prevalence and predictors of depressive symptoms in a nursing
home. J Geriatr Psychiatry 11:629–634, 1996
Ruckdeschel K, Thompson R, Datto CJ, et al: Using the Minimum Data Set 2.0 mood disturbance
items as a self-report screening instrument for depression in nursing home residents. Am J
Geriatr Psychiatry 12(1):43–49, 2004 14729558
Ruths S, Straand J, Nygaard HA, et al: Effect of antipsychotic withdrawal on behavior and
sleep/wake activity in nursing home residents with dementia: a randomized, placebo-
controlled, double-blinded study. The Bergen District Nursing Home Study. J Am Geriatr Soc
52(10):1737–1743, 2004 15450054
Sabin TD, Vitug AJ, Mark VH: Are nursing home diagnosis and treatment inadequate? JAMA
248(3):321–322, 1982 7087125
Sadavoy J: Psychotherapy for the institutionalized elderly, in Practical Psychiatry in the Nursing
Home: A Handbook for Staff. Edited by Conn DK, Herrman N, Kaye A, et al. Toronto, ON,
Canada, Hogrefe and Huber, 1991, pp 217–236
Saliba D, Jones M, Streim J, et al: Overview of significant changes in the Minimum Data Set for
nursing homes version 3.0. J Am Med Dir Assoc 13(7):595–601, 2012 22784698
Saxton J, Silverman M, Ricci E, et al: Maintenance of mobility in residents of an Alzheimer
special care facility. Int Psychogeriatr 10(2):213–224, 1998 9677508
Schneider LS, Pollock VE, Lyness SA: A meta-analysis of controlled trials of neuroleptic
treatment in dementia. J Am Geriatr Soc 38(5):553–563, 1990 1970586
Schneider LS, Olin JT, Doody RS, et al: Validity and reliability of the Alzheimer’s disease
Cooperative Study–Clinical Global Impression of Change. The Alzheimer’s Disease
Cooperative Study. Alzheimer Dis and Assoc Disord 11 (suppl 2):S22–S32, 1997 9236949
Schneider LS, Dagerman KS, Insel P: Risk of death with atypical antipsychotic drug treatment for
dementia: meta-analysis of randomized placebo-controlled trials. JAMA 294(15):1934–1943,
2005 16234500
Schnelle JF, Newman DR, White M, et al: Reducing and managing restraints in long-term-care
facilities. J Am Geriatr Soc 40(4):381–385, 1992 1556366
Schnelle JF, Wood S, Schnelle ER, et al: Measurement sensitivity and the Minimum Data Set
depression quality indicator. Gerontologist 41(3):401–405, 2001 11405438
Schulz R: Effects of control and predictability on the physical and psychological well-being of the
institutionalized aged. J Pers Soc Psychol 33(5):563–573, 1976 1271225
Seitz DP, Gruneir A, Conn DK, et al: Cholinesterase inhibitor use in U.S. nursing homes: results
from the national nursing home survey. J Am Geriatr Soc 57(12):2269–2274, 2009 19874411
Seitz D, Purandare N, Conn D: Prevalence of psychiatric disorders among older adults in long-
term care homes: a systematic review. Int Psychogeriatr 22(7):1025–1039, 2010 20522279
Seitz DP, Gill SS, Herrmann N, et al: Pharmacological treatments for neuropsychiatric symptoms
of dementia in long-term care: a systematic review. Int Psychogeriatr 25(2):185–203, 2013
23083438
Semla TP, Palla K, Poddig B, et al: Effect of the Omnibus Reconciliation Act 1987 on
antipsychotic prescribing in nursing home residents. J Am Geriatr Soc 42(6):648–652, 1994
7911134
Shader RI, Harmatz JS, Salzman C: A new scale for clinical assessment in geriatric populations:
Sandoz Clinical Assessment—Geriatric (SCAG). J Am Geriatr Soc 22(3):107–113, 1974
4464879
Shea DG, Russo PA, Smyer MA: Use of mental health services by persons with a mental illness in
nursing facilities: initial impacts of OBRA87. J Aging Health 12(4):560–578, 2000 11503732
Shorr RI, Fought RL, Ray WA: Changes in antipsychotic drug use in nursing homes during
implementation of the OBRA-87 regulations. JAMA 271(5):358–362, 1994 8283585
Siegler EL, Capezuti E, Maislin G, et al: Effects of a restraint reduction intervention and OBRA
’87 regulations on psychoactive drug use in nursing homes. J Am Geriatr Soc 45(7):791–796,
1997 9215327
Simard J, Volicer L: Effects of Namaste Care on residents who do not benefit from usual
activities. Am J Alzheimers Dis Other Demen 25(1):46–50, 2010 19332652
Sloane PD, Mathew LJ, Scarborough M, et al: Physical and pharmacologic restraint of nursing
home patients with dementia. Impact of specialized units. JAMA 265(10): 1278–1282, 1991
1995975
Sloane PD, Mitchell CM, Preisser JS, et al: Environmental correlates of resident agitation in
Alzheimer’s disease special care units. J Am Geriatr Soc 46(7):862–869, 1998 9670873
Smalbrugge M, Jongenelis L, Pot AM, et al: Incidence and outcome of depressive symptoms in
nursing home patients in the Netherlands. Am J Geriatr Psychiatry 14(12):1069–1076, 2006
17138812
Smith M, Buckwalter KC, Albanese M: Geropsychiatric education programs. Providing skills and
understanding. J Psychosoc Nurs Ment Health Serv 28(12):8–12, 1990 2126559
Smyer M, Brannon D, Cohn M: Improving nursing home care through training and job redesign.
Gerontologist 32(3):327–333, 1992 1499997
Smyer MA, Shea DG, Streit A: The provision and use of mental health services in nursing homes:
results from the National Medical Expenditure Survey. Am J Public Health 84(2):284–287,
1994 8296955
Sneed JR, Roose SP, Keilp JG, et al: Response inhibition predicts poor antidepressant treatment
response in very old depressed patients. Am J Geriatr Psychiatry 15(7): 553–563, 2007
17586780
Sneed JR, Keilp JG, Brickman AM, et al: The specificity of neuropsychological impairment in
predicting antidepressant non-response in the very old depressed. Int J Geriatr Psychiatry
23(3):319–323, 2008 17726720
Snow LA, Hovanec L, Brandt J: A controlled trial of aromatherapy for agitation in nursing home
patients with dementia. J Altern Complement Med 10(3):431–437, 2004 15253846
Snowden MB: The Minimum Data Set Depression Rating Scale (MDSDRS) lacks reliability for
identifying depression among older adults living in nursing homes. Evid Based Ment Health
7(1):7, 2004 14769651
Snowden M, Roy-Byrne P: Mental illness and nursing home reform: OBRA-87 ten years later.
Omnibus Budget Reconciliation Act. Psychiatr Serv 49(2):229–233, 1998 9575011
Snowden M, Sato K, Roy-Byrne P: Assessment and treatment of nursing home residents with
depression or behavioral symptoms associated with dementia: a review of the literature. J Am
Geriatr Soc 51(9):1305–1317, 2003 12919245
Snowdon J: Dementia, depression, and life satisfaction in nursing homes. Int J Geriatr Psychiatry
1:85–91, 1986
Snowdon J, Donnelly N: A study of depression in nursing homes. J Psychiatr Res 20(4):327–333,
1986 3806427
Sommer OH, Aga O, Cvancarova M, et al: Effect of oxcarbazepine in the treatment of agitation
and aggression in severe dementia. Dement Geriatr Cogn Disord 27(2): 155–163, 2009
19182483
Spagnoli A, Foresti G, Macdonald A, et al: Dementia and depression in Italian geriatric
institutions. Int J Geriatr Psychiatry 1:15–23, 1986
Steele C, Rovner B, Chase GA, et al: Psychiatric symptoms and nursing home placement of
patients with Alzheimer’s disease. Am J Psychiatry 147(8):1049–1051, 1990 2375439
Steiner A, Neu CR: Monitoring the Changes in Use of Medicare Posthospital Services.
Washington, DC, RAND/UCLA/Harvard Center for Health Care Financing Policy Research,
1993
Stevenson DG, Decker SL, Dwyer LL, et al: Antipsychotic and benzodiazepine use among
nursing home residents: findings from the 2004 National Nursing Home Survey. Am J Geriatr
Psychiatry 18(12): 1078–1092, 2010 20808119
Stotsky B: Multicenter study comparing thioridazine with diazepam and placebo in elderly,
nonpsychotic patients with emotional and behavioral disorders. Clin Ther 6(4):546–559, 1984
6380725
Street JS, Clark WS, Gannon KS, et al; The HGEU Study Group: Olanzapine treatment of
psychotic and behavioral symptoms in patients with Alzheimer disease in nursing care
facilities: a double-blind, randomized, placebo-controlled trial. Arch Gen Psychiatry
57(10):968–976, 2000 11015815
Streim JE, Katz IR: Federal regulations and the care of patients with dementia in the nursing
home. Med Clin North Am 78(4):895–909, 1994 8022236
Streim JE, Katz IR: Psychiatric aspects of long-term care, in Comprehensive Textbook of
Psychiatry, 9th Edition, Vol II. Edited by Sadock BJ, Sadock VA, Ruiz P. Philadelphia, PA,
Lippincott Williams & Wilkins, 2009, pp 4195–4200
Streim JE, Oslin DW, Katz IR, et al: Drug treatment of depression in frail elderly nursing home
residents. Am J Geriatr Psychiatry 8(2):150–159, 2000 10804076
Streim JE, Porsteinsson AP, Breder CD, et al: A randomized, double-blind, placebo-controlled
study of aripiprazole for the treatment of psychosis in nursing home patients with Alzheimer
disease. Am J Geriatr Psychiatry 16(7):537–550, 2008 18591574
Sunderland T, Silver MA: Neuroleptics in the treatment of dementia. Int J Geriatr Psychiatry
3:79–88, 1988
Sutcliffe C, Burns A, Challis D, et al: Depressed mood, cognitive impairment, and survival in
older people admitted to care homes in England. Am J Geriatr Psychiatry 15(8):708–715,
2007 17504909
Svarstad BL, Mount JK, Bigelow W: Variations in the treatment culture of nursing homes and
responses to regulations to reduce drug use. Psychiatr Serv 52(5):666–672, 2001 11331803
Swanson EA, Maas ML, Buckwalter KC: Catastrophic reactions and other behaviors of
Alzheimer’s residents: special unit compared with traditional units. Arch Psychiatr Nurs
7(5):292–299, 1993 8257198
Szczepanska-Gieracha J, Kowalska J, Pawik M, Rymaszewska J: Evaluation of a short-term group
psychotherapy used as part of the rehabilitation process in nursing home patients. Disabil
Rehabil 36(12): 1027–1032, 2014 23962232
Tariot PN, Podgorski CA, Blazina L, et al: Mental disorders in the nursing home: another
perspective. Am J Psychiatry 150(7):1063–1069, 1993
Tariot PN, Erb R, Podgorski CA, et al: Efficacy and tolerability of carbamazepine for agitation
and aggression in dementia. Am J Psychiatry 155(1):54–61, 1998 9433339
Tariot PN, Cummings JL, Katz IR, et al: A randomized, double-blind, placebo-controlled study of
the efficacy and safety of donepezil in patients with Alzheimer’s disease in the nursing home
setting. J Am Geriatr Soc 49(12):1590–1599, 2001a 11843990
Tariot PN, Schneider LS, Mintzer J, et al: Safety and tolerability of divalproex sodium in the
treatment of signs and symptoms of mania in elderly patients with dementia: results of a
double-blind, placebo-controlled trial. Curr Ther Res Clin Exp 62: 51–67, 2001b
Tariot PN, Farlow MR, Grossberg GT, et al; Memantine Study Group: Memantine treatment in
patients with moderate to severe Alzheimer disease already receiving donepezil: a
randomized controlled trial. JAMA 291(3):317–324, 2004 14734594
Tariot PN, Raman R, Jakimovich L, et al; Alzheimer’s Disease Cooperative Study; Valproate
Nursing Home Study Group: Divalproex sodium in nursing home residents with possible or
probable Alzheimer disease complicated by agitation: a randomized, controlled trial. Am J
Geriatr Psychiatry 13(11):942–949, 2005 16286437
Teeter RB, Garetz FK, Miller WR, Heiland WF: Psychiatric disturbances of aged patients in
skilled nursing homes. Am J Psychiatry 133(12):1430–1434, 1976 985657
Teri L, Logsdon RG, McCurry SM: Nonpharmacological treatment of behavioral disturbances in
dementia. Med Clin North Am (86):641–656, 2002
Teri L, Huda P, Gibbons L, et al: STAR: a dementia-specific training program for staff in assisted
living residences. Gerontologist 45(5):686–693, 2005 16199404
Thapa PB, Gideon P, Cost TW, et al: Antidepressants and the risk of falls among nursing home
residents. N Engl J Med 339(13): 875–882, 1998 9744971
Thomasma M, Yeaworth RC, McCabe BW: Moving day: relocation and anxiety in
institutionalized elderly. J Gerontol Nurs 16(7):18–25, 1990 2370428
Tondi L, Ribani L, Bottazzi M, et al: Validation therapy (VT) in nursing home: a case-control
study. Arch Gerontol Geriatr 44 (suppl 1):407–411, 2007 17317483
Toseland RW, Diehl M, Freeman K, et al: The impact of validation group therapy on nursing home
residents with dementia. J Appl Gerontol 61:31–50, 1997
Trappler B, Cohen CI: Using fluoxetine in “very old” depressed nursing home residents. Am J
Geriatr Psychiatry 4:258–262, 1996
Trappler B, Cohen CI: Use of SSRIs in “very old” depressed nursing home residents. Am J Geriatr
Psychiatry 6(1):83–89, 1998 9469218
Trichard L, Zabow A, Gillis LS: Elderly persons in old-age homes: a medical, psychiatric and
social investigation. S Afr Med J 61(17):624–627, 1982 7079853
U.S. Food and Drug Administration: Risperdal (Risperidone) Dear Healthcare Professional Letter,
April 2003. Available at: http://www.fda.gov/Safety/MedWatch/SafetyInformation/Safety-
AlertsforHumanMedicalProducts/ucm168933.htm. Accessed January 5, 2015.
U.S. Food and Drug Administration: Public Health Advisory: Deaths with Antipsychotics in
Elderly Patients with Behavioral Disturbances, April 11, 2005. Available at:
http://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandprovider
s/ucm053171. Accessed January 5, 2015.
van Weert JC, van Dulmen AM, Spreeuwenberg PM, et al: Behavioral and mood effects of
snoezelen integrated into 24-hour dementia care. J Am Geriatr Soc 53(1):24–33, 2005
15667372
Vink AC, Zuidersma M, Boersma F, et al: The effect of music therapy compared with general
recreational activities in reducing agitation in people with dementia: a randomized controlled
trial. J Geriatr Psychiatry 10:1031–1038, 2013
Volicer L, Frijters DH, Van der Steen JT: Relationship between symptoms of depression and
agitation in nursing home residents with dementia. Int J Geriatr Psychiatry 27(7):749–754,
2012 21956820
von Gunten A, Mosimann UP, Antonietti JP: A longitudinal study on delirium in nursing homes.
Am J Geriatr Psychiatry 21(10): 963–972, 2013 23567403
Wagner AW, Teri L, Orr-Rainey N: Behavior problems of residents with dementia in special care
units. Alzheimer Dis Assoc Disord 9(3):121–127, 1995 8534409
Wang PS, Schneeweiss S, Avorn J, et al: Risk of death in elderly users of conventional vs. atypical
antipsychotic medications. N Engl J Med 353(22):2335–2341, 2005 16319382
Webber AP, Martin JL, Harker JO, et al: Depression in older patients admitted for postacute
nursing home rehabilitation. J Am Geriatr Soc 53(6):1017–1022, 2005 15935027
Wells Y, Jorm AF: Evaluation of a special nursing home unit for dementia sufferers: a randomised
controlled comparison with community care. Aust N Z J Psychiatry 21(4):524–531, 1987
3449047
Werner P, Cohen-Mansfield J, Braun J, Marx MS: Physical restraints and agitation in nursing
home residents. J Am Geriatr Soc 37(12):1122–1126, 1989 2592719
Wetzels RB, Zuidema SU, de Jonghe JFM, et al: Course of neuropsychiatric symptoms in
residents with dementia in nursing homes over 2-year period. Am J Geriatr Psychiatry
18(12):1054–1065, 2010 21155143
Williams-Barnard CL, Lindell AR: Therapeutic use of “prizing” and its effect on self-concept of
elderly clients in nursing homes and group homes. Issues Ment Health Nurs 13(1):1–17, 1992
1737699
Winblad B, Kilander L, Eriksson S, et al; Severe Alzheimer’s Disease Study Group: Donepezil in
patients with severe Alzheimer’s disease: double-blind, parallel-group, placebo-controlled
study. Lancet 367(9516):1057–1065, 2006 16581404
Won A, Lapane KL, Vallow S, et al: Long-term effects of analgesics in a population of elderly
nursing home residents with persistent nonmalignant pain. J Gerontol A Biol Sci Med Sci
61(2):165–169, 2006 16510860
Wood S, Cummings JL, Hsu MA, et al: The use of the neuropsychiatric inventory in nursing home
residents. Characterization and measurement. Am J Geriatr Psychiatry 8(1):75–83, 2000
10648298
Yesavage JA, Brink TL, Rose TL, et al: Development and validation of a geriatric depression
screening scale: a preliminary report. J Psychiatr Res 17(1):37–49, 1982 7183759
Youssef FA: The impact of group reminiscence counseling on a depressed elderly population.
Nurse Pract 15(4):32–38, 35–38, 1990 2325924
Yudofsky SC, Silver JM, Jackson W, et al: The Overt Aggression Scale for the objective rating of
verbal and physical aggression. Am J Psychiatry 143(1):35–39, 1986 3942284
Zerhusen JD, Boyle K, Wilson W: Out of the darkness: group cognitive therapy for depressed
elderly. J Psychosoc Nurs Ment Health Serv 29(9):16–21, 1991 1941727
Zhong KX, Tariot PN, Mintzer J, et al: Quetiapine to treat agitation in dementia: a randomized,
double-blind, placebo-controlled study. Curr Alzheimer Res 4(1): 81–93, 2007 17316169
Zimmer JG, Watson N, Treat A: Behavioral problems among patients in skilled nursing facilities.
Am J Public Health 74(10): 1118–1121, 1984 6476166
Suggested Readings
American Geriatrics Society; American Association for Geriatric Psychiatry: The American
Geriatrics Society and American Association for Geriatric Psychiatry recommendations for
policies in support of quality mental health care in U.S. nursing homes. J Am Geriatr Soc
51(9):1299–1304, 2003
Bharucha AJ, Dew MA, Miller MD, et al: Psychotherapy in long-term care: a review. J Am Med
Dir Assoc 7(9):568–580, 2006
Boyce RD, Hanlon JT, Karp JF, et al: A review of the effectiveness of antidepressant medications
for depressed nursing home residents. J Am Med Dir Assoc 13(4):326–331, 2012
Centers for Medicare and Medicaid: State Operations Manual, Appendix PP: Guidance to
Surveyors for Long Term Care Facilities. Baltimore, MD, Centers for Medicare and
Medicaid, Rev 107, 04-04-14. Baltimore, MD, Centers for Medicare and Medicaid. 2014.
Available at: https://www.cms.gov/Regulations-and-
Guidance/Guidance/Manuals/downloads/som107ap_pp_guidelines_ltcf.pdf. Accessed June
12, 2014.
Karlin BE, Visnic S, Shealy McGee J, et al: Results from the multisite implementation of STAR-
VA: a multicomponent psychosocial intervention for managing challenging dementia-related
behaviors of veterans. Psychol Serv 11(2):200–208, 2014
Saliba D, Jones M, Streim J, et al: Overview of significant changes in the Minimum Data Set for
nursing homes version 3.0. J Am Med Dir Assoc 13(7):595–601, 2012
Index
Page numbers printed in boldface type refer to tables or figures.
B lymphocytes, 43
Baby boomers, 3, 33, 415, 459
substance-related disorders among, 459, 460, 468, 655
suicide among, 20
Balance problems, 51
assessment of, 52
falls due to, 47, 48, 637
in Lewy body neurocognitive disorder, 202
medication-related, 47
nonpharmacological interventions for, 224
exercise, 637
Balint’s syndrome, 138
Baltimore Longitudinal Study of Aging, 34
Bapineuzumab, 213
Barbiturates, 112, 267, 470, 594
before electroconvulsive therapy, 268–269
Baroreceptor reflex, 34
BDI (Beck Depression Inventory), 131, 698–700
Beck Anxiety Inventory, 131
Beck Depression Inventory (BDI), 131, 698–700
Beers Criteria for Potentially Inappropriate Medication Use in Older
Adults, 50, 165, 168
Behavior therapy. See also Cognitive-behavioral therapy
brief interventions for substance-related disorders, 476
for depression, 264
dialectical behavior therapy, for personality disorders, 500–501,
650
for inappropriate sexual behaviors, 407
for insomnia, 446
in psychosis of Alzheimer’s disease, 316
Behavioral disturbances. See also Aggression; Agitation; Paranoia;
Psychosis/psychotic symptoms
agitation, 507–519
in Alzheimer’s disease, 47
genetic contributions to, 72
sundowning, 193, 195, 441, 510, 514
in bipolar disorder, 284
in dementia, 180–182, 181
management of, 221–230, 233
family tolerance of, 95, 670, 678
in frontotemporal lobar degeneration, 73, 140, 204, 206
sexual inappropriateness or aggression, 405–406
Benign prostatic hypertrophy, 45, 49, 443
Benton Facial Recognition Test, 131
Benton Visual Retention Test, 137
Benzodiazepine receptor agonists, 168, 441, 443, 448, 448–449, 450,
556–557. See also specific drugs
Benzodiazepines, 556–557, 560, 561. See also specific drugs
adverse effects of, 360, 470, 556–557
cognitive and motor effects, 448, 450, 557
delirium, 162, 166. 168, 212
fall and fracture risk, 360, 450, 470, 556
sexual dysfunction, 399
for agitation in dementia, 518
for anxiety disorders, 360, 363
for benzodiazepine or alcohol withdrawal, 513–514, 557
drug interactions with, 557
alcohol, 470
electroconvulsive therapy and, 267, 601
elimination half-lives of, 448, 449, 556, 557
flumazenil reversal of, 601
for grief reactions, 427
misuse of prescribed drugs, 465
for sleep disorders, 448, 449, 557
in Alzheimer’s disease, 439
restless legs syndrome and periodic limb movement disorder,
439
for symptoms of personality disorders, 502
urine toxicology testing for, 112
use in chronic obstructive pulmonary disease, 443, 557
withdrawal from, 478
Bereavement, 415–428. See also Grief reaction
adaptation to, 421–424, 428
acceptance of loss, 422–423
gender differences in, 421–422
preloss relationship and, 423
social support and, 423
after violent, stigmatized, or unexpected death, 422
complicated, 418, 419, 421, 424–426, 428
course of symptoms in, 418–420
cultural differences in responses to, 420, 428
definition of, 415
depression and, 255, 256, 416, 419, 421, 422, 426
sleep disturbances in, 416, 439–440
DSM-5 definitions related to, 420–421
persistent complex bereavement disorder, 424
dual-process model of, 418, 422, 426, 428
interventions for, 424–427, 428
normal grief reactions, 426–427
persistent complex bereavement disorder, 424–426
key points related to, 427–428
losses associated with, 415
normal vs. abnormal reactions to, 419–420, 428
personality disorders and, 497
resilience and, 418–419, 428
spousal, prevalence of, 415, 417
stage model of, 416–417
theories about adjustment to permanent losses, 416–418
Berlin Aging Group, 257
Beta-blockers. See β-Adrenergic blockers
Bethanechol, 400
Bibliotherapy, 653
BIN1 gene, in Alzheimer’s disease, 71, 190, 191
Binge drinking, 11, 26, 460, 462, 465, 476
Binswanger’s disease, 139, 141
Biomarkers, 14, 23, 99
for alcohol and drug use, 475
for Alzheimer’s disease, 70, 113, 127, 128, 184, 188, 199, 207,
214
therapies targeting, 214, 215
for depression, 262
for frontotemporal lobar degeneration, 141
for nutritional status, 630
for prion diseases, 216
Bipolar and related disorders, 244, 283–300. See also Mania
age at onset of, 249, 289–291, 294, 299
biological basis of, 291
bipolar I disorder, 283–284
bipolar II disorder, 284
clinical course and prognosis for, 249, 288–289
comorbidity with, 286–288
anxiety disorders, 286
dementia, 286, 287–288
medical conditions, 287
personality disorders, 287
posttraumatic stress disorder, 286
psychotic depression, 259
substance-related disorders, 286–287
cyclothymic disorder, 284
depressive episodes in, 285, 298, 299, 554, 555
diagnosis of, 258, 283–284
in DSM-5, 283–284
due to another medical condition, 283, 284
epidemiology of, 284–286
functional impairment in, 288, 289, 291
genetics of, 61, 75
overlap with schizophrenia and depression, 75
imaging studies in, 292–294
magnetic resonance imaging, 292–294
poststroke, 292
volumetric findings, 292
key points related to, 299–300
mortality and, 288, 299
among nursing home residents, 245
other specified or unspecified, 283, 284
prevalence of, 245, 284–286, 299
gender and, 286
in treatment settings, 285–286
with psychosis, 291
substance/medication-induced, 283, 284
suicidality and, 289
Bipolar disorder treatment, 294–299
effect of age of first manic episode on, 249
electroconvulsive therapy, 249, 299, 591, 593
key points related to, 299–300
omega-3 fatty acids, 628
pharmacotherapy, 249
antidepressants, 298
antipsychotics, 298–299, 547
carbamazepine, 295, 296–297, 555
for depressive episodes, 298, 299, 554, 555
gabapentin, 556
lamotrigine, 295, 297–298, 554–555
lithium, 295, 300, 553–554
oxcarbazepine, 295, 555
pregabalin, 556
topiramate, 556
valproate, 295, 296, 554
Systematic Treatment Enhancement Program for Bipolar
Disorder, 289, 298
Bisexuality, 109, 391, 677
Bisphosphonates, 42
Bladder outlet obstruction, 49, 403
Bleeding, drug-related, 527
selective serotonin reuptake inhibitors, 532
serotonin-norepinephrine reuptake inhibitors, 537
Blood glucose testing, 108, 109
Blood pressure. See also Hypertension; Hypotension
changes with aging, 36, 53
electroconvulsive therapy effects on, 268, 596, 597, 602, 603
Blood urea nitrogen, 108
“Blues,” 91
BMI (body mass index), 52, 632
Body dysmorphic disorder, 373
Body mass index (BMI), 52, 632
Bone changes with aging, 39, 40, 42, 51. See also Osteoporosis
Bone metabolism, drug effects on
selective serotonin reuptake inhibitors, 536
serotonin-norepinephrine reuptake inhibitors, 537
valproate, 554
Borderline personality disorder, 492, 493, 494, 498, 499–500
Boston Naming Test, 40
Bradycardia
drug-induced
antipsychotics, 115
β-blockers, 114
cholinesterase inhibitors, 218, 558, 559
selective serotonin reuptake inhibitors, 532
electroconvulsive therapy and, 596, 597
Bradykinesia, 137, 142, 205
Bradyphrenia, 137, 142
Brain biopsy, 187
Brain imaging. See Imaging studies
Brain stimulation therapies, 589–611
deep brain stimulation, 607, 609–610
electroconvulsive therapy, 266–270, 589–607
future of, 610
key points related to, 610–611
transcranial direct current stimulation, 607, 608–609
transcranial magnetic stimulation, 270, 607, 607–608
vagus nerve stimulation, 607, 609
Breathing changes with aging, 36–37
Breathing-related sleep disorders, 435, 437–438
Brief dynamic therapy, for depression, 264
Brief Intervention and Treatment for Elders (BRITE) project, 655–656
Brief interventions for substance-related disorders, 475–476, 655–656
Brief Visuospatial Memory Test—Revised, 130, 135
Bright light therapy, 222, 441, 696
Briquet’s syndrome, 375
BRITE (Florida Brief Intervention and Treatment for Elders) project,
655–656
Bromocriptine, 400
Bronchospasm, electroconvulsive therapy-induced, 598
Browning, Robert, 33, 34
Buprenorphine patch, 518
Bupropion, 540–541
adverse effects of, 265, 397, 541
psychosis, 541
seizures, 541
for antidepressant-induced sexual dysfunction, 400
in bereavement, 425
cytochrome P450 enzyme inhibition and drug interactions with,
535, 541
for depression, 265, 539, 540–541
augmentation therapy, 541
compared with paroxetine, 530
in dementia, 228
dosing of, 265, 266, 534, 539
pharmacokinetics of, 534
Buschke Selective Reminding Test, 130, 135
Buspirone, 400, 557–558
ECA (Epidemiologic Catchment Area) study, 6, 8, 15, 17, 18, 23, 24,
102, 245, 250, 285, 288, 338, 349, 467, 480
ECG. See Electrocardiography
Economic costs
of aging population, 4
of delirium, 155
of dementia, 177
ECT. See Electroconvulsive therapy
ED. See Erectile disorder
Educational strategies with families, 223, 225, 509–510, 680
EEG. See Electroencephalography
EIF4G1 gene, in Parkinson’s disease, 74
Ejaculation
delayed, 397
premature (early), 397, 404–405
Elder mistreatment, 670
Electrocardiography (ECG), 113–115. See also QTc interval
before/during drug treatment
carbamazepine, 297
tricyclic antidepressants, 265
drug effects on
antipsychotics, 114, 114, 514, 552
β-blockers, 114
citalopram, 114–115, 227, 516, 529, 532
lithium, 114, 114
mirtazapine, 540, 542
serotonin-norepinephrine reuptake inhibitors, 537
trazodone, 542
tricyclic antidepressants, 113–114, 114, 265
before electroconvulsive therapy, 267
for suspected drug overdose, 113, 115
Electroconvulsive therapy (ECT), 589–607
for bipolar disorder, 249, 299, 591, 593, 606
for catatonia, 591
continuation or maintenance treatment with, 269, 592–593, 606–
607
for depression, 247–248, 259, 266–270, 590–591, 600
compared with antidepressants, 590
compared with repetitive transcranial magnetic stimulation,
608
in dementia, 269–270
effectiveness of, 266, 268–269
evaluation before initiation of, 266, 599–601
components of, 599
deciding whether to recommend ECT, 593, 599–600
informed consent, 600–601
medication management, 266, 267–268, 601
future of, 610
history of, 589–590
indications for, 590–591
key points related to, 610
for Parkinson’s disease, 591
patient/family education about, 266
response of older adults to, 591–592
risks and adverse effects of, 268, 593–599
cardiovascular disorders, 596–597
central nervous system disorders, 595–596
cognitive effects, 268, 269, 594, 596, 598, 607
endocrine disorders, 597
gastrointestinal disorders, 598
genitourinary disorders, 598
hematological disorders, 597–598
metabolic disorders, 597
mortality, 593–594
musculoskeletal disorders, 598
in older adults, 598–599
pulmonary disorders, 598
for schizoaffective disorder, 591
for schizophrenia, 589, 590, 591
technique for, 268, 602–606
anesthesia, 602–603
anticholinergic medications, 268, 596, 597, 602
determining seizure adequacy, 268, 605
inpatient vs. outpatient settings, 602
number and frequency of treatments, 268, 605–606
physiological monitoring, 603
stimulus dosing, 604–605
stimulus electrode placement, 268, 603–604
trends in utilization of, 590
use in older adults, 269, 590
use in patients with medical illness, 270, 594–598
Electroencephalography (EEG), 119–120
in Creutzfeldt-Jakob disease, 113, 120, 186, 209
in delirium, 119, 161–162, 162, 187
in dementia, 119–120, 186–187
drug-induced abnormalities on
olanzapine, 549
risperidone, 547
for electroconvulsive therapy, 268, 599, 603
in frontotemporal lobar degeneration, 186
indications for, 119, 162
Electrolyte disorders, 43. See also specific electrolytes
agitation and, 509
delirium and, 46, 162, 163, 164, 169
laboratory testing for, 108–109, 163
before carbamazepine use, 296
lithium-induced, 553
sexual dysfunction and, 399
Elimination half-life of drugs, 44–45, 53
antidepressants, 533–534
benzodiazepines, 448, 449, 556, 557
cholinesterase inhibitors, 559
lithium, 295
sleep medications, 448, 449, 450, 557
valproate, 554
Employment
of family caregivers, 670
of older persons, 4
retirement from, 33, 251, 261, 381, 395, 446, 464, 468, 494, 497
work rehabilitation programs for patients with psychosis, 321
Encephalopathy
cancer-associated, 207
due to alcohol intoxication, 207
Hashimoto’s, 207, 209
hepatic, 514, 591
limbic, 209
spongiform, 207, 208
subcortical arteriosclerotic, 267
Wernicke’s, 117, 117
Endocrine system
changes with aging, 38–41
adrenal androgens, 38
adrenal medulla and sympathetic nervous system, 39
antidiuretic hormone, 38
corticotropin and cortisol, 38
estrogen, 40–41
growth hormone, 39
insulin, 41
musculoskeletal effects of, 51
parathyroid hormone, vitamin D, and calcium regulation, 39
prolactin, 38
renin, angiotensin and aldosterone, 39
testosterone, 40
thyroid, 41
depression and, 252–253, 260
electroconvulsive therapy in disorders of, 597
Energy requirements with aging, 632
Environmental factors, 22, 26
assessment of, 93
falls due to, 47, 48
interaction with genetic factors, 21–22, 66, 80
in depression, 22, 79
in management of neuropsychiatric symptoms of dementia, 225
nutritional status and, 629
EPESE. See Established Populations for Epidemiologic Studies of the
Elderly
EPHA1 gene, in Alzheimer’s disease, 190, 191
Epidemiologic Catchment Area (ECA) study, 6, 8, 15, 17, 18, 23, 24,
102, 245, 250, 285, 288, 338, 349, 467, 480
Epidemiology of psychiatric disorders in elderly persons, 3–26
anxiety disorders, 334–335, 338
bipolar and related disorders, 284–286
case identification, 4, 5–8
definition of, 3
delirium, 155–156, 157
delusional disorder, 314
demographic factors, 3–5
depression, 244–245
descriptive epidemiology, 5
distribution of disorders, 4, 8–18, 10, 12, 13, 16–17, 26
in treatment settings, 15, 18, 19
etiological studies, 5, 21–23
health service utilization, 5, 21–23
historical studies, 5, 18–20, 21
key points related to, 26
longitudinal studies, 8, 18, 23
Alzheimer’s disease and folate intake, 111
anxiety disorders, 333, 357
bereavement, 416, 418, 422, 423
bipolar disorder, 291
depression, 249, 253, 254, 255, 256
maturation of defenses, 496
nutrition and dementia, 620, 621, 622
personality disorders, 495, 496, 498, 503
schizophrenia, 310
substance-related disorders, 468, 480
substance-related disorders, 8–9, 18, 459–460, 460, 461, 465–
468, 482
uses of epidemiological studies, 4–5
Epigenomics, 123, 123
Epilepsy. See also Seizures
electroconvulsive therapy in, 589, 601
electroencephalography in, 119
psychogenic nonepileptic seizures and, 378
schizophrenia and, 589
Epinephrine, 39, 164
Epworth Sleepiness Scale, 229
Erectile disorder (ED), 396, 397, 403–404, 408
Alzheimer’s disease and, 405
etiology of, 403
laboratory testing for, 399
testosterone deficiency and, 40
treatment of, 389, 400, 403–404
Erikson’s stage-theory of late-life development, 496, 500
Erythromycin, 297, 554, 557
Escitalopram, 528
adverse effects of
cardiovascular effects, 115, 227
cognitive effects, 532
for anxiety disorders, 361, 528
cytochrome P450 enzyme inhibition and drug interactions with,
535
for depression, 264, 528, 530, 532
with insomnia, 440
vs. serotonin-norepinephrine reuptake inhibitors, 536–537,
540
dosing of, 266, 533
indications for, 528
for neuropsychiatric symptoms in dementia, 227, 529
pharmacokinetics of, 533
Esophageal function, 37
Established Populations for Epidemiologic Studies of the Elderly
(EPESE), 34
East Boston, 11, 12
Hispanic, 13, 22
Iowa, 13
North Carolina, 250
Estazolam, 448
Estrogen(s)
Alzheimer’s disease and, 189, 213
bone mass and, 42, 51
postmenopausal decline in production of, 40, 393, 394, 403
nocturia and, 443
symptoms related to, 444
to reduce aggression in dementia, 407, 518
replacement therapy with, 41
for psychosis, 312
thyroid effects of, 110
Eszopiclone, for insomnia, 448, 448, 449, 556, 557
in depression, 440
dosing of, 449
with hot flashes, 444
in Parkinson’s disease, 442
Ethical issues
Alzheimer’s prevention studies, 215
genetic testing, 120, 121–122, 123
genomic medicine, 80
sexuality and dementia, 405
Etiological studies of psychiatric disorders in elderly persons, 5, 21–
23, 26
Etomidate, for electroconvulsive therapy, 602
Euphoria
delirium and, 159
mania and, 244, 283, 284
orgasm and, 393
Evidence-Based Caregiver Intervention Resource Center, 673
Executive function, 46, 180
in Alzheimer’s disease, 134, 315
in bipolar disorder, 291
in cerebrovascular disease, 140
in dementia, 181
discussing deficits with family, 674
effect of exercise programs on, 210
in frontotemporal lobar degeneration, 136, 140, 204
in generalized anxiety disorder, 357, 654
in hoarding disorder, 357
in Lewy body neurocognitive disorder, 201
in major neurocognitive disorder, 181
in mild neurocognitive disorder, 183
normal age-related declines in, 144
in normal-pressure hydrocephalus, 206
in Parkinson’s disease, 137, 202
effect of cholinesterase inhibitors on, 220
in personality disorders, 499
psychotherapy and, 654, 657–658
tests of, 99, 129, 130, 144, 185
in vascular dementia, 184
Exercise. See Physical activity
Exposure therapy
for anxiety disorders, 359–360, 653
for complicated grief, 425
Extrapyramidal symptoms
in Alzheimer’s disease, 201, 315
in corticobasal syndrome, 205
in Creutzfeldt-Jakob disease, 138, 208, 209, 209
drug-induced
antipsychotics, 322, 514, 515, 545
atypical antipsychotics, 321, 322, 515, 545, 547, 549,
551, 707–709, 711, 714
rivastigmine, 218
selective serotonin reuptake inhibitors, 536
in frontotemporal lobar degeneration, 205
in Lewy body neurocognitive disorder, 142
antipsychotic sensitivity and, 226–227, 322
in paraneoplastic syndromes, 209
in Parkinson’s disease, 137, 142
in progressive supranuclear palsy, 137
Extraversion, 193, 495, 498
Fluoxetine, 528
combined with olanzapine, for bipolar disorder, 298
for depression, 264, 265, 266, 530, 532
with insomnia, 440
dosing of, 265, 266, 533
drug interactions with
bupropion, 541
cytochrome P450 enzyme inhibition and, 535, 541
valproate, 554
warfarin, 93
indications for, 528
pharmacokinetics of, 533
switching to monoamine oxidase inhibitor from, 266
for vasomotor menopausal symptoms, 444
Flurazepam, 448, 449, 557
Fluvoxamine, 528
cytochrome P450 enzyme inhibition and drug interactions with,
535
olanzapine, 549
for depression, 530, 532
dosing of, 532, 533
indications for, 528
pharmacokinetics of, 533
Folate
deficiency of, 111, 621
alcoholism and, 627
laboratory testing for, 111, 262
dementia and, 189, 621
depression and, 262, 622
L-methylfolate treatment, 628
selective serotonin reuptake inhibitor response and, 628
dietary intake of, 633
mild cognitive impairment and, 210
supplementation of, 635–636
Follicle-stimulating hormone, 40
Four pillars of dementia care, 211–212, 233
Fractures
electroconvulsive therapy–related, 268, 598
hip
alcohol consumption and, 470
delirium and, 169
depression and, 247, 253
due to falls, 47
medication–related, 536, 557
subacute nursing home care for, 727
imaging of, 115, 116, 267
incontinence and, 50
medication-related, 527
antidepressants, 227, 536, 540
atypical antipsychotics, 545
benzodiazepines, 470, 557
bisphosphonates, 42
minimizing risk during exercise, 638
nutritional deficiencies and, 625
benefits of vitamin D and calcium supplementation, 636
osteoporotic, 36, 268, 638
estrogen replacement therapy and, 41
skull, 116
vertebral, 36, 268
Frailty, 45, 50–51, 52, 54
agitation and, 513
physical restraint for, 718
definition of, 51
delirium and, 155, 513
dementia and, 215, 228, 513
depression and, 246–247, 694
inadequate energy intake and, 632
of nursing home residents, 690, 694, 717, 718, 732
nutritional, 625, 632
pharmacotherapy and
antidepressants, 228
mirtazapine, 542
selective serotonin reuptake inhibitors, 532, 717
venlafaxine, 717
benzodiazepines, 470, 557
effect on drug protein binding, 44
lithium, 295
for nursing home residents, 717, 732
Frontal Assessment Battery, 131, 136, 185
Frontotemporal lobar degeneration (FTLD; frontotemporal dementia),
190, 204–206
behavioral variant, 73, 141, 204
clinical presentations of, 73, 141, 204
corticobasal syndrome and, 205
diagnosis of, 206
genetic testing, 73–74, 81
plasma assays, 113
TDP-43, 73, 113, 141, 190, 206
differentiation from Alzheimer’s disease, 118, 206
genetics of, 73–74, 113, 206
with motor neuron disease, 73, 141, 205
neuropathology of, 73, 74, 140, 141, 190, 192, 196, 198–199,
205–206
neuropsychological assessment in, 136, 140–141, 144
personality changes in, 73, 136, 140, 190, 204, 499
pharmacotherapy for, 233
antidepressants, 228
cholinesterase inhibitors, 220
prevalence of, 140, 204
primary progressive aphasia, 73, 140–141, 204–205
progressive supranuclear palsy and, 205
with psychosis, 317
semantic dementia, 73, 141, 204, 205
Functional Adaptation Skills Training (FAST), 320
Functional magnetic resonance imaging, 116
in dementia, 188
in generalized anxiety disorder, 358
in somatic symptom and related disorders, 380
Functional neurological symptom disorder. See Conversion disorder
Functional status/impairment, 46. See also Activities of daily living
performance; Instrumental activities of daily living performance
agoraphobia and, 336
Alzheimer’s disease and, 190, 192–193, 194, 195
pharmacotherapy effects on, 213, 217, 219
with psychosis, 316
assessment of, 51, 52, 91, 101, 104
functional analysis, 222–223
instrument for, 101
neuropsychological evaluation, 128, 129, 132
by occupational therapist, 186, 231
bipolar disorder and, 288, 289, 291
capacity to live alone and, 683
case identification based on, 6–7
conversion disorder and, 378
delirium and, 155, 157, 165, 167, 170
dementia and, 180, 185, 186, 187, 659
depression and, 23, 143, 144, 243–244, 246, 247, 250, 253, 263,
421, 694
exercise and, 637, 638
falls and, 352, 618
frailty and, 51
frontotemporal lobar degeneration and, 144
generalized anxiety disorder and, 347
Lewy body neurocognitive disorder and, 141, 142
mild cognitive impairment and, 135, 184
of nursing home residents, 692, 693, 694, 720, 728, 731
in special care units, 726, 727
nutritional status and, 630, 634
pain and, 377
Parkinson’s disease and, 141
personality disorders and, 491, 492
schizophrenia and, 310–311
somatoform disorders and, 374, 376, 381
substance-related disorders and, 459, 469
vascular dementia and, 200
working with families of patients with, 94, 669, 674
Funeral observances, 420
FUS gene, in frontotemporal lobar degeneration, 73, 141
Gabapentin
for bipolar disorder, 556
dosing of, 556
for neuropathic pain, 556
for restless legs syndrome and periodic limb movement disorder,
439
for vasomotor menopausal symptoms, 444
GAD. See Generalized anxiety disorder
GAF (Global Assessment of Functioning) Scale, 289
Gait disturbances. See also Ataxia
dementia and, 182
drug-induced
antipsychotics, 515, 549, 709
bupropion, 541
falls and, 48
hyponatremia and, 109
Lewy body neurocognitive disorder and, 201
normal-pressure hydrocephalus and, 137, 206, 215–216
Parkinson’s disease and, 137, 202
prion diseases and, 120, 208, 209
vascular neurocognitive disorder and, 200
Galantamine
adverse effects of, 211, 218, 559
for Alzheimer’s disease, 47, 216–217, 218, 220, 558
dosing of, 218, 559
drug interactions with, 559
in mild cognitive impairment, 211
preparations of, 218
Gallbladder function, 38
Gastrointestinal system
changes with aging, 37–38, 529
drug effects on, 45, 527
calcium supplements, 636
chloral hydrate, 448
cholinesterase inhibitors, 218, 558, 559
clozapine, 552
memantine, 218
olanzapine, 549
phosphodiesterase 5 inhibitors, 404
selective serotonin reuptake inhibitors, 227, 264–265, 532,
541
serotonin-norepinephrine reuptake inhibitors, 537
tricyclic antidepressants, 544
vilazodone, 543
vortioxetine, 543
electroconvulsive therapy effects on, 598
GDS (Geriatric Depression Scale), 6, 18, 52, 100, 131, 243, 262, 263,
706, 729
Gender factors, 4
anxiety disorders and, 339
bereavement and, 415–416, 421–422
bipolar disorder and, 286
dietary reference intakes and, 633
late-onset schizophrenia and, 311–312
prolactin levels and, 38
restless legs syndrome and, 439
sexual behavior and, 390
sexual response cycle and, 393–395, 394
sexually transmitted diseases and, 395
substance-related disorders and, 465, 468
prescription drugs, 469
treatment response and, 479
suicide and, 20
Generalized anxiety disorder (GAD), 346–349
age at onset of, 337
case example of, 346
chronicity of, 348
clinical features of, 339, 340, 346, 348
cognitive deficits in, 357
comorbidity with
dementia, 358
depression, 7, 346, 348, 349, 355
diagnosis in older adults, 336
in DSM-5, 347
hypothalamic-pituitary-adrenal axis in, 357–358
iatrogenic outcomes of lack of detection of, 346, 348
imaging studies in, 358
poststroke, 339
prevalence of, 15, 16–17, 18, 334, 338, 339, 347–348
treatment of
pharmacotherapy, 361, 528
psychotherapy, 359, 360, 653–654, 659
Genetic Information Nondiscrimination Act, 67, 122
Genetic risk factors for psychiatric disorders, 21–22, 26
interaction with environmental factors, 21–22, 66, 79, 80
Genetic testing, 66–67, 80, 81, 107, 120–122, 123
for Alzheimer’s disease, 70–72, 188
apolipoprotein E ε4 allele, 71–72, 120
early-onset, 70
late-onset, 71–72
confidentiality of, 122
ethical and psychological concerns in, 121–122
for frontotemporal lobar degeneration-associated genes, 73–74
HLA-B*1502 allele test, 121, 297
for Parkinson’s disease, 75
pharmacogenomics, 121
Genetics, 65–66, 68–81
of Alzheimer’s disease, 22, 61, 64, 66, 68–72, 189–192, 191, 620
of bipolar disorder, 75, 251
definitions related to, 63–64
of depression, 22, 75, 79, 251–252
vascular depression, 252
family assessment of, 92
of frontotemporal lobar degeneration, 73–74, 113, 206
key points related to, 80–81
modes of inheritance, 66
of obsessive-compulsive disorder, 349
of Parkinson’s disease, 64, 74–75
of schizophrenia, 75–79
Genito-pelvic pain/penetration disorder, 41, 396, 397
Genome-wide association studies (GWASs), 64, 65, 67, 77–78, 79
in Alzheimer’s disease, 66, 71, 72, 190–192
in schizophrenia, 78
Genomics in geriatric psychiatry, 61–67, 123
copy-number variations, 64–65
de novo mutations, 65
genomic medicine, 66, 80
Human Genome Project, 61, 62, 122
practical framework for clinician, 65–66
single-nucleotide polymorphisms, 62–64
Genotype, 63
Geriatric assessment, 51–53, 54
components of, 52
definition of, 52
effectiveness of, 52–53
in-home, 52
neuropsychological, 97, 99, 127–145, 180, 185–186
to prevent delirium, 169
psychiatric interview, 89–104
Geriatric Depression Scale (GDS), 6, 18, 52, 100, 131, 243, 262, 263,
706, 729
Geriatric Mental State Schedule, 100–101
Geriatric syndromes, 45–51
falls, 47–48
frailty, 50–51
key points related to, 54
memory loss and dementia, 46–47
polypharmacy, 50
urinary incontinence, 48–50
Geriatrician, 53, 54
Gerstmann-Sträussler-Scheinker syndrome, 208
“Get up and go” test, 52
Gingko biloba, 210, 213
Global Assessment of Functioning (GAF) Scale, 289
Glomerular filtration rate, 44, 527, 529
Glucose dysregulation, 41, 108, 619. See also Diabetes mellitus;
Hyperglycemia; Hypoglycemia
atypical antipsychotic–induced, 109, 109, 299, 317, 319, 545,
547, 549, 551
delirium and, 164, 167
medroxyprogesterone-induced, 407
Glutamate
in Alzheimer’s disease, 199
in bipolar disorder, 291
in delirium, 164
medication actions on
acamprosate, 478
memantine, 217, 559
in schizophrenia, 78
Glycopyrrolate, for electroconvulsive therapy, 602
Go4Life program, 636
Gonadotropin-releasing hormone, 40
Gonorrhea, 395
GPNMB gene, in Parkinson’s disease, 75
GRIA1 gene, in schizophrenia, 78
Grief reaction, 14, 415. See also Bereavement
as adjustment to permanent loss, 416–418
anticipatory, 415, 419
assessment measure for, 423
chronic, 418–419
course of, 418–420
culture and, 420, 421, 424, 428, 439
vs. depression, 418
duration of, 417
interventions for, 424–427
losses associated with, 415
normal vs. abnormal, 419–420, 428
resolution of, 417, 418
risk factors for intensification of, 421–424
stages of, 416–417
traumatic, 419
GRIN2A gene, in schizophrenia, 78
GRM3 gene, in schizophrenia, 78
GRN gene, in frontotemporal lobar degeneration, 73
Grooved Pegboard test, 131
Group psychotherapy
for depression, 651, 652
in nursing homes, 697, 698–702, 729
for patients with cognitive impairment, 658, 729
Growth hormone, 39, 51
Growth hormone–releasing hormone, 39
Guilt feelings of family caregivers, 675
GWASs. See Genome-wide association studies
Labetalol, 602
Laboratory testing, 107–123
for agitation, 508
for alcohol and drug use, 112, 475
in bipolar disorder, 294
for carbamazepine use, 296–297
in delirium, 161, 162, 163
in dementia, 46, 186–188
cerebrospinal fluid analysis and plasma assays, 112–113
in depression, 262, 263
for diabetes mellitus, 108, 112
electrocardiography, 113–115, 114
before electroconvulsive therapy, 267, 599
electroencephalography, 119–120
genetic testing, 66–67, 80, 81, 107, 120–122
imaging studies, 115–119, 117
key points related to, 123
for mental status changes, 108, 112, 115, 119, 162, 508
to monitor for metabolic effects of atypical antipsychotics, 109,
319
for nutritional assessment, 630
omics technologies, 122–123, 123
serologic tests, 107–108
chemistry tests, 108–109, 109
hematologic tests, 108
for HIV infection, 110
for syphilis, 109–110
testosterone level, 40
thyroid function tests, 110–111, 111
vitamin B12, folate, and homocysteine levels, 111–112
for sexual dysfunction, 399
for sleep disorders, 445
toxicology tests, 112, 475
urinalysis, 112
Lacunar infarcts, 117, 117, 179, 200
Lamotrigine, 554–555
adverse effects of, 297–298, 555
for bipolar disorder, 295, 297–298, 554–555
drug interactions with, 554, 555
interaction with valproate, 296
pharmacokinetics in elderly persons, 297
Language
age-related changes in, 46
assessment of, 99, 129
tests of, 130, 133
Language deficits
Alzheimer’s disease and, 134
delirium and, 158, 160
dementia and, 179, 180, 181, 183, 184
frontotemporal lobar degeneration and, 73, 140, 190, 204–205,
206
Lewy body neurocognitive disorder and, 142
prion diseases and, 208
vascular neurocognitive disorder and, 136
LASA (Longitudinal Aging Study Amsterdam), 13, 23, 255, 334, 338
LBD. See Lewy body neurocognitive disorder
Learning, 46, 53
Leiden 85+ Study, 13, 23
Leipzig Longitudinal Study of the Aged, 12
Lennon, John, 33, 34
Lesbian, gay, bisexual, or transgender (LGBT) persons, 109, 391, 677
Leukocytosis, 51
lithium-induced, 108
Leukoencephalopathy, 199
Leukopenia, drug-induced
carbamazepine, 297
clozapine, 319
Leuprolide, 399
Levomilnacipran, 536
cytochrome P450 enzyme inhibition and drug interactions with,
535
for depression, 537, 538
dosing of, 266, 533, 538
pharmacokinetics of, 533
Lewy bodies, 178, 200, 203
Alzheimer’s disease with, 179
in cognitively normal persons, 178, 179
Lewy body disorders, 190, 200–204
clinical presentations of, 201
neuropathology of, 142, 178–179, 200, 202
neuropsychological assessment in, 136, 137, 141–142, 144
pharmacotherapy for psychosis in, 322, 547
α-synuclein in, 74, 141, 190, 200, 203, 203, 441
Lewy body neurocognitive disorder (Lewy body neurocognitive
disorder (DLB)), 190, 200–202
clinical presentation of, 142, 201
cognitive enhancers for, 220
course and prognosis of, 202
diagnosis of, 201
differential diagnosis of, 202
imaging studies in, 201
neuropsychological assessment in, 136, 141–142, 144
prevalence of, 12, 46, 201
with psychosis, 317, 322, 517
treatment of, 233
antidepressants for depression, 228
neuroleptic sensitivity and, 136, 190, 201, 226–227, 322,
517
for psychosis, 322, 517, 547
rivastigmine for neuropsychiatric symptoms, 517
Lexical fluency tests, 130
LGBT (lesbian, gay, bisexual, or transgender) persons, 109, 391, 677
Life events
bereavement, 415–428
financial problems, 256
as focus of cognitive-behavioral therapy, 651
health problems (See Medical conditions)
legal problems, 256
marital problems/divorce, 22, 255, 256, 261, 396, 415, 424, 469
personality disorders and, 497
retirement, 33, 251, 261, 381, 395, 446, 464, 468, 494, 497
as risk factors for psychiatric disorders, 22–23
adjustment disorder with depressed mood, 261–262
anxiety associated with medical illness, 356
depression, 79, 245, 247, 248, 252, 254, 255
hoarding disorder, 337, 349
mania, 249
personality disorders, 497
substance-related disorders, 468
Life expectancy, 4, 33–34, 53
Life review therapy, 652, 653
Ligament changes with aging, 42
Limbic encephalopathy, 209
Lithium, 553–554
adverse effects of, 295, 553
cognitive effects, 553
delirium, 553
hypothyroidism, 111, 295
renal effects, 108, 553
sexual dysfunction, 399
for bipolar disorder, 295, 553
dosing of, 249, 295, 300, 553
drug interactions with, 295, 553
effect on suicidality, 554
electrocardiogram effects of, 114, 114
electroconvulsive therapy and, 267, 595, 601
laboratory monitoring for use of, 112
neuroprotective effects of, 554
pharmacokinetics of, 295, 553
prophylactic, after course of electroconvulsive therapy, 592
therapeutic plasma level of, 112, 295
toxicity of, 295, 553
for treatment-resistant depression, 553
Liver
changes with aging, 38, 527, 529
drug metabolism by, 44
Liver function tests, 109, 163, 186
Living arrangements of older adults, 3–4
assisted living facilities, 212, 629, 676, 679, 684, 690, 691, 696
living alone, 232, 625, 670
assessing capacity for, 681, 684
documenting problems with, 244
nursing homes, 689–733
Long-term care. See also Nursing homes
behavioral problems of residents in, 693
agitation, 515
dementia among residents in, 11, 231, 670
depression among residents in, 18, 245, 693
selective serotonin reuptake inhibitors for, 528, 530–531
elders living in facilities for, 3–4
genetic discrimination and, 67
nutritional assessment of residents in, 629
pain among residents in, 376
prevalence of psychiatric disorders among residents in, 9, 11, 18
quality of facilities for, 679
sexuality in settings for, 389, 391–392, 406, 407
somatic preoccupation among residents in, 381
Long-Term Care Minimum Data Set, 630
Longitudinal Aging Study Amsterdam (LASA), 13, 23, 255, 334, 338
Lorazepam, 448, 556, 557, 710
Loxapine, use in nursing homes, 704, 705
LRRK2 gene, in Parkinson’s disease, 74, 75
Lumbar puncture, 162, 163, 206, 215
Lung changes with aging, 36–37
Lurasidone, 552
for bipolar depression, 298
receptor blockade of, 550
Luteinizing hormone, 40
Race/ethnicity, 4
antidepressant use and, 25
apolipoprotein E ε4 allele, Alzheimer’s disease risk and, 70–71
cultural norms and bereavement responses, 420
depression and, 9
mental health service use and, 24
pharmacogenomic testing and, 121
HLA-B*1502 allele test, 121, 297
suicide mortality and, 20, 26
vascular depression and, 253
Radiography, plain film, 115
Ramelteon, 443, 448, 448, 449
Rapid eye movement (REM) sleep behavior disorder, 435
Lewy body neurocognitive disorder and, 201
Parkinson’s disease and, 202, 204, 441
quetiapine for, 230
Rapid plasma reagin test, 110
Rapidly progressive dementias (RPDs), 207–210
differential diagnosis of, 209–210
nonprion causes of, 207
prion diseases, 207–209, 207–210
treatment of, 216
Rash, drug-induced
carbamazepine, 297, 555
lamotrigine, 297, 555
Rating scales, 98–101. See also Assessment instruments
Receptors
acetylcholine
muscarinic, 547, 549, 550, 551, 552
nicotinic, 217
α-adrenergic, 39, 542, 550, 551
β-adrenergic, 39
age-related changes in, 527
angiotensin II, 252
atypical antipsychotic blockade of, 550, 551, 552
dopamine D2, 550, 551
glucocorticoid, 164
histamine H1, 551
insulin, 41
serotonin
5-HT1A, 541, 543
5-HT2A, 252, 542, 550
Relaxation techniques
for anxiety disorders, 359, 360, 654, 655, 660
for delirium, 170
for insomnia, 446
for nursing home residents, 697, 702
for sexual dysfunctions, 403, 404
Reliability of diagnosis, 7–8
Religious Order Study, 200
Religious participation
bereavement and, 419, 420, 421
effect on recovery from depression, 248, 264
of family, 681
psychotherapy and, 650
REM sleep behavior disorder. See Rapid eye movement sleep behavior
disorder
Remifentanil, 602
Reminiscence therapy, 210, 653
in nursing homes, 697, 698–702
Renal effects of drugs
angiotensin-converting enzyme inhibitors, 44
gabapentin, 556
lithium, 108, 553
nonsteroidal anti-inflammatory drugs, 44
Renal system changes with aging, 39, 43–44, 527, 529
drug prescribing and, 44, 53–54
Renin, 39, 44
Repetitive transcranial magnetic stimulation (rTMS), for depression,
270, 608
Research Domain Criteria, 333, 341
Reserpine, 267, 399
Resident Assessment Instrument, 720
Respiratory system changes with aging, 36–37
Respite care, 94, 232, 233, 672, 675, 676, 720
Restless legs syndrome (RLS), 229, 230, 435, 439, 451
Restlessness
during alcohol withdrawal, 478
Alzheimer’s disease and, 193, 195
generalized anxiety disorder and, 346, 347
sexually inappropriate behavior and, 406
Retirement, 33, 251, 261, 381, 395, 446, 464, 468, 494, 497
Rey Auditory Verbal Learning Test, 130, 135
Rey-Osterrieth figure, 136
Ribonucleic acid (RNA), 62, 63, 123
messenger, 62, 63
noncoding, 62, 63, 64, 77, 78
schizophrenia and, 78
Rigidity
in corticobasal syndrome, 205
in Lewy body neurocognitive disorder, 201
in Parkinson’s disease, 74, 142
in progressive supranuclear palsy, 137, 205
pupillary, 35
Risk factors for psychiatric disorders in elderly persons, 21–23
environmental factors, 22
genetic factors, 21–22
social factors, 22–23
Risperidone, 546–547
adverse effects of, 319, 546, 547
hyperprolactinemia, 117, 547
metabolic effects, 547
mortality and stroke risk in elderly dementia patients, 299,
516, 546
sexual dysfunction, 398
for agitation, 515
in Alzheimer’s disease, 47, 518, 546
in bipolar disorder, 547
for delirium, 514, 547
for delusional disorder, 320
dosing of, 320, 547
for generalized anxiety disorder, 361
long-acting injectable, 546–547
for mania, 298
for neuropsychiatric symptoms in dementia, 226, 546
agitation, 47, 518, 546
for psychosis
in Alzheimer’s disease, 316, 321
schizophrenia, 318, 319, 546–547
very-late-onset schizophrenia-like psychosis, 320
receptor blockade of, 550
for tardive dyskinesia, 318
use in Lewy body disorders, 547
use in nursing homes, 697, 703, 707, 708, 711, 712
Rivastigmine, 216–217, 558
adverse effects of, 218, 559
in Alzheimer’s disease, 47, 217, 218, 220
for agitation, 517
delirium and, 168
dosing of, 218, 559, 559
in Parkinson’s disease dementia, 204, 220
preparations of, 218, 220
patch, 217, 220, 559
for psychosis in Lewy body neurocognitive disorder, 322, 517
use in nursing homes, 716
RLS (restless legs syndrome), 229, 230, 435, 439, 451
RNA. See Ribonucleic acid
Rofecoxib, in mild cognitive impairment, 210
Ropinirole, 439, 545
Rosalynn Carter Institute for Caregiving, 673
RPDs. See Rapidly progressive dementias
rTMS (repetitive transcranial magnetic stimulation), for depression,
270, 608
T lymphocytes, 43
T3 (triiodothyronine), 41, 110–111, 111, 262, 263
T4 (thyroxine), 41, 110–111, 111, 262, 263, 630
Tachycardia, electroconvulsive therapy–induced, 602
Tacrine, 216, 217, 558
Tadalafil, for erectile disorder, 400, 404
T’ai chi, 638
Tardive dyskinesia, 101, 313, 314, 317, 318, 321, 323, 324, 550, 703
Task structuring, 225
Tau proteins, 188
in Alzheimer’s disease, 71, 112, 119, 122, 128, 196, 197, 198,
203, 316
therapies targeting, 213
in corticobasal degeneration, 73
in frontotemporal lobar degeneration, 73, 113, 141, 190, 205–206
in Parkinson-plus syndromes, 113
positron emission tomography imaging of, 118, 119
in progressive supranuclear palsy, 73
TBI. See Traumatic brain injury
TCAs. See Antidepressants, tricyclic
tDCS (transcranial direct current stimulation), 607, 608–609
TDEEs (total daily energy expenditures), 632
TDP-43 (transactive response DNA-binding protein 43), 73, 113, 141,
190, 206
Temazepam, 448, 449, 557
Temperament, 284, 492
Temporal arteritis, 45
Tendon changes with aging, 42
Terfenadine, 297
Testosterone
age-related decline in, 40, 252, 394
bone mass and, 51
replacement therapy with, 40
adverse effects of, 403
in depression, 253
for erectile disorder, 403
for female orgasmic disorder, 404
for female sexual interest/arousal disorder, 403
testing for deficiency of, 40
Texas Revised Inventory of Grief—Present (TRIG-Present), 423
Theophylline, 45, 443
electroconvulsive therapy and, 595, 598
interaction with lithium, 295
Therapeutic plasma level
of carbamazepine, 516
of lithium, 112, 295
of tricyclic antidepressants, 112, 265, 544
of valproate, 296
Thiamine intake, 633
Thioridazine
delirium and, 168
QTc interval prolongation induced by, 114, 552
use in nursing homes, 704, 705
Thiothixene
before electroconvulsive therapy, 269
use in nursing homes, 706
Thirst response, 635
Thought disturbances
assessment of, 96–97
in delirium, 160
3MS (Modified Mini-Mental State), 185
Thrombocytopenia, drug-induced, 108
valproate, 296, 554
Thrombophlebitis, prevention during electroconvulsive therapy, 597–
598
Thyroid changes with aging, 41
Thyroid disorders. See Hyperthyroidism; Hypothyroidism
Thyroid function tests, 110–111, 111, 262, 263, 294
Thyroid-stimulating hormone (TSH), 110–111, 111
Thyroxine (T4), 41, 110–111, 111, 262, 263, 630
Thyroxine-binding globulin, 41, 110
Timeline followback (TLFB) method, to assess substance use, 472
TMS. See Transcranial magnetic stimulation
Tolterodine, 49
Topiramate, 554, 556
Torsades de pointes, drug-induced
antipsychotics, 114, 114
citalopram, 114–115, 227, 532
mirtazapine, 540, 542
Total daily energy expenditures (TDEEs), 632
Toxicology tests, 112, 475
Trail Making Test, 130, 133, 135–137
Training of mental health workers, 25
in nursing homes, 695, 696, 724, 725, 730, 731
to prevent delirium, 169
Transactive response DNA-binding protein 43 (TDP-43), 73, 113, 141,
190, 206
Transcranial direct current stimulation (tDCS), 607, 608–609
Transcranial magnetic stimulation (TMS), 607, 607–608
deep TMS, 608
repetitive TMS for depression, 270, 608
safety of, 607, 608
technique for, 607–608
Transcriptomics, 123, 123
Transference, 103
Transurethral resection of prostate, 50
Tranylcypromine, 266, 540, 542, 544
Trauma- and stressor-related disorders, 333
acute stress disorder, 339, 352
in DSM-5, 335
posttraumatic stress disorder, 351–352, 353–354
Traumatic brain injury (TBI), 22
agitation and, 509
conversion disorder and, 378
due to falls, 47
electroencephalography for, 119
imaging studies for, 162, 163
inappropriate sexual behavior and, 406
mania and, 249, 294
neurocognitive disorders due to, 22, 181, 183, 185
Alzheimer’s disease, 189
personality disorders and, 499, 502
Trazodone, 542–543
adverse effects of, 265, 448, 542–543
for antidepressant-induced sexual dysfunction, 400
cytochrome P450 enzyme inhibition and drug interactions with,
535, 543
for depression, 265, 542–543
compared with other antidepressants, 538
dosing of, 265, 266, 534, 542
mechanism of action of, 542
for neuropsychiatric symptoms of dementia, 227, 230, 441, 517,
542
pharmacokinetics of, 534
for sleep disorders, 230, 441, 448, 450, 542
Treatments for psychiatric disorders
brain stimulation therapies, 589–611, 607
deep brain stimulation, 607, 609–610
electroconvulsive therapy, 266–270, 589–607
transcranial direct current stimulation, 607, 608–609
transcranial magnetic stimulation, 270, 607, 607–608
vagus nerve stimulation, 607, 609
nutrition and physical activity, 617–639
pharmacotherapy, 527–560
psychotherapy, 649–660
working with families, 669–684
TREM gene, in Alzheimer’s disease, 71
Tremor
alcohol-related, 475, 478
conversion disorder and, 377
corticobasal syndrome and, 205
drug-induced
galantamine, 559
lithium, 295, 553
selective serotonin reuptake inhibitors, 264, 541
valproate, 554
Lewy body neurocognitive disorder and, 201
clozapine for, 322
Parkinson’s disease and, 74
progressive supranuclear palsy and, 137
Triazolam, 448, 557
for insomnia, 444, 449
in chronic obstructive pulmonary disease, 443
in dementia, 229
Tricyclic antidepressants (TCAs). See Antidepressants, tricyclic
Triflusal, in mild cognitive impairment, 210
TRIG-Present (Texas Revised Inventory of Grief—Present), 423
Triiodothyronine (T3), 41, 110–111, 111, 262, 263
TSH (thyroid-stimulating hormone), 110–111, 111
Twelve-step programs, 476–477
Twin studies, 61
of Alzheimer’s disease, 189
of depression, 22, 251
of personality change, 495
Yohimbine, 400
Z-Access
https://wikipedia.org/wiki/Z-Library