Vous êtes sur la page 1sur 23

Journal of Gerontology: MEDICAL SCIENCES Copyright 2003 by The Gerontological Society of America

2003, Vol. 58A, No. 4, 331–353

Review Article

Management of Alzheimer’s Disease


George T. Grossberg and Abhilash K. Desai

Department of Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, Missouri.

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
A LZHEIMER’S disease (AD) is a devastating and
debilitating neurodegenerative condition and the most
common cause of dementia in the elderly. AD may not be
anxiety and depression. During the moderate stage these
symptoms appear to abate, as neuropsychiatric manifesta-
tions such as visual hallucinations, delusions, and reversal
a single disease but rather a group of diseases with over- of sleep patterns emerge. The severe and final stage is char-
lapping pathogenetic mechanisms and clinical manifesta- acterized by motor signs such as motor rigidity and prom-
tions. The condition accounts for an estimated 60–70% of inent cognitive decline. Cognitive and functional decline
all dementing disorders in the elderly (1). Between 5% and tend to be linear throughout the 3 stages of the disease,
10% of the population aged 65 years and older, and up to whereas caregiver burden is high in severe AD, since at this
50% of those older than 85 years of age, are estimated to point patients require almost total care; the physical care
suffer from AD (2). One hundred thousand deaths per year burden is the greatest, although the neuropsychiatric care
are attributed to AD in the United States. In the last 50 burden can also be considerable.
years, AD has grown from relative obscurity to becoming
a defining characteristic of industrialized society. In 1950, at MANAGEMENT oF AD
the most 200,000 people in the United States suffered from Although the focus of many physicians is the management
the ailment. The total stands at 4 million today (3). By 2050, of AD with cholinesterase inhibitors (ChEI), it is crucial that
barring a cure, the number of U.S. sufferers is expected physicians develop a global management strategy for their
to reach 16 million, out of a total of 80 million sufferers patients with AD and their caregivers. Global management
worldwide (4). includes early accurate diagnosis and providing counseling
Despite consensus on both clinical (5) and neuropatho- and pharmacological treatment to the patient and the family/
logic (6) definitions of AD, only limited information is caregiver (Table 2). The patient and family must agree with
known about its etiology and pathogenesis. It is probably the physician on the treatment ultimately selected for use;
caused by several mechanisms that include both genetic this decision must include duration and cost considerations.
and environmental influences (7). The disease manifests as Both the patient’s premorbid functioning and potential im-
a relentless decline in a broad range of intellectual and func- pact on their quality of life must be taken into account when
tional abilities, followed eventually by death. In addition, making any treatment decisions for AD patients. A thorough
as many as 90% of people with AD demonstrate clinically evaluation of comorbid medical and psychiatric disorders
significant behavioral and psychological symptoms at some and a review of current medications (including discontinu-
point in the course of the illness, causing severe emotional ation of unnecessary or harmful medications) are important.
suffering. Caring for patients with AD imposes an immense As over-the-counter products are popular, physicians are en-
burden on caregivers (8). couraged to ask about their use and to provide appropriate
Although most patients with AD develop the disease after counsel regarding their safety and efficacy. During the eval-
the age of 65, 5–10% develop the disease at a younger age uation phase, emphasis must be placed on establishing a
(early-onset AD). Less than 10% of patients with AD, good rapport with the patients and their families. Regularly
almost all being early-onset AD, have a familial AD with an scheduled visits, every 1–2 months initially and then every 3
autosomal dominant pattern of inheritance. Abnormal genes months, for health maintenance and routine patient assess-
on chromosomes 21, 14, and 1 appear to account for the vast ment will assist in maximizing independence and functioning
majority of cases of the early-onset familial AD (9–11). All and will minimize behavioral disturbances associated with
these genes are almost fully penetrant. the progression of the disease. Families with early-onset fa-
There are 3 stages of AD—mild, moderate, and severe— milial AD may be encouraged to enroll in a research database
with cognitive and functional decline stretching over 5–8 for potential future curative therapies.
years on an average (range 2–20 years) (Table 1) (12). A
recent study indicates that people with AD often die within Naming the Disease
about 3 years of diagnosis, especially those above the age of The management of AD begins with naming the disease
85—a far grimmer prognosis than was previously thought to and evaluating its severity. The clinical diagnosis of AD can
exist (13). Without treatment, the initial, mild stage usually be accurate 90% of the time. Recent data indicates that AD
lasts 2–3 years, during which time patients show short-term can be accurately diagnosed even in very mildly impaired
memory impairment, often accompanied by symptoms of individuals (14). A definitive diagnosis of AD still can be

331
332 GROSSBERG AND DESAI

Table 1. Measures of Global and Cognitive Dysfunction Associated Table 2. Goals of Management of Alzheimer’s Disease (AD)
With the 3 Stages of Alzheimer’s Disease
Patient care goals
Measure Educating the patient regarding the disease
Global Mini-Mental Symptomatic treatment with a cholinesterase inhibitor (ChEI)
Duration Deterioration State Exam Global Reducing excess disability
Stage (years) Scale (score)* (score)y Autonomy Addressing safety concerns (driving, firearms, wandering, poisonous
substances, etc.)
Mild 2–3 3–4 26–18 Independent living Addressing ability to make medical and financial decisions and capacity to
Moderate 2 5 17–10 Supervision required live independently
Severe 2–3 6–7 9–0 Total dependence Screening for abuse and neglect
* Scale measures progressive need for assistance in daily activities (e.g., Treating medical comorbidity with intensity appropriate to the stage of the
choosing clothes, dressing); scores range from 1–2 (normal) through 6–7 (severe disease and patient/family wishes

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
dysfunction (219). Treating psychiatric comorbidity (depression, psychosis, agitation) with
y nonpharmacological and, if necessary, pharmacological interventions
This 22-item scale measures cognitive function; scores range from 30 (ex-
cellent function) to 0 (severe dysfunction) (220). Addressing end-of-life-issues
Note: Reprinted with permission of Dr. S. Gauthier and Can Med Assoc J. Counseling regarding research options
Family care goals
Increasing effectiveness of care and coping strategies
made only by histopathologic examination of brain tissue Increasing satisfaction with the family member’s preferred level of
after the patient’s death (15). Early and accurate diagnosis involvement—often, no matter how much an individual is doing for his
or her family member with AD, he or she harbors feelings that it is
affords the patient, family, and physician the time to plan
not enough
the best way to manage the disease. If diagnosis is made in Decreasing negative consequences on the family
the early stages of AD, the patient can assume a significant Minimizing family conflicts
role in planning financial, legal, and care issues as well as
housing arrangements for the future. At the time of di-
agnosis, physicians need to reinforce with caregivers that
the condition is a brain disorder, and they need to clear up referral to a geneticist or a genetic counselor for the whole
any misconceptions that the family may have. family and the patient is recommended.

Role of the Primary Care Physician Educating the Patient and Caregiver
The primary care physician will manage a growing num- During the workup and in disclosing the diagnosis to
ber of persons with AD and their family members or other the patient and the family, the clinician begins to educate
caregivers upon whom these patients depend. In addition to about AD. This discussion should include a frank, honest,
the diagnostic and pharmacological treatment components balanced view of what is known about the disease process
of managing AD, the physician will be called upon to assist and available treatment options. Education regarding the
with the behavioral, social, economic, legal, and living- recognition of current symptoms and the symptoms likely
environment problems facing the patient and her or his fam- to occur with progression of the disease will assist in
ily. A team approach to managing these complex problems formulating plans for safety and health maintenance. Addi-
of AD is a practical and effective management strategy. In tional sources of education should be provided. These may
addition to nurses and physician assistants, the team may include material written for the lay person, such as The
include those with expertise in neurology, geriatric psychi- 36-Hour Day (16) and Forget Me Not (17). The 3 ‘‘R’s’’—
atry, social work, clinical psychology, and elder law. The repeat, reassure, and redirect—can help caregivers reduce
neurologist assists in the differential diagnosis of patients escalating behaviors and limit the need for pharmacologic
with atypical dementia presentation and in the management management. Caregivers should be counseled to help pa-
of later stage neurologic features of AD, such as seizures. The tients with AD maintain social and intellectual activities as
geriatric psychiatrist assists in the differential diagnosis of tolerated, especially important family events.
complex cases and in the recognition and psychopharma- Referrals to local and national groups, such as the
cologic management of behavioral problems such as agita- Alzheimer’s Association, that are dedicated to the education
tion, psychosis, and depression. The social worker assists and support of patients with AD is strongly recommended.
in maintaining the integrity of the patient’s family unit and Information about agencies such as the Administration on
in identifying and mobilizing community care resources and Aging (202-619-1006), the Alzheimer’s Association (800-
may provide psychotherapy for patients and caregivers. The 272-3900, www.alz.org), the Alzheimer Research Forum
clinical psychologist assists in the diagnosis of early-stage or (www.alzforum.org), and the Alzheimer’s Disease Educa-
questionable dementia and provides expertise in behavioral tion and Referral Center (800-438-4380, www.alzheimers.
approaches to such problems as depression. The elder law org) should be provided. An additional resource may be the
attorney assists in addressing issues such as guardianship American Bar Association Commission on Legal Problems
and health-care financial planning. Experts from other disci- of the Elderly (202-662-8690). Other community services,
plines such as pharmacy, nutrition, physical therapy, and such as the local chapters of the Agency on Aging, Meals
occupational therapy can also make important contributions on Wheels, and organizations providing transportation, may
to management. For patients with early-onset familial AD, all be important as a means of reducing the burden of care-
MANAGEMENT OF ALZHEIMER’S DISEASE 333

giving. The physician is the critical conduit for providing of the illness. Family and caregiver interventions will help
this information to the caregiver. not only the caregivers but also the patients with AD by pre-
venting premature institutionalization (24,25). Educational
Genetic Testing program, training program, support, and respite services
Testing for the apolipoprotein E-4 (APOE-4) gene, one directed toward caregivers of dementia patients may improve
form of a gene on chromosome 19 that is more common their coping skills and reduce stress (26–29).
in individuals with AD than in age-matched individuals
without dementia, is not currently recommended for use in Reducing Excess Disability/Functional Impairment
diagnosis because it is found in many undemented elderly Many factors contribute to functional impairment in pa-
and is not found in many patients with AD (18,19). Em- tients with AD. They include social issues, cultural expec-
pirical data on the benefits and potential harm of genetic tations, environmental factors, sensory deficits (hearing

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
susceptibility testing with APOE-4 are currently being col- and vision), pain, coexisting disease states (e.g., over- or
lected and studied in the multicenter REVEAL study (Risk undercorrection of hypothyroidism, vitamin B12 deficiency,
Evaluation and Education for Alzheimer’s Disease) (20). etc.), fear of falling, and lack of motivation. Treatment of
Patients with early-onset familial AD should be referred to medical comorbidity can improve function (30) and can
Alzheimer Disease Research Centers for counseling regard- be important in delaying the progression of frailty (31).
ing the potential benefits of genetic testing. An impersonal For patients who need them, the value of new eyeglasses
relationship or an encounter created solely for the purpose or a hearing aid cannot be overemphasized. Such devices
of risk assessment would seem more vulnerable to mis- facilitate patients’ understanding of and participation in
communication than would an encounter discussing risk the choices surrounding care at the end of their lives. Ad-
assessment that is embedded in a long-term clinical re- dressing these issues can greatly improve AD patients’ func-
lationship (20). tional and even cognitive abilities. In addition, nursing
care and care by caregivers that creates dependency is a sig-
Caring for the Caregiver nificant factor in causing excess functional impairment.
Caring for the caregiver is an essential element of Teaching the caregivers (at home as well as in long-term
managing the patient with AD. The majority of patients with care facilities [LTCFs]) to implement a restorative philos-
AD are cared for by family members or other caregivers ophy of care is recommended. Restorative care focuses on
in their own homes. They frequently continue to devote the restoration and/or maintenance of physical function and
a substantial amount of time to caring for a patient after helps AD patients to compensate for functional impairment
admission to a nursing home. Caregivers are responsible so that the highest level of function is obtained.
for administering medication prescribed by the physician.
The burden of caregiving exacts a heavy toll: it is estimated COUNSELING REGARDING SPECIFIC ISSUES
that about half of all caregivers of people with dementia
suffer severe emotional distress, and dementia caregivers Should the Patients Be Told of Their Diagnosis?
have significantly higher rates of depression, physical illness, In sharing the diagnosis of dementia, one needs to
and other health-related problems (21,22). Almost 90% of consider each patient individually (32). Generally speaking,
caregivers interviewed in one study reported fatigue, anger, patients in their early stages need to know the nature and
and depression directly linked to caring for a demented prognosis of their disease once the diagnosis is clear to the
family member (23). Fatigue is probably the most under- treating physician. Physicians must not allow their own dis-
estimated reaction. Caregiving is also associated with a comfort—or the misguided requests of family members—to
greater use of sedative hypnotics and a higher mortality subvert the honesty with which they relate to patients. It is
rate among caregivers. Caregivers must be warned of the also important to ensure that the patient’s ability to com-
dangers of emotionally, physically, and financially deplet- prehend is at its highest possible level. Disclosure may also
ing themselves, and regular assessments must be made to assist in persuading the patient to accept help and in man-
screen for psychiatric disorders in the caregivers. Spouses aging social needs (33). It also enables the issue of driving
of patients with AD should be also screened for a dement- safety to be addressed. With the development of new drug
ing illness if, during the patient’s evaluation, the physician treatments, disclosure allows patients to consent to partic-
learns about cognitive impairment in the spouse. Caregivers ipation in clinical trials when they still have the capacity to
often do not recognize that their guilty feelings lead them consent. Currently most research relies on relatives to give
to make unrealistic demands. It also is not uncommon for proxy consent, although this has been challenged as legally
spouses or children to feel that they would be betraying their unacceptable (34). This said, there are circumstances in which
relatives by sending them to a nursing home. it may be ill-advised to tell a patient that he/she has AD
Many caregivers are reluctant to ask for help. It is im- during the first few visits. For example, if the patient has
portant that health care providers ask the caregivers if they no support system, there is a potential that such a disclosure
need assistance and validate their feelings. Clinicians must may undermine his will to live. In such situations, it may
be prepared to help the caregivers deal with anger, denial, be prudent to first assist the patient in establishing support
anxiety, guilt, grief, and clinical depression as they adjust to networks and relevant services and then sensitively dis-
the progression of the disease. The grief and adjustment close the diagnosis.
process for a caregiver is complex and cyclical, potentially In the late stages of disease the truth may neither benefit
reactivated by the additional impairments at each new stage nor harm the patient (33). Of course, there exists a greater
334 GROSSBERG AND DESAI

dilemma in the cases in which some patient understanding tronic guards to prevent wandering are recommended. Regis-
remains. With sensitivity, flexibility, and discretion, such bad tration with the Safe Return Program through the Alzheimer’s
news may still be delivered. Disclosure of diagnosis should Association should be encouraged. Ethical considerations
not be a one-office-visit event and must be seen as an on- should be kept in mind when using electronic tagging de-
going, dynamic process and a fundamental part of the care vices in patients with AD who have a tendency to wander
of a patient with dementia. (42). When used, particularly in mild AD patients, these
devices are often demoralizing and dehumanizing.
Discussion of Future Financial,
Health Care, and End-of-Life Issues Poisonous or harmful substances.—Families and care-
Physicians should help both the family and patient to givers need to be counseled to keep such substances out of
establish medical and legal advance directives for patients the reach of patients with AD.

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
with AD and should recommend updating the patient’s
will early in the course of treatment (35). Open discussions Other safety concerns.—Families and caregivers should
regarding role and responsibility changes within the family be counseled to modify the place of living in order to reduce
system should be encouraged. Assignment of health care objects and hurdles that might increase the risk of falls.
proxy, durable power of attorney, and discussion of end- Sharp objects should be kept out of the patient’s reach, and
of-life issues, including living wills, should be addressed appliances and power tools should be kept unplugged and
while the patient is competent to make informed decisions out of sight. Smoke alarms should be kept in working order.
regarding these concerns. It is important to introduce the A home visit for safety assessment and guidance by a social
topic of advance directives sooner rather than later. The un- worker may be helpful.
predictability of serious events such as hip fracture, pneu-
monia, etc., drive home the point that the discussion should Counseling Regarding Long-Term Care Placement
take place before the crisis occurs. Discussions about ad- As many as 90% of patients with AD reportedly become
vance planning do not have to be lengthy or conclusive. institutionalized before death (43). However, most patients
But the topic should be an agenda item in encounters with with AD continue to live in the community until family
patients with dementia, much like nutrition and safety are. caregivers are no longer able to care for them. Patient char-
acteristics (e.g., high cognitive impairment, one or more
Safety Issues dependencies in ADL, difficult behaviors) and caregiver
Clinicians should counsel the caregivers regarding vari- characteristics (e.g., high caregiver burden) are both impor-
ous safety issues in the home. Caregivers need to be told when tant determinants of long-term care placement for patients
the patient will start needing 24-hour care and supervision. with dementia (44). Planned admissions to a LTCF may
be better than unplanned admissions (45). Numerous local,
Driving.—Many elderly patients with AD may continue state, and federal agencies monitor nursing homes, but vigi-
to drive despite having impaired abilities (36–38). Physi- lant, assertive family members may be most helpful in as-
cians often fail to identify signs of impaired mental perfor- suring quality care. High-quality LTCFs encourage family
mance in their older patients, further compounding this issue. participation. Before choosing a LTCF, consumers should
A history of getting lost, misjudging distances, inappropri- examine reports of the deficiencies found in state inspec-
ate speed, missing signs or signals, moving violations, motor tions. These can be accessed at the website www.medicare.
vehicular accidents or near misses, passenger panic, etc., gov. Family members should also contact their state Depart-
should be documented (39,40). A substantial proportion of ment of Health with concerns about general conditions or
even mildly demented people are not safe drivers when di- care in nursing homes; the ombudsman in their local aging
rectly observed. A substantial proportion of mildly demented department about problems with finances, property, or other
people can pass performance tests. For patients with very consumer concerns; and local police or their aging depart-
mild AD, formal, serial performance evaluations are recom- ment’s office of protective services with any concerns about
mended. Patients with more advanced AD should be recom- abuse.
mended to discontinue driving. All patients with AD should
be systematically evaluated regarding their driving abilities.
OTHER IMPORTANT ISSUES
Presence of firearms.—Many elderly in the United States Abuse and Neglect
have firearms in their households, most of which are stored Patients with AD are certainly at high risk of abuse and
loaded (41). Each patient and his or her family/caregiver neglect (46). Estimates of the prevalence of abuse of older
must be specifically asked about access to firearms. The adults suffering from dementia range from 5.4% to 11.9%
physician must advocate for the safe storage or removal of (46). They far exceed the 1–4% prevalence rates typically
these firearms. Any firearms in the patient’s house should be cited for all older adults, cognitively intact as well as de-
disabled in order to prevent accidental gunshot wounds or mented (47). The physician must acknowledge that not only
deaths. will some family members have conflicts with AD patients,
but a small number will actually neglect or abuse their AD
Wandering.—Families should be warned of the hazards of relatives. This also applies to nonfamily members or profes-
wandering. Supervised walks and using door locks or elec- sional caregivers. Physicians must be able to identify the
MANAGEMENT OF ALZHEIMER’S DISEASE 335

potential for emotional, physical, sexual, and financial abuse In the presence of dementia, successful cardiopulmonary
or neglect, detect its occurrence, and deal with situations in resuscitation (CPR) is 3 times less likely to succeed, as
which abuse is present. Financial abuse by telemarketers compared to CPR in patients with metastatic cancer (56). In
and other people (including those on television) who make addition, in the demented population, most cardiac arrests
patients with AD easy victims of solicitation should also occur in long-term care institutions, not acute-care hospitals.
be suspected and reported to the state’s attorney general The success rate of CPR is only 1% in demented LTCF
office. A prior history of abuse is associated with a greater residents. The benefits of successful CPR are further dimin-
likelihood of abusiveness once dementia occurs. Malnutri- ished because of injuries, such as broken ribs, associated
tion, noncompliance with medications, bruises, decubitii, with CPR and because of the need for mechanical venti-
poor personal hygiene, and other evidence of trauma are lation. Families should be counseled about these outcomes
some of the signs that should alert physicians that abuse and whether the patient and family would like ‘‘do not

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
or neglect may be an issue. Physicians need to learn their resuscitate’’ directives instituted. These discussions should
state’s law on reporting of suspected elder abuse. Interven- ideally occur when the patient is in the early stages of AD
tions such as supportive counseling, individual or family and thus can be an active participant.
psychotherapy for caregivers, respite or in-home care ser- Antibiotic therapy may be withheld in some frail AD
vices for patients, and alternative living situations for all patients who are expected to die soon (such as patients in
parties concerned may be helpful. their severe to terminal stages of AD). Most severely de-
mented AD patients are indeed frail and at high risk of dying
End-of-Life Care from pneumonia or sepsis, in spite of antibiotic treatment
Dementia severity and other patient characteristics are (54). Palliative management of symptoms such as pain, dys-
important for informed end-of-life decision making and pnea, etc., in patients with severe to end-stage AD should be
for assessing effectiveness of interventions in severely de- similar to that used in patients who are dying from cancer.
mented patients in order to prevent an unfavorable outcome An agreed-upon, documented goal and treatment approach
of medical comorbidity, such as pneumonia or hip fracture for all health-care team members is an essential component
(48). Patients with advanced dementia commonly develop of end-of-life care for AD patients. Management should
difficulty eating, often when they become bedridden and involve vigilance to ensure good symptom management,
dependent in all activities of daily living (49–51). They may support to families of patients with AD in their end-stages of
resist or be indifferent to food or fail to manage the food dementia, communication with other health team members,
bolus properly once it is in the mouth (oral phase dys- and objective documentation. Palliative care is an appro-
phagia). In moderately demented patients, prevention of priate treatment strategy for the management of patients with
aspiration and weight loss, if possible, may improve mid- advanced AD. Palliative care should never mean that the
term outcome (52). Enteral tube feeding is intended to patient with AD receives less care. Rather, palliative care
prevent aspiration pneumonia, to forestall malnutrition and replaces aggressive intervention, with care oriented toward
its sequelae, including death by starvation, and to provide comfort. Hospice care may be appropriate for patients with
comfort. There is no evidence to indicate that tube feeding AD in the terminal stages of the disease (57).
improves any of these clinically important outcomes, and
some data seem to indicate that it does not (53,54). Indeed,
tube feeding may increase the risk of aspiration. In addition, PHARMACOTHERAPY oF AD
the risks of this treatment are substantial. Nasogastric tubes
may promote sinus and middle ear infections, and gastro- ChEI Therapy
stomy tubes may cause cellulitis, abscesses, necrotizing This class of drugs is presently regarded as the standard
fasciitis, and myositis. Many patients are put in physical treatment of AD. Four ChEIs have been approved by the
restraints to prevent tube removal. For severely demented U.S. Food and Drug Administration (FDA) for the treatment
patients, the practice of tube feeding should be discouraged of mild to moderate AD. They are tacrine, donepezil, rivas-
on clinical grounds, but this decision needs to be made in tigmine, and galantamine. These compounds increase the
the context of the patients’ and families’ moral and religious concentration of acetylcholine and the duration of its action
belief systems. There are no published studies that compare in synapses by inhibiting the degradation of acetylcholine.
tube feeding to oral feeding. These compounds provide symptomatic treatments and may
It is possible to convert tube feeding to hand feeding and, also have disease-modifying effects. They have been shown
in some cases, patients may be able to feed themselves again in several large, multicenter, randomized, double-blind,
(55). Withdrawing sedatives and psychotropic medications placebo-controlled trials (of 3–6 months’ duration) to im-
should be considered in AD patients with swallowing prob- prove cognitive function, global outcome, and activities of
lems, because these medications may reduce consciousness daily living (58–61). There is also accumulating evidence
and thus predispose the patient to aspiration pneumonia. that ChEIs may improve behavioral and psychological symp-
Improving oral care is recommended to reduce oropharyn- toms of AD, such as psychosis and apathy (62). Treat-
geal colonization by pathogens. Inadequate staffing and lack ment with ChEIs may also help caregivers by reducing the
of supervision at mealtime may contribute to weight loss in burden of caregiving (63). The mean effect of drug over
nursing homes. If fed quickly, AD patients may cough or placebo represents an improvement in cognition roughly
choke or aspirate. Adequate staffing and allowing patients equivalent to stemming 6–12 months of natural decline in
with AD to take their time to eat is thus very important. untreated patients. There is preliminary evidence from a retro-
336 GROSSBERG AND DESAI

spective return drop-out data indicating that the effects of The recommended starting dose is 5 mg daily, which is
rivastigmine on cognition may be sustained in rivastig- increased to 10 mg daily after 4–6 weeks. Morning dosing
mine-treated patients and indicating that rivastigmine may of donepezil may be preferable in some patients who ex-
affect disease progression (64). Evidence with the ChEIs perience nightmares or insomnia. It may be taken without
also suggests that the effect is extremely variable, with large regard to meals unless gastrointestinal side effects occur, in
improvements in some patients and none in others. At the which case it should be taken with meals. Although there is
moment we have no reliable way of distinguishing potential 1 case report of fulminant hepatitis with the concomitant use
responders from nonresponders. When response occurs, it of donepezil and sertraline (70), laboratory monitoring of
does so relatively quickly (12 weeks). The only certain way liver enzymes is not required.
of proceeding is therefore to use a ChEI for at least 12 weeks
at the proper dose and to observe the results systematically. Rivastigmine.—This was the third ChEI approved by the

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
The physician should seek evidence of changes in all 3 FDA for symptomatic treatment of mild to moderate AD in
domains (cognition, ability to perform ADL, and behavior) the United States. Rivastigmine should be titrated every 4
to determine if the patient has benefited from the ChEI. weeks, as opposed to every 2 weeks, as recommended when
Group means hide a marked heterogeneity of response, as the drug was first made available. Slower titration and
40–50% of patients show a definite clinical improvement taking rivastigmine with a full meal significantly improves
(4 points on the AD assessment scale–cognitive subscale tolerability, especially with regard to gastrointestinal side
[ADAS-cog], equivalent to stemming half a year or more effects. One unique feature of rivastigmine that distingui-
of natural cognitive decline), whereas 20% show a stronger shes it from other ChEIs is the very low risk of drug inter-
response (7 points on the ADAS-cog, equivalent to stem- actions in AD patients receiving multiple medications for
ming a year or more of natural cognitive decline). Respon- ‘‘real-world’’ comorbidities (71). This is because the meta-
ders are maintained close to baseline for 12–18 months on bolism of rivastigmine occurs primarily via enzymatic
both cognitive and noncognitive measures. No reliable pre- cleavage (hydrolysis) by cholinesterases at the site of action
dictors of response have emerged, and in each patient, and does not require the cytochrome P450 enzyme system.
careful assessment of benefit needs to be undertaken after The starting dose is 1.5 mg twice a day with meals (break-
2–4 months of treatment. fast and supper), and this dose is increased by 3 mg/day, not
faster than every 4 weeks (as tolerated), to a therapeutic dose
Tacrine.—Tacrine was the first ChEI to be approved of 6–12 mg/day. The highest tolerated dose is recommen-
specifically for the symptomatic treatment of patients with ded, as there is some evidence that higher doses may pro-
mild to moderate AD. The starting dose for tacrine is 10 mg vide greater benefits. A more rapid progression of AD while
4 times daily, and this dose is increased by 40 mg/day no receiving placebo treatment was predictive of a significantly
more frequently than every 4 weeks (according to tole- stronger patient response to rivastigmine therapy on various
rance), to a maximum daily dose of 160 mg (40 mg 4 times measures (72). Laboratory monitoring is not required.
daily). Tacrine has been associated with hepatotoxicity (65)
and thus requires baseline and multiple follow-up liver Galantamine.—This was the fourth ChEI approved by the
enzyme determinations. This along with the need for mul- FDA for symptomatic treatment of mild to moderate AD in
tiple daily dosing makes it unsuitable as a ChEI of first the United States. Metabolism is hepatic via glucuronidation
choice. We recommend that its use be restricted to patients and the cytochrome P450 isoenzymes 2D6 and 3A4; inter-
who do not tolerate or respond to ChEIs. Tacrine is exten- actions with other drugs that are metabolized through this
sively metabolized by the liver via the cytochrome P450 pathway are therefore possible. Caution should be used in
1A2 isoenzyme system; therefore, it has the potential to patients with liver disease. The starting dose is 4 mg twice
interact with other medications metabolized by this isoen- a day, and this dose is increased every 4 weeks. The thera-
zyme, such as theophylline, fluvoxamine, and cimetidine. peutic dose is 16–24 mg/day. A 6-month study showed no
Tacrine should be avoided in patients with liver disease. additional benefit and a higher rate of side effects with a dose
of 32 mg/day (73). Laboratory monitoring is not required.

Donepezil.—Donepezil was the second ChEI approved by Role of Butyrylcholinesterase and Nicotinic Modulation
the FDA for symptomatic treatment of mild to moderate AD Humans have 2 types of cholinesterase: acetyl and buty-
in the United States. Two placebo-controlled clinical trials ryl. The physiological role of butyrylcholinesterase is being
of donepezil have been reported in which efficacy was investigated, but levels of this enzyme have been shown to
demonstrated for 1 year in mild-to-moderate AD based on increase as AD progresses, whereas levels of acetylcholin-
cognitive (66) and functional (67) measures. Donepezil is esterase decrease (74). Both enzymes are found in neuritic
largely metabolized by the liver, although some of the dose plaques, and their inhibition with ChEIs may modify the
is recovered in the urine as unchanged drug (11–17%) (68). deposition of beta-amyloid, a key component of the patho-
It is metabolized by the cytochrome P450 isoenzymes 2D6 physiology of AD as we currently understand it. The clinical
and 3A4. Clinically relevant drug interactions with other significance of this action, if any, in terms of slowing pro-
drugs have not been studied. Interaction with paroxetine gression of the disease or better symptomatic efficacy in
(patient developing increased confusion and agitation) has later stages has yet to be fully established. Of the currently
been reported (69). Caution should be exercised in using available ChEIs, only tacrine and rivastigmine have the
donepezil in patients with severe hepatic or renal disease. ability to inhibit butyrylcholinesterase. Of the currently avail-
MANAGEMENT OF ALZHEIMER’S DISEASE 337

Table 3. Characteristics of Commonly Used Cholinesterase Inhibitors (ChEIs)


Donepezil Rivastigmine Galantamine Clinical Relevance
1. Elimination half-life (hr) 70–80 0.6–2* 7–8 Easier to switch from drug with shorter half-life
2. Metabolism Liver By AchE Liver Liver metabolism involves P450 system: potential for drug
interaction
3. Protein binding 96% 40% 8% Potential for interaction with drugs having high protein
binding
4. Food interaction No Yes No Need to take the drug with meals if food interaction
5. Dosing Once daily Twice daily Twice daily Compliance issues
6. Pricing 2 Level Flat rate 3 Level Cost implications
7. BuChE inhibition No Yes No Unknown

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
8. Nicotinic modulation No No Yes Unknown
9. Specific AchE subtype inhibition No Yes No Unknown
10. Titration 1 Step 2–4 Steps 2–3 Steps Complex titration may influence decisions in busy
practices
* Enzyme inhibition significantly outlasts elimination half-life.
Note: BuChE ¼ butyrylcholinesterase; AchE ¼ acetylcholinesterase.
Reprinted (with modification) by permission of Dr. R. Bullock and Br J Psychiatr. 2002;180:135–139.

able ChEIs, only galantamine has the property of allosteric Importance of Starting ChEI Early
modulation of the presynaptic nicotinic receptors. The Patients with AD who begin treatment with a ChEI later
potential significance of this effect is additional increase in do not do as well as those who began treatment early
cholinergic neurotransmission. Whether this provides any (73,80,81). In a 1-year study, in which the double-blind,
additional clinical benefit over other ChEI has not been placebo-controlled phase lasted for 26 weeks, all subjects—
proved at the present time. whether started on rivastigmine or placebo—were switched
to open-label rivastigmine and followed for an additional 26
Which ChEI Should Be the First Choice? weeks (81). Patients who were initially placed on placebo
Only direct comparisons between various currently avai- and later started on rivastigmine seemed to respond to the
lable ChEIs will provide definitive data that can be used ChEI and had a significant increase in cognitive function,
to maximize patient outcome. In general, these agents all but then they started to decline. They never caught up to
have similar degrees of efficacy. However, there are some the group that started on rivastigmine initially. Similar find-
important differences (refer to Table 3). Certain patients may ings were seen with galantamine and donepezil (73,80). An
benefit more from one particular agent over the others. For open-label extension study found that the group that was
AD patients living alone who do not have close daily super- started on donepezil (10 mg/day) and continued on this
vision over their medications, donepezil may be preferable dosage functioned better for 18 to possibly 24 months
because of its once-a-day dosing. For patients with AD who longer than persons who were initially on placebo and then
also have hepatic disease or who are on numerous other started on donepezil (10 mg) (80). These findings indicate
medications metabolized by cytochrome P450 2D6 and 3A4 the importance of starting ChEI as early as possible in AD.
enzymes, rivastigmine may be preferable because of its lack Early institution of ChEI may also delay the emergence of
of cytochrome P450 metabolism. Patients with AD who ex- neuropsychiatric symptoms in patients with mild to mod-
perience impaired sleep or excessive dreaming with done- erate AD, as shown in a 5-month placebo-controlled study
pezil may benefit more from rivastigmine or galantamine. with galantamine (82).

Switching From One ChEI to Another More Advanced AD and ChEI Therapy
There is some preliminary evidence that if a patient There is preliminary evidence that the beneficial effects of
does not respond to one ChEI, switching to another may ChEIs may extend to more advanced (moderate to severe
be beneficial (75,76). Switches can also be performed to AD) stages and that effects may even be more robust than
cope with side effects (75,77). In general it is not difficult seen in mild to moderate AD (83). In this 24-week, random-
to switch from one drug to another among the 3 ChEIs ized, double-blind placebo-controlled study, the donepezil
(donepezil, rivastigmine, and galantamine). Combination of group showed significant improvement in the neuropsychia-
ChEI is not recommended. Standard dose escalation using tric inventory (NPI) scores, whereas the placebo group
monthly titration is recommended. Preliminary data indicate worsened slightly. The donepezil group was better than the
that for patients experiencing no safety/tolerability problems placebo group for all individual items in the NPI, with sta-
on a ChEI, another ChEI can be administered the follow- tistically significant differences seen for depression, anxiety,
ing day with no washout period (78,79). If the patient is and apathy (83).
experiencing safety/tolerability problems with a ChEI, a
washout period of 7 days, or until symptoms resolve, is rec- Treatment of AD With ChEIs
ommended before switching to another ChEI (78,79). As in the Nursing Home Setting
yet, no guidelines for switching from one ChEI to another Very little controlled data is available for the potential
have been published. benefits of ChEIs in patients with AD in the LTCF setting.
338 GROSSBERG AND DESAI

Donepezil was found to be beneficial in the domain of cog- that patients should continue on these medications until
nition and overall dementia severity compared to placebo in they no longer have meaningful interactions with other indi-
a 6-month treatment period of AD patients in the nursing viduals, because ChEIs may still help alleviate behavior
home setting (84). Unfortunately, no significant differences problems even when cognition is severely impaired. Several
were obtained in the noncognitive domains of behavioral factors influence medication prescribing for most older pa-
disturbances and ADL in this study. An open-label, 52- tients with AD, resulting in considerable variability and the
week study of rivastigmine in nursing home patients with need to individualize treatments. Elderly patients often take
AD demonstrated improvement in many of the behavioral multiple medications, so the clinician must be aware of
and psychiatric symptoms, such as irritability, anxiety, de- potential drug interactions. Patients with symptomatic bra-
lusions, hallucinations, disinhibition, aberrant motor activ- dyarrhythmia, active peptic ulcer disease, and acute exacer-
ity, nighttime behavior, and appetite (85). In addition, about bation of chronic obstructive pulmonary disease and asthma

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
40% of patients who were on neuroleptic medications for may experience worsening of their problems because of the
these symptoms were able to reduce or discontinue these mild peripheral cholinergic effects of ChEIs. Generally,
medications over the course of the study. Patients with AD ChEIs can be safely instituted once these conditions are
in the nursing home are generally older, exhibit greater se- stabilized. Adverse effects are similar for all ChEIs and
verity of dementia, and have more comorbid illness than typically are gastrointestinal (nausea, vomiting, diarrhea,
do AD patients in the community. anorexia, and weight loss) in nature. They are usually seen
during dose escalation. Other adverse effects such as abdom-
Use of ChEIs in Non-Alzheimer’s Dementias inal pain, dizziness, syncope, and headache have been also
ChEIs are approved for use in mild to moderate AD only, described but are less common. Adverse effects are usually
and use of these agents to treat other dementing disorders is mild to moderate in severity and resolve spontaneously or
an off-label use. There is initial evidence from randomized, after dosage reduction. The frequency of adverse effects
double-blind, placebo-controlled studies that ChEIs may dramatically increases when the dose of a ChEI is increased
also benefit cognition, daily functioning and behavioral too rapidly (in 1–2 weeks). Their frequency is very low dur-
symptoms in vascular dementia (VaD) and Lewy body ing the maintenance phase. Potential for prolonging the
disease/dementia (LBD) (86–88). Both of these dementing effects of the muscle relaxant succinylcholine, used during
disorders have been found to have an associated cholinergic anesthesia, exists with all ChEIs but does not seem to be
deficit. Preliminary data indicates that early and widespread clinically significant, although the anesthetist should be
cholinergic losses may help differentiate LBD from AD informed of the possibility. We do not recommend discon-
(89), suggesting that cholinergic replacement therapy may tinuing these drugs a few days prior to surgery because of
be even more effective in LBD than in AD, especially in the risk of cognitive deterioration. If for any reason a ChEI
mild-stage disease. We recommend a trial of ChEIs for is discontinued for a significant period and need to be re-
patients with VaD and LBD. Other disorders associated with instituted, it should be done by again starting at the lowest
cholinergic deficit include but are not limited to Parkinson’s dose and gradually titrating upward to the highest tolerated
disease with dementia and Down’s syndrome with pro- therapeutic dose. These drugs have not been well studied
gressive cognitive decline. These disorders may also benefit in patients with severe hepatic or renal impairment. Hence,
from a trial with ChEI, although large controlled studies are caution should be exercised when using a ChEI in this pa-
lacking. The only double-blind, placebo-controlled 24-week tient population.
pilot study in patients with Down’s syndrome Dose titration should be as slow as necessary to prevent
and AD showed that improvement in the donepezil group the development of gastrointestinal or other side effects. The
was not significantly better than improvement with placebo. risk of nausea can be reduced by administering the medi-
Surprisingly, noncognitive symptoms showed less improve- cation on a full stomach, and intermittent antiemetics can
ment than they did in the placebo group (90). The biological be used if necessary. Patients should be titrated up to the
basis of these findings is not yet certain. No therapeutic highest therapeutic dose they can tolerate (for donepezil it is
benefit is anticipated in dementia syndromes without a 10 mg/day, for rivastigmine it is 12 mg/day, and for galan-
cholinergic deficit, such as frontotemporal dementia or tamine it is 24 mg/day). Once therapy with ChEI is begun
Huntington’s dementia. Nonprogressive dementias, such as and the therapeutic dosage reached, patients should be eval-
those secondary to traumatic brain injury or anoxic enceph- uated every 3 months to monitor response to treatment in 3
alopathy, may also not respond to ChEIs. domains, cognitive, functional, and behavioral. If unaccept-
able side effects occur at higher doses, the dose should be
Patient Selection, Management of Adverse Effects, decreased as long as it is in the therapeutic range. If side
and Therapeutic Outcome effects develop with dose escalation, we recommend going
All pivotal studies have investigated patients with mild to back to the tolerated dose and then increasing the interval
moderate AD. How early in the course of AD ChEIs should of dosage escalation. ChEI should be discontinued if side
be initiated and for how long they are of use have not been effects persist at the lowest therapeutic dose, if the patient
elucidated fully. Although most studies have been done shows accelerated decline after a 6-month trial, or if a
in patients with mild to moderate AD (Mini-Mental State medical condition develops that significantly increases the
Exam [MMSE] scores between 10 and 26), clinicians are risk benefit ratio. The lowest therapeutic dose is 5 mg/day
recommended to consider ChEI even in patients with AD for donepezil, 6 mg/day for rivastigmine, and 16 mg/day for
and scores of more than 26 or less than 10. We recommend galantamine. If patients with AD are not able to tolerate
MANAGEMENT OF ALZHEIMER’S DISEASE 339

more than donepezil 5 mg/day, rivastigmine 3 mg/day, or Table 4. Clinical Expectations From Cholinesterase Inhibitor
galantamine 8 mg/day, another agent should be considered. Therapy
Patients who have been on one ChEI and failed to benefit Primary benefits
(due to intolerable side effects or accelerated decline) should Maintain current level of daily functioning or slow the decline in current
be considered for a trial with another ChEI. Incontinence level of functioning
and increased behavioral disturbances (irritability) have been Maintain current level of cognition or slow the cognitive decline associated
described with donepezil use and may be seen with other with Alzheimer’s disease (AD)
ChEIs as well. Decrease emergence of behavioral and psychological disturbances
associated with AD
The course of AD tends to be slowly progressive, with
a loss of 3–5 points per year on a standard assessment in- Secondary benefits
strument such as the MMSE. Caregivers and patients must Decrease caregiver burden and distress

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
have realistic expectations (Table 4); these drugs’ effects are Decrease overall health-care cost
Delay institutionalization
modest, and sometimes no symptomatic improvement is
noted. In fact, a report of Ôno change’ means that these drugs Caregiver and physician expectations
are helping, because without ChEI treatment, one would ‘‘No change’’ means cholinesterase therapy is helping
have seen a decline in functioning.
If a ChEI is stopped, rapid deterioration can occur in
some patients. Also, reintroducing the drug after the AD pa- provement in noncognitive behavioral or ADL functions
tient has been taken completely off a ChEI may not yield the with Ginkgo biloba extract, since this is critical in eval-
same benefit. By tapering the dosage slowly, we can detect uating the use of treatment in AD. Also, there is no data
early those patients who are going to lose function or show regarding the safety of the use of Ginkgo biloba extract
cognitive deterioration. If they do, then there exists a good along with ChEIs.
reason not to take them off the drug. Only very early in the
disease will the patient be able to self-report improvement Antioxidants (vitamin E and selegiline).—In a double-
or benefit. Many of the patients, by the time they are diag- blind, randomized, placebo-controlled study (94), fewer
nosed, have already lost insight and are unable to appreciate participants (58%) in group taking vitamin E (2000 IU/
the benefits of ChEIs. day) and selegiline (5 mg bid) reached 1 of the 4 endpoints
(death, institutionalization, loss of 2 out of 3 basic ADL,
Other Agents for Treatment of AD or severe dementia), compared to 74% with a placebo.
However, more participants taking vitamin E suffered a fall
Metrifonate.—Metrifonate is another ChEI that has been compared to patients receiving a placebo. It was not possible
investigated for the treatment of mild to moderate AD. to interpret the reported results for specific endpoints (95).
Metrifonate was found to be beneficial in areas of cognition, There was no difference between the vitamin E group and
global functioning, and ADLs, compared to placebo in the selegiline group. There appeared to be no additive bene-
patients with mild to moderate AD, in a meta-analysis of fit to treatment with both agents. The principal concerns
4 randomized, double-blind, placebo-controlled trials (91). with high-dose (2000 IU) vitamin E are gastrointestinal
Unfortunately, development of this agent was halted re- upset and prolonged clotting time, with easy bruisability or
cently secondary to problems with muscle weakness and bleeding. A lower dose (400 iu bid) of vitamin E is recom-
respiratory paralysis. mended by many experts for patients with AD and may be
associated with lowered risk of adverse effects without
Memantine.—Memantine, a noncompetitive, highly volt- compromising the beneficial effects. In patients with AD,
age-dependent NMDA antagonist, has been approved for selegiline leads to small short-term improvement in cogni-
use in the treatment of dementia in Germany for over 10 tion and activities of daily living. Selegiline does not improve
years and recently was approved for use in the treatment of emotional state or global response (96). For patients with
AD in the European Union. It has been found to be useful AD who can tolerate vitamin E, there is no reason to take
in more advanced (moderate to severe) cases of AD (92). selegiline.
Patients with AD in the United States may import the drug
or consider participating in double-blind, placebo-controlled
studies currently underway at numerous sites all over the Estrogen.—Clinical trials indicate that oral conjugated
country. The FDA is currently reviewing the data for ap- equine estrogen is not an effective treatment for AD in
proval of its use in the United States. postmenopausal women (97,98). Hence, estrogen is not rec-
ommended for the treatment of cognitive or functional
Ginkgo biloba extract.—Oken and colleagues reviewed deficits attributable to AD. Preliminary data indicate that
the published literature on efficacy of Ginkgo biloba for AD short-term estrogen therapy may safely decrease the fre-
(93). They identified 4 well-designed, randomized, placebo- quency and severity of behavioral disturbances of dementia
controlled studies that met their inclusion criteria. They in elderly patients (99,100). Larger randomized, controlled
concluded that treatment with Ginkgo biloba extract (120 to studies are needed to further explore the potential benefits of
240 mg/day for 3–6 months) had a small but significant estrogen in the treatment of behavioral disturbances (aggres-
effect on objective measures of cognitive function in AD. sion, sexual disinhibition) in patients with AD. There is in-
We need further research to determine whether there is im- creasing evidence that estrogen may decrease the risk for or
340 GROSSBERG AND DESAI

Table 5. Common Behavioral Disturbances in Dementia


Aggression Affect mood
Verbal Anxiety
Screaming Depressive symptoms
Cursing Apathy
Irritability
Physical
Anger outbursts
Hitting
Biting Thought and perception
Kicking Delusions
Scratching Hallucinations
Grubbing Illusions

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
Misperceptions
Nonaggressive behavioral
Verbal Vegetative symptoms
Repetitive questioning Sleep disturbances
Complaining Insomnia
Increased daytime napping
Physical
Sundowning
Wandering
Pacing Sexual
Hoarding Hyposexuality
Rummaging Hypersexuality
Hiding Sexual disinhibition
Taking other people’s belongings Appetite
Voiding in inappropriates places Poor food intake
Shadowing Hyperphagia
Resistance to care
Intrusiveness
Fatigability
Mannerisms

delay the onset of AD in postmenopausal women (101,102).


However, this has not been universal (103,104), and a num-
ber of methodological shortcomings in these studies have
been identified (105). These potential benefits have to be
weighed against the known risks of estrogen, such as in-
creased risk of thromboembolism and gynecological cancers.

Figure 1. An algorithm for the management of behavioral disturbances in


TREATMENT oF BEHAVIORAL AND PSYCHOLOGICAL patients with Alzheimer’s disease (AD).
SYMPTOMS oF AD
The most challenging aspect of AD care is management
of behavioral and psychological signs and symptoms of anticonvulsants. For a comprehensive review of manage-
dementia (BPSD) (106,107). BPSD are common, serious ment of BPSD in patients with AD, the reader is referred
problems that affect the quality of life of both patient and to the review article on recognition and management of
caregiver and that frequently result in premature institu- behavioral disturbances in dementia by the authors (107).
tionalization. The majority of BPSD in AD are of clinical Figure 1 provides an algorithm for the management of
significance based on their severity and because of co-occur- behavioral disturbances in patients with dementia. Before
rence of multiple symptoms (108). BPSD are associated with any behavioral symptoms are ascribed to AD, the possibility
more rapid rates of cognitive and functional decline. BPSD that the symptom is produced by environmental influences
include agitation, aggression, delusions, hallucinations, de- or by intercurrent medical problems needs to be eliminated.
pression, apathy, sleep disturbances, and sexually inappro- Environmental influences that may affect behavior include
priate behaviors (Table 5). These difficulties occur in 90% temperature extremes, excessive noise, and physical re-
of patients at some point in the course of the disease and straints. In addition, many medical conditions, including
have been described in all dementing illnesses. Many clinical cardiovascular disease, chronic obstructive pulmonary
practice guidelines (109,110) have addressed the treatment disease, infection, anemia, and metabolic disorders, may
of these disturbances, noting that even modest improvement produce behavioral symptoms. One of the more common
in these behaviors can markedly improve quality of life for conditions that may elicit behavioral disturbances in patients
both patient and caregiver. Practice guidelines frequently with AD is unrecognized pain. Because AD individuals fre-
recommend starting with behavioral and environmental quently are unable to express pain verbally, any patient exhib-
approaches, followed by a wide variety of pharmacologic iting new or increased behavioral symptomatology should
interventions, including antipsychotics, antidepressants, and be carefully evaluated for the presence of pain.
MANAGEMENT OF ALZHEIMER’S DISEASE 341

After environmental or physical reasons for behavioral Table 6. Nonpharmacological Interventions for the Treatment
symptoms are eliminated, it is essential to determine which of Alzheimer’s Disease (AD)
psychiatric syndrome of dementia is producing agitation. Interventions with the patient
Treatment of depression, psychotic symptoms, and anxiety Specific nonpharmacological interventions
symptoms is more effective than treating several individual Behavior-oriented approaches
peripheral symptoms such as insomnia, aggression, weight Behavioral treatment (increasing pleasant events): especially useful for
loss, etc. Psychotropic medications play a critical role in the treatment of depression in moderate stages of AD
management of behavioral disturbances of patients with AD. Describe the problem behavior, assess specific antecedents and
consequences and implement specific strategies suggested by
Table 8 lists the principles of using pharmacotherapy to
the first 2 steps
manage behavioral disturbances in patients with demen-
tia. Although improved quality of life for AD patients and Emotion-oriented approaches

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
caregivers can be achieved in many cases with appropriate Supportive psychotherapy: especially in early stages of AD
Reminiscence therapy: in mild to moderate stages of AD
pharmacotherapy, there is currently no FDA-approved medi-
Validation therapy: in mild to moderate stages of AD
cation for the treatment of behavioral symptoms of AD. For Simulated presence therapy: in moderate to severe stages, especially for
the treatment of mild behavioral problems, ChEIs are an patients in long-term care facilities
effective treatment option, although in some patients they
Cognition-oriented approaches
may exacerbate BPSD (111).
Cognitive therapy: especially for treating depression in early stages of
AD
NONPHARMACOLOGICAL INTERVENTIONS
Stimulation-oriented approaches
Nonpharmacological interventions are the key to man-
Recreational therapies (crafts, games, pets)
agement of BPSD. The majority of AD patients are calmer
Art therapies (music, dance, art)
and better adjusted when treated with low-tech, nondrug
approaches that help to decrease problem behaviors and Nonspecific nonpharmacological interventions
promote independence (Table 6). The foundation of non- Reassurance
Distraction
pharmacologic management is recognizing that the person
Bright-light therapy
with dementia is no longer able to adapt and that instead Touch therapy: hand massages, back rubs
the environment must be adapted to the patient’s specific One-to-one therapy
needs. Caregivers should be counseled to learn to change Gardening
what can be changed through information gathering and di- Aromatherapy
rect action. They must also train themselves to cope with Light exercise
their reactions to what cannot be changed (and ‘‘intrapsy- Interventions with the family member/caregivers
chic’’ tricks or reframing perspectives—‘‘tomorrow will be Interventions to benefit the patient
better,’’ ‘‘my husband is difficult but he could be worse’’). Problem-solving therapy for the caregivers: especially for treating
Positive outcomes derived from support groups include depression in moderate stages of AD
not only increased knowledge of the illness and services Skills training for the caregivers: to improve personal care skills
Structured educational programs to decrease use of inappropriate
available as well as decreased feelings of isolation but also medications and psychotherapeutic agents for behavioral and
creative and practical suggestions for dealing with BPSD psychological symptoms of AD (BPSD)
using nonpharmacological interventions. Adult day services
Interventions to benefit the family member/caregivers
are often an effective method for managing demented pa-
Family therapy: especially to address conflicts, abuse
tients and postponing the need for institutionalization. Support groups: to decrease loneliness, increase knowledge of illness
Caring for loved ones with AD is psychologically and physi- and services available, and learn creative and practical
cally challenging but also provides the family with oppor- nonpharmacological interventions
tunities for personal growth and deepening of relationships Psychoeducational groups
with the patient and other family members. Behavioral Self-help groups
disturbances are often provoked through interaction with Respite care
Cognitive behavioral approaches: especially for treatment of
caregivers. The manner in which caregivers approach the
depression in the caregiver
patient with AD is critical, because most episodes of aggres-
sive behavior occur during contact with caregivers. Effective
strategies include leaving the patient and returning later or
about themselves, to be respected, to have the approval of
having one caregiver distract the patient while another is
others who are important to them, to be stimulated in body,
providing care. As ADL dependence on others increases,
mind, and spirit, to feel secure, to be included (not alienated
patients are at risk of invalidism, social isolation, lower self-
and marginalized), and to be needed. Every effort must be
esteem, depression, and a loss of control over their destiny,
made by the health-care providers to assist the caregivers in
which may in turn generate a sense of helplessness and hope-
meeting these needs of patients with AD. Some more-specific
lessness. Inactivity also increases the risk of pressure sores,
nonpharmacological interventions are as follows:
falls, loss of range of motion, and muscle wasting. Structured
and unstructured activities improve subjective well-being, 1. People with very early or mild AD who have been told
help maintain function, and decrease BPSD. People with AD their diagnosis and prognosis are faced with psycho-
are still people, with the same emotional needs as all people. logical problems that may be helped by regular counsel-
They need to know they are loved and they need to feel good ing or psychological support.
342 GROSSBERG AND DESAI

2. In the early stages of AD, patients can use diary and Specific Therapies/Approaches and Their Efficacy
dictaphone to help them remember events and con- Whatever the intervention, it is critical to match the level
versations. Patients should try and concentrate on doing of demand on the patient with his or her current capacities,
things they are good at, rather than pursuing tasks they avoiding both infantilization and frustration, and to modify
cannot manage. the environment insofar as possible to compensate for defi-
3. The desire to ‘‘go home’’ is common in people who have cits and to capitalize on the patient’s strengths.
AD, even when they’re still residing in the homes in
which they have lived independently for years. The care- Behavior-oriented approaches.—Behavioral approaches
giver should avoid correcting the patients or reassuring can be effective in lessening or abolishing problem behav-
them that they are in their homes. It is better to inquire iors. The steps involve careful description of the problem
where home is and who the patients might be worried behavior, assessment of specific antecedents and conse-

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
about there. quences, and implementing specific strategies that are often
4. Often people with dementia report being worried about suggested from the first 2 steps (116).
their young children—even though they’re now grown—
or about their parents—even if they’re deceased. We Emotion-oriented approaches.—These interventions in-
recommend reassuring these persons that everyone at clude but are not limited to supportive psychotherapy, remi-
home is OK and that those at home know that the pa- niscence therapy, validation therapy, sensory integration,
tients are OK. Asking what the patients like best about and simulated presence therapy (116–120). These interven-
‘‘home’’ and helping patients with AD to reminisce tions have been shown to improve mood and behavior.
about the good times in the home are also beneficial.
5. People with AD may curse or use language that the
family finds shocking. This may be a sign that the per- Cognition-oriented approaches.—These techniques in-
son has lost inhibition and impulse control and now says clude reality orientation and skills training (121,122). These
the first thing that enters his or her mind. It’s important interventions have shown slight transient improvement in
to remind the caregivers that this isn’t intentional and cognition, but there have also been reports of anger and
should not be taken personally. frustration. The slight improvements observed with some of
6. The caregivers should listen attentively and watch for these treatments have not lasted beyond the treatment ses-
nonverbal clues that might indicate hunger or thirst, sions and thus do not appear to warrant the risk of adverse
implied meanings, and expressed feelings. effects.
7. Caregivers need to adjust their expectations regarding
a loved one with AD throughout the course of the ill- Stimulation-oriented approaches.—These treatments in-
ness. Activities such as folding and stacking items gives clude activities (structured and unstructured) or recreational
the person with AD a sense of purpose and accom- therapies (e.g., crafts, games, pets) and art therapies (e.g.,
plishment. Maintaining a routine of meaningful activ- music, dance, art). There is some evidence that, while they
ities that promote success is important for people with are in use, these interventions decrease behavioral problems
AD, whose days are filled with frustration and failure. and improve mood (123–125).
8. Religious worship is a safe and soothing activity for
many persons with AD. It enables them to respond to Interventions for Families and Caregivers
their faith and spiritual needs through long-remembered Practical and creative problem solving, prayer, humor, the
rituals that connect past and present. For persons living dependable support of family and friends, expressive outlets,
alone, members of the congregation may be the first to the ability to forgive others and one’s self, and learning
notice the early signs of AD. how to economize the personal and family ‘‘energy’’ that is
9. Although a recent randomized, controlled trial did not available, for example, can all be effective coping strategies
find walking and or talking to provide any benefits in that families and caregivers should be encouraged to use. For
communication, ambulation, and functional status in resi- families who are struggling with various aspects of the issues
dents with AD (112), exercise has been documented to that arise while caring for a patient with AD, sessions of
improve muscle strength (thereby reducing frailty, func- family therapy have proven beneficial (126). For alleviating
tional decline, and injuries), even in frail residents in caregiver and family distress, a broad array of psychosocial
nursing homes (113,114). In addition, walking and other interventions was assessed in a meta-analysis of 18 studies
forms of light exercise may decrease problem behaviors. (127). The interventions included psychoeducation, support,
10. Meaningful activities may improve depression and di- cognitive-behavioral techniques, self-help, and respite care.
minish agitation, apathy, insomnia, and repetitive vocal- Exercise has been shown to improve caregiver outcomes (128).
ization. Individual and respite programs were found moderately
11. Graded assistance, practice, and positive reinforcement effective at reducing caregiver burden and dysphoria, but
should be used to increase functional independence. group interventions were only marginally effective.
12. Patients with AD may experience decreased problem Subsequent research buttressed the utility of adult day care
behaviors with music, particularly during meals and in reducing caregiver’s stress and depression and in en-
bathing. hancing their well-being (129). Targeted behavioral techni-
13. Use of therapeutic touch, including massage, may de- ques also improved the quality of caregivers’ sleep (130),
crease agitation/irritability (115). whereas psychoeducation and family support appeared to
MANAGEMENT OF ALZHEIMER’S DISEASE 343

promote better patient management. A range of behavioral with AD was provided by a controlled clinical trial (144). In
interventions for nursing home staff have been shown to be this randomized clinical trial, researchers compared 2
effective in improving behavioral symptoms of AD, such as behavioral therapies with a typical care condition, in which
incontinence (131,132), dressing problems (133), and verbal family caregivers were given information, advice, and sup-
agitation (134,135). A major problem is that interventions are port in their efforts to manage patient problems, and a wait
not maintained or implemented correctly by nursing home list control in 72 individuals who had major depression and
staff (136). Additional patient and caregiver benefits may AD (145). One type of behavioral treatment consisted of
be obtained by the use of computer networks to provide teaching caregivers to focus on increasing pleasant events for
education and support to the caregivers. the patient, while the other focused on improving the
caregivers’ approach to problem solving. Those in the 2
Depression behavior therapy conditions and those in typical care received

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
Population studies have shown that major depression the intervention in the form of 9 weekly, 60-minute sessions.
occurs in 12% of demented patients, while higher per- The study provided strong support for the efficacy of specific
centages have been reported in clinical samples. Depressive psychosocial treatments; the 2 behavior therapy conditions
symptoms are much more common in AD than major were equally beneficial in reducing depressive symptoms and
depression and afflict more than 30% of these patients (137). were superior to both the typical care and wait list conditions.
Depressed mood and anhedonia have been identified in Outcomes were similar for the typical care and wait list
49% of AD patients; these symptoms do not correlate with conditions, suggesting that, in the absence of a structured
either vegetative symptoms or cognitive symptoms of de- approach to treatment, professional contact alone is not
pression. Family history of depressive disorders increases effective. Depressed mood may also respond to improve-
the probability of developing depression during the course ments in living situation or stimulation-oriented treatments.
of AD. Common depressive symptoms and signs in AD in Somatic treatments for depression can be used in
those who do not meet criteria for major depression include demented patients to improve mood, functional status, and
loss of interest, lack of energy, thinking and concentration quality of life. These treatments should be considered even
difficulties, and psychomotor disturbances. Depressive symp- for patients with depressed mood who do not meet the
toms as a rule do not coexist with apathy in demented diagnostic criteria for major depression (132). Patients with
patients (138). Apathy appears to be a syndrome distinct severe or persistent depression should be treated with anti-
from depression, and its prevalence increases as the cogni- depressants. The choice among agents is based on the side
tive impairment in AD increases (138). Depressive symp- effect profile and the characteristics of a given patient.
toms add to functional impairment of AD patients and are Selective serotonin reuptake inhibitors (e.g., citalopram, ser-
associated with increased psychopathology in their care- traline, paroxetine) are probably the first-line treatments and
givers. If left untreated, depressive symptoms or syndromes are supported by many RCTs, although other agents such
persist or subside to some extent and then recur. As AD as mirtazapine and venlafaxine may be more appropriate for
progresses, depressive symptoms become less prominent some patients (146–150). Electroconvulsive therapy (ECT)
and often are overshadowed by other behavioral abnormal- is effective in the treatment of depression in dementia pa-
ities, including delusions, aggressive behavior, and agita- tients (151). ECT may be beneficial for patients with severe
tion. Depression of AD involves fewer required symptoms major depression who are ineligible for, cannot tolerate, or
than a major depressive episode, and less emphasis is do not respond to antidepressants (152). Individuals whose
placed on verbally weighted items. Provisional diagnostic cognitive symptoms recover fully with treatment of depres-
criteria for depression of AD have been proposed (139). sion are considered not to have been demented but rather
Health-care providers need to be aware that suicide to have a condition called dementia syndrome of depression;
attempts are not rare in AD patients (140). Thus, all AD however, about half of such persons may develop dementia
patients with depression should be carefully evaluated for in 5 years (153). The management of depression has been
suicide potential. Treatment is likely to be of value, with reviewed in detail in a recent issue of the Journal (154).
reported response rates of up to 85% (141).
There is some evidence that it is possible to modify Psychotic Symptoms
cognitive-behavioral therapies for depression to allow them It is estimated that approximately 30–50% of patients
to be administered to patients with mild dementia and to with AD develop delusions and hallucinations at some point
modify behavioral therapies to allow them to be provided for during the course of AD (155). Psychosis with AD may run
those with moderate to severe dementia (142,143). Cognitive in families (156). The psychotic symptoms consist of non-
therapy, seen as more promising in the early stages of elaborate persecutory delusions (e.g., accusations of some-
dementia, strives to help patients cope with depression by one stealing the patient’s possessions) and simple visual
reducing cognitive distortions and by fostering more adap- or, less commonly, auditory hallucinations. Psychotic symp-
tive perceptions (142). Behavioral therapy, seen as more toms often cause severe agitation and aggression as well as
promising for more moderately or severely affected adults emotional distress and may precipitate institutionalization or
with dementia, targets family caregivers directly—and pa- hospitalization.
tients indirectly—by helping caregivers identify, plan, and Mild psychotic symptoms associated with AD are best
increase pleasant activities for the patient, such as taking a managed with nonpharmacological interventions and cho-
walk, designed to improve their mood (130). Further affir- linesterase inhibitors. If antipsychotics are used, the goals
mation for behavioral therapy for depression of patients should be modest (reduction of emotional distress and
344 GROSSBERG AND DESAI

behavioral disturbances) and should not be resolution of on conventional antipsychotics can be safely switched to
psychotic symptoms. A dose decrease or discontinuation is atypical antipsychotics to minimize the risk of EPS and TD
recommended periodically for all patients with psychoses (167). Compared to conventional antipsychotics, atypical
associated with AD who receive antipsychotic medications antipsychotic drugs cause less TD (168). Cholinesterase in-
(157). The major predictors of relapse, if antipsychotics hibitors may benefit mild psychotic symptoms in dementia
are discontinued, are agitation and aggressive features. Al- patients (62,87). A recent study found the antidepressant
though conventional or typical antipsychotics (e.g., Halo- citalopram to be more efficacious than placebo in the short-
peridol, thioridazine, chlorpromazine, perphenazine, term hospital treatment of psychotic symptoms and behav-
fluphenazine, etc.) are somewhat better than placebo for ioral disturbances in nondepressed, demented patients (169).
psychosis of AD (158,159), the effect is modest, and the These drugs may be considered in AD patients with psychotic
high risk of toxicity (especially extrapyramidal syndrome symptoms before antipsychotics are considered.

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
[EPS] and tardive dyskinesia [TD]) usually makes them
inappropriate. Despite this, conventional antipsychotics Agitation
continue to be chosen over atypical antipsychotics for psy- The most common behavioral disturbance associated
chotic symptoms and aggression in patients with demen- with AD is agitation, which affects nearly three fourths
tia, even in industrialized countries (160). Even low-potency of patients and typically increases in severity as the disease
conventional antipsychotics (e.g., Thioridazine, chlorprom- progresses. Agitation involves verbal and physical aggres-
azine) are associated with high frequency of EPS in patients sion, increased psychomotor activity, repeated verbaliza-
with AD. Risperidone has the largest body of currently tion, screaming, etc. Agitation often co-occurs with anxiety,
available published evidence amongst the atypical anti- psychotic symptoms, and irritability. However, clinicians
psychotics for the treatment of psychoses associated with should be sure to exclude potential causes unrelated to AD,
AD. Two 12-week, randomized, double-blind, placebo- such as pain, medical illness, medication adverse effects, or
controlled trials of risperidone included nearly 1000 test environmental provocation.
subjects who were suffering from dementia with psychosis Mild to moderate agitation is best managed by non-
and agitation (AD, VaD, and related disorders) (161,162). pharmacologic interventions, as environmental factors and
They found that 1 mg was the optimal dose. Doses of caregiver patient interaction can significantly influence agi-
0.5–1.5 mg daily do not produce a significantly increased tation. Severe agitation may need a trial of psychotropics.
incidence of side effects, but nonetheless they are effective Controlled clinical trials for the treatment of agitation have
in controlling psychosis-driven behavioral disturbances and reported efficacy of atypical antipsychotics (161,162,164),
aggression. Risperidone was superior to haloperidol in terms antidepressants (148), beta-adrenergic blockers (170), and
of efficacy, while presenting a significantly more benign anticonvulsants (divalproex, carbamazepine) (171,172). A
side-effect profile. Risperidone at higher than 1.5-mg doses 16-week RCT in community-dwelling patients with AD and
is associated with increased EPS, although in some patients agitation did not find any statistically significant difference
with AD, EPS can occur even at lower dosages. Risperidone between the group receiving trazodone, the group receiving
was found to be effective and well tolerated over 13–46 haloperidol, and the group receiving behavioral approaches
months for nursing home residents with dementia and behav- (behavior management techniques to caregivers). There was
ioral disturbances, despite high rates of medical comor- slight worsening of cognition and function in the medica-
bidity and use of concomitant medications (163). A 6-week tion group (173). Uncontrolled studies and small controlled
randomized, double-blind, placebo-controlled study of olan- studies have suggested use of gabapentin and buspirone for
zapine was conducted in 206 nursing home patients with the treatment of agitation (174,175).
AD or VaD plus psychosis or severe agitation (164). Pa- Despite the widespread utilization of benzodiazepines
tients received either placebo or olanzapine in 5-mg, 10-mg, (BZ) in community-dwelling elders as well as in the nursing
or 15-mg doses. The most effective doses turned out to be home population, data supporting the use of BZ and other
5 mg or 10 mg. A 15-mg dose was not better than placebo antianxiety agents in AD are very limited. BZ may be used
but produced significant gait disturbance and sedation. In for a short duration in AD patients who have moderate to
some patients with AD, gait disturbance can occur even severe free-floating anxiety or who may be apprehensive
at lower dosages. In a 10-week, double-blind, placebo- about particular events, such as medical procedures or
controlled, randomized trial with nursing home residents admission to the hospital or a LTCF. Although treatment
with dementia and psychosis, quetiapine (average dose 120 with BZ may be efficient in the short term, their long-term
mg/day) was better than placebo in reducing symptoms of usage is limited by their gradual decline in efficiency and
agitation but not psychosis (165). A large, multicenter, the potential side effects of sedation, decreased cognitive
open-label study of quetiapine (average daily dose 100 mg) functioning, loss of coordination and unsteadiness, and
found it to be useful for psychotic symptoms in the elderly, paradoxical disinhibition of behavior (176). When in-
many of whom had dementia (166). The most common side dicated, a BZ with a short half-life, such as lorazepam or
effects associated with quetiapine were somnolence and oxazepam, is recommended. BZ have not been rigorously
falls. Ziprasidone is the newest atypical antipsychotic, and investigated in AD, and their effectiveness in controlling
to date there are very few data available in the elderly. behavioral disorders in patients with AD has not been
Randomized, controlled studies are needed before ziprasi- conclusively demonstrated. BZ perform better than placebo
done can be recommended as the drug of first choice for but not as well as antipsychotics in reducing agitation in
treatment of psychosis of AD. Patients with AD and psychosis patients with AD (177). Discontinuation of BZ has been
MANAGEMENT OF ALZHEIMER’S DISEASE 345

found to improve cognitive performance in nursing home Table 7. Psychotropics in the Management of Behavioral and
patients with dementia (178). Psychological Symptoms of AD (BPSD)
Daily Dose
Sleep Disturbances Indicated Clinical
Medication Starting (mg) Therapeutic (mg) Condition
Sleep disorder is common in AD and may be manageable
with nonpharmacologic interventions in many cases (179). Antidepressants
SSRIs
The most disturbing sleep disturbances include day/night
reversal of sleep pattern and the waking by the patient of the Citalopram 10 mg 20–40 mg Depression, anxiety,
agitation
caregiver. Appropriate sleep hygiene (including regular sleep Sertraline 25 mg 50–100 mg Depression, anxiety
and waking times, limited daytime sleeping, avoidance Paroxetine 10 mg 10–20 mg Depression, anxiety
of fluid intake in the evening), calming bedtime rituals, and Mirtazepine 7.5–15 mg 15–30 mg Depression, anxiety

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
adequate daytime physical and mental activities should be Venlafaxine 37.5 mg 75–150 mg Depression, anxiety
tried before pharmacological interventions. There is pre- Antipsychotics
liminary evidence from 2 small open trials for elderly sub- Risperidone 0.25 mg 0.5–1.5 mg Psychosis, aggression
jects with dementia (180,181) suggesting that early-morning Olanzapine 2.5 mg 5–10 mg Psychosis, aggression
or evening bright-light therapy may improve sleep (and Quetiapine 25 mg 50–200 mg Psychosis, aggression
possibly behavior as well). It is important to investigate Anticonvulsants
sleep disorders such as restless leg syndrome, REM sleep Valproate 125 mg 500–1000 mg Mania, aggression
behavior disorder, nocturnal myoclonus, and sleep apnea, Carbamazepine 100 mg 200–600 mg Mania, aggression
which are relatively common in elderly individuals. Sleep SSRI ¼ selective serotonin reuptake inhibitor.
apnea contraindicates the use of benzodiazepines or other
agents that suppress respiratory drive. Short-term use of zol-
pidem (5–10 mg) may be considered in certain situations management techniques to reduce difficult patient behaviors.
(182). Other drugs that may be tried judiciously include tra- Many of these interventions can be easily combined and may
zodone and sedating antidepressants such as mirtazepine. have an even greater impact on delaying institutionalization.
Benzodiazepine (lorazepam, temazepam) use should be
limited to short-term use for a few days in certain situations,
such as a short hospital stay. Any drug used as a sleep aid FACTORS MODIFYING TREATMENT DECISIONS
can increase the risk of falls. Patients should be counseled
not to use over-the-counter sleep aids containing anticho- Treatment of Comorbidities
linergic agents such as diphenhydramine (present in Tylenol Several diseases are more common in AD because they
PM) because of risk of delirium and hallucinations. are a consequence of the dementing process. These diseases
include new onset seizures, respiratory and urinary infec-
Initiating, Monitoring, and Maintaining tions, Parkinsonism, and pressure sores (185). In one study,
Safe Pharmacologic Treatment for BPSD more than 60% of patients with AD suffered from 3 or more
Selection of the right drug and the proper dosage should be comorbid conditions and complaints; virtually all (93%)
guided by the nature of the patient’s symptoms (Tables 7 and had at least 1 (186). A case-control study using 7195 death
8). A drug should be continued as long as it is effective, but no certificates found that patients who died of AD had higher
longer than needed. Its efficacy can be gauged more easily if incidences of Parkinson’s disease, seizures, sensory impair-
realistic goals have been set and target behaviors specified ments, infections, malnutrition, hip fractures and other in-
before initiating treatment. Achieving significant efficacy may juries, and pressure sores, compared to control subjects
require a few weeks of treatment. Rapidly switching from one (187). Treatment of individuals with advanced AD should
agent to another deprives the patient of an adequate trial. weigh the possible benefits for the patient against the burden
Recent data on beneficial effects of ChEIs and antidepressants imposed by such treatment. Most medical interventions
on behavioral disturbances including psychotic symptoms and cause discomfort, especially to patients with dementia (56).
agitation suggests that the intuitive approach of treating agita- Patients with advanced AD do not recognize the need for
ted AD patients experiencing psychotic symptoms with anti- therapeutic interventions, do not cooperate with treatment,
psychotics may not be the safest approach for many patients. and often actively oppose it. Minor routine procedures, such
as blood drawing or blood pressure measurement, typically
Preventing Premature Institutionalization precipitate behavioral disturbances. The dementing process
ChEI may postpone nursing home placement for AD also may limit the benefits of therapeutic interventions be-
(183,184). Day care for people with AD can delay insti- cause of reduced life expectancy and decreased treatment
tutionalization. Potentially treatable BPSD are risk factors effectiveness. Preventive measures, such as restricted diets
for institutionalization, and treating these symptoms might and screening procedures, are appropriate only in the earlier
delay or prevent institutionalization. A comprehensive sup- stages of AD. Similarly, treatment of chronic conditions such
port and counseling intervention for spouse-caregivers of pa- as hypertension and diabetes should be aggressive only in
tients with AD reduced time to nursing home placement by the earliest stages of AD. In more advanced stages, treat-
nearly 1 year, compared with those not receiving the inter- ment of comorbidities should be conservative and directed
vention (24,25). Interestingly, one of the key components toward prevention of potentially serious side effects (e.g.,
of that intervention was to teach the caregivers behavioral postural hypotension that may lead to falls and hip fracture).
346 GROSSBERG AND DESAI

Table 8. Principles of Pharmacotherapeutic Management considered potentially inappropriate for use in the elderly
of Behavioral Disturbances in Dementia Patients (191). Avoidance of unnecessary medications, use of the
1. Use drugs only when secondary causes (environmental, medical) of lowest effective dose, using medications with least anticho-
behavioral disturbances have been ruled out and nonpharmacologic linergic activity, vigilant monitoring aimed at early recog-
interventions have failed. nition, a thorough search for causes, and prompt treatment
2. Review current medications and consider tapering or discontinuing may diminish the prevalence and morbidity of delirium.
unnecessary, ineffective, or harmful medications.
3. Consider adverse drug reactions or drug–drug interactions as a
potential cause of behavioral disturbances.
Neurologic Issues
4. Target symptoms and their impact on functioning should be In later stages, extrapyramidal signs such as rigidity may
documented prior to initiation of drug therapy. become prominent, especially in patients treated with anti-
5. If pharmacotherapy is used, start low and go slow and use it in psychotic drugs (even the more modern ‘‘atypical’’ agents).

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
conjunction with nonpharmacologic interventions. Seizures have been reported in 10–20% of AD patients,
6. Select agents based on target symptoms, side-effect profile, and again, often late in the disease (192). Anticonvulsants such
individual patient characteristics.
as divalproex, carbamazepine, and oxcarbazepine may not
7. Give the medication for an adequate time at an adequate dose.
8. Closely monitor for and document side effects and beneficial effects.
only benefit seizures but may also decrease agitation and
9. If found beneficial, continue the medication for a few months. If the aggression in patients with late-stage AD. Myoclonus, or
patient has been stable for that period, consider decreasing or brisk irregular muscle contraction, occurs in 5–10% of AD
discontinuing the medication. patients (193). A high index of suspicion and treatment of
10. Educate patient and family regarding the benefits and side effects of seizures and myoclonus in later stages of AD is recom-
medications. mended. Recent epidemiological and clinicopathologic data
suggest overlaps between AD and cerebrovascular lesions
that may magnify the effects of mild AD pathology and
promote progression of cognitive and functional decline
Antibiotic therapy preferably should be limited to oral (194). Many AD patients also have comorbid cerebrovas-
preparations. Intravenous therapy is to be avoided in moder- cular disease (CVD) or risk factors for CVD. Good control
ate to severe AD patients because the demented patients of blood pressure, treatment of atrial fibrillation, and anti-
in advanced stages do not understand the need for the platelet therapy, such as low-dose aspirin, may help prevent
medication and frequently try to remove the intravenous further strokes.
catheters and often end up with physical restraints or psy-
chotropic drugs. Intramuscular administration of antibiotics
such as cephalosporins should be considered before intra- SITE-SPECIFIC ISSUES
venous therapy. The effectiveness of antibiotic therapy may
be limited by the recurrent nature of infections in advanced Emergency Room (ER),
dementia. Also, adverse effects of antibiotic use, such as Inpatient General Medical, or Surgical Services
gastrointestinal upset, diarrhea, allergic reactions, etc., should Patients with AD visit the ER twice as often as unaffected
be factored in before the decision is made to administer patients with similar conditions; these patients also remain
the drug. hospitalized for a longer time (195). Lyketsos and col-
leagues reported that hospitalized patients with dementia
Urinary Incontinence had longer stays, higher costs, and a greater frequency of
Behavior modification, scheduled toileting, and prompted medication-induced psychosis and delirium (196). Statisti-
voiding should be used to reduce urinary incontinence. Uri- cally significant findings were discerned despite the likeli-
nary incontinence with donepezil has been reported (188), hood that many cases of dementia were unrecognized,
and all ChEIs have a potential to precipitate or exacerbate which reflects the substantial impact of dementia on clinical
this problem. Treating urinary tract infection, fecal impac- services. For many patients with AD, hospitalization rep-
tion, and other causes of urinary incontinence may suffice resents a failure to prevent an unintentional injury, properly
in many cases. Use of anticholinergic agents such as tolte- manage a chronic disorder, or use an appropriate alternate
radine and oxybutynin should be minimized, as these drugs therapy, such as Hospice or temporary LTCF stay. Transfer
may counteract the effects of ChEIs. of AD individuals to an ER or hospital exposes patients with
AD to serious risks, such as increased confusion, agitation,
Delirium anorexia, incontinence, and falls (197). These risks and po-
Delirium is a significant problem for patients with AD, tential benefits of treating acute medical problems should
and it occurs much more frequently than it does in cog- be discussed with the family before transfer. The risks are
nitively intact patients (189). Possible causes of delirium even higher in more advanced AD patients because of the
include an acute illness (urinary tract infection, respiratory very low rates of successful outcome in terms of improved
infection) or medication toxicity. Compounds with anticho- survival and quality of life after treatment. Transfer to an ER
linergic effects (e.g., tricyclic antidepressants, low-potency or hospital should be used only when it is consistent with
antipsychotics, diphenhydramine, disopyramide phosphate) the overall goals of care and not as a default option. Having
or histamine-2 activity (cimetidine, ranitidine) are particu- family members or aides stay with the patient may help de-
larly likely to cause delirium, but many other classes of crease agitation and wandering that may occur in hospital-
medications can do so (190). Many of these medications are ized patients with AD.
MANAGEMENT OF ALZHEIMER’S DISEASE 347

Inpatient Psychiatric Units some cases and reinstate them in others, as deemed clin-
Individuals with AD may need hospitalization to inpatient ically necessary, is recommended. A structured education
psychiatric units for the treatment of psychotic, affective, program for nursing and medical staff has been shown to
and behavioral symptoms that are causing harm to self decrease antipsychotic usage in the nursing home setting
or others, severe symptoms not responding to outpatient without adverse outcomes (206). Pet therapy and/or the
treatment, or if there is need for ECT (198,199). A thorough introduction of the Eden Alternative may improve outcomes
search for psychosocial, general medical, or noncognitive in some, but not all, patients with dementia (207–209).
psychiatric difficulties that may be leading to the distur-
bance will often reveal a treatable problem. Both non- Special Care Dementia Unit
pharmacologic and pharmacologic interventions can be tried The social environment is also significant in mollifying
more readily and aggressively on inpatient units than in the behavioral manifestations of dementia. If demented indi-

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
outpatient settings. Clinical experience suggests that patients viduals are grouped together on a special care dementia unit,
with AD may not do well if treated in general adult units, many symptoms may be minimized. Grouping demented
where younger violent patients may pose some danger. Ge- patients together further eliminates the problem of demented
riatric-psychiatry units, which are designed to address the patients disrupting the routine of cognitively intact indi-
unique needs of the demented patient with staff trained in viduals by wandering into their rooms, rummaging through
the management of dementia patients, may be preferable. their belongings, or approaching them repeatedly with ques-
tions and unwanted physical contact. Well-designed studies
Long-Term Care are needed to demonstrate the benefits of such units.
According to the American Health Care Association,
more than 1.5 million individuals in the United States reside MILD COGNITIVE IMPAIRMENT (MCI)
in nursing homes. Within this population (elderly as well as MCI is a cognitive disorder with a high rate of conversion
nonelderly persons), 67–77% have dementia (100,201,202). to AD over many years (210). Short-term memory loss is
Institutionalization offers many AD patients the best dura- the most common abnormality. Patients with MCI perform
tion of survival, followed by a formal care package at home. similarly on memory tests when compared to patients with
In the nursing home, AD is underrecognized and under- AD, but they perform similarly to normal elderly controls on
treated. By the time patients with AD reach the nursing other cognitive tests and in activities of daily living (197).
home, the bulk of their needs are no longer for health care Neuropsychiatric symptoms in MCI have a prevalence inter-
but rather for custodial care. Most do not need high-tech mediate to that in healthy participants and those with AD
care. They need care that is mediated by touch and delivered (108). Recent evidence suggests that most patients with MCI
by professionals whose skilled eyes and hands can detect eventually develop AD (211). Patients with MCI should be
deterioration–professions who can intervene early and who carefully evaluated and followed. Biological markers, in-
can identify opportunities for improvement and rehabilita- cluding APOE, may be helpful in identifying patients at risk
tion. Psychoactive agents are one of the most prevalent class for conversion to dementia. While controlled trials for MCI
of drugs with potential for inappropriate use in elderly treatments are in progress, patients may be offered empiric
LTCF residents (203). Staff of LTCF should receive edu- treatment with vitamin E and ChEIs. The potential for ChEIs
cation about AD to reduce the use of unnecessary psycho- to delay onset of dementia in patients with MCI is currently
tropics. These facilities should be tailored to meet the needs being rigorously investigated. Patients with MCI may be
of patients with dementia and to adequately address be- encouraged to participate in such studies with ChEIs.
havioral symptoms. Skills training for people with AD in
LTCF may lead to an improvement in personal care skills. THE FUTURE oF AD TREATMENT
Structured activity programs can improve both behavior and
mood (204). LTCFs need to take advantage of the latest Pharmacotherapy
thinking in design, which can help both staff and patients Definitely effective treatments for AD are few. Research
with AD. AD-specific design features include high levels of to increase understanding of the underlying disease process
visual access, highly visible and signed toilet doors, indoor/ and to seek new and better treatments is robust. Clinical
outdoor wander-safe areas, increased lighting, age-appro- research, with active patient participation, is a current and
priate fixtures and fittings, and individualized personal space valuable means of treatment development for AD, with
(205). The interdisciplinary care model is recommended for government- and private industry–sponsored therapeutic
patient care to best address the increasingly complex needs trials currently numbering in the hundreds. Epidemiological
of AD residents in LTCF settings. The Omnibus Budget and postmortem studies have established a number of
Reconciliation Act of 1987 regulates the use of physical testable hypotheses (212). Secretase inhibitors (beta and
restraints and many psychotropic medications in nursing gamma) may stall A-beta amyloid production, thus pre-
homes. Health professionals practicing in nursing homes venting neuronal plaque formation that is theorized to cause
must be familiar with these regulations. Indications for anti- neuronal cell death. Such drugs are in a Phase-I stage of
psychotic medication treatment and available alternatives study. Beta-sheet blockers latch onto critical portions of
and outcomes should be carefully documented. A clinical spiral-shaped A-beta, helping them maintain their shape,
strategy of carefully considering which patients may be thereby stalling the formation of new fibrils and allowing
appropriate for withdrawal of antipsychotic medications and the body to clear A-beta from the brain, thus forestalling
for being prepared to maintain use of the medications in plaque formation. Such drugs are also currently in devel-
348 GROSSBERG AND DESAI

opment. Studies involving metal chelators such as desfer- from which point it empties CSF into the peritoneal cavity.
rioxamine and clioquinol to reduce plaque formation are The theory is that poor circulation of the CSF is contributing
also underway. The theory behind this is that metals such to AD by allowing the deposition of amyloid and or other
as aluminum, iron, zinc, and copper promote A-beta aggre- toxins. The study also found promising results in 12 pa-
gation and plaque formation, and drugs that bind to these tients: either a 3-month improvement or stabilization. A
metals (chelators) may prevent A-beta aggregation. Anti- larger, randomized, double-blind, controlled clinical trial is
bodies targeted to plaques (amyloid) can mark them for underway to elucidate the potential for this intervention in
destruction by the immune system, resulting in increased the treatment of AD.
clearance of plaque and reduced neuronal loss, and in
animal studies, antibodies to beta-amyloid have enhanced
memory (213,214). An Alzheimer vaccine works on this SUMMARY AND CONCLUSIONS

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
principle and is currently under investigation. However, the AD is one of the principal causes of disability and de-
initial human study with amyloid vaccine caused brain creased quality of life among the elderly and is a leading
inflammation and had to be halted. New research with a obstacle to successful aging. AD is a treatable condition.
more refined vaccine that has a high degree of selectivity Today’s treatment options are greatly improved over those
for the pathogenic target structures offers new hope (215). available a few years ago. The complexity and heterogeneity
Blocking of A-beta production with an antisense directed of AD requires comprehensive assessment and possibly
against the amyloid precursor protein also improves memory multiple layers of intervention for the patient as well as
in animals (216). Chronically high levels of glutamate have the caregiver. Current management focuses on establishing
been implicated in causing neuronal cell loss. Glutamate an early accurate clinical diagnosis, early institution of
antagonists such as memantine (which is already approved ChEIs, treating medical comorbidities and dementia-related
in Europe) may thus protect neuronal loss in AD patients. complications, ensuring appropriate services are provided,
Large multicenter trials are currently underway with nerve supporting caregivers, and treating cognitive and behavioral
growth factors (compounds that promote growth, survival, problems and functional deficits with appropriate non-
and regeneration of neurons), as well as drugs such as mem- pharmacological and pharmacologic interventions. A ther-
antine. However, the key event that will provide direction apeutic alliance between physician and caregiver is critical
for cure would be a full understanding of AD etiology. to the success of AD management. AD is a terminal illness,
The potential usefulness of hormone replacement, anti- and the patients and caregivers should be given an early
inflammatory drugs, and Ginkgo biloba extract for the pre- opportunity to decide about intensity of care before the dis-
vention of AD is currently being studied. Further information ease inevitably progresses. Physician involvement in ethical
about these studies is available from the Alzheimer’s Disease issues raised in the management of AD and a multidisci-
Education and Referral Center of the National Institutes of plinary team approach is strongly recommended.
Health (http://www.alzheimers.org/ir.html or 800-438- ChEIs should be considered in all patients with mild to
4380). These and other promising areas of investigation moderate AD. ChEIs have been shown to temporarily stabi-
bring hope that in the near future we will be able to turn our lize cognition and ADL and may delay nursing home place-
attention from the palliative treatment of AD to primary ment and reduce demands on caregiver time. Preliminary
prevention, or at least to delaying the progression of the evidence also indicates that these benefits may extend to
disease to its most disabling stages. patients with more advanced AD and rapidly progressive
AD as well as to patients with VaD and LBD. Patients not
responding to one agent in the class may respond to another.
Research Involving Genetics of AD
Discontinuation should be monitored; deterioration during
A 30-year-old woman with a genetic disorder linked
withdrawal indicates therapeutic benefit and the medication
to early onset of AD has become the first to successfully
should be reinstated. Other drugs such as memantine look
undergo in vitro fertilization with preimplantation genetic
promising in their effect in slowing functional decline
diagnosis (PGD) of the embryos to avoid passing the de-
in patients with moderate to severe AD. Estrogen should not
fect on to her children (217). PGD, prenatal diagnosis, pre-
be prescribed to treat AD.
implantation embryo selection, and presymptomatic testing
Psychotropics play a critical role in the management of
has been offered to families of patients who have early-onset
moderate to severe behavioral disturbances in patients with
familial AD. Complex legal and ethical issues surrounding
AD. Short-term programs directed toward educating family
these interventions need to be addressed before these inter-
caregivers about AD should be offered to improve caregiver
ventions can be routinely recommended. There is great poten-
satisfaction. Intensive long-term education and support ser-
tial for stem cell treatment of patients with AD, but ethical
vices (when available) should be offered to caregivers of
issues will need to be addressed before fetal stem cell re-
patients with AD to delay time to nursing home place-
search can be carried out.
ment. Professional caregivers need higher expectations and
better training and support. Staff of long-term care facilities
Surgical Interventions should receive education about AD to reduce the use of
Low-flow cerebrospinal fluid (CSF) drainage using a unnecessary psychotropics. This article is by no means ex-
shunt is a recent surgical intervention that was found to be haustive. Readers are encouraged to read other articles for
relatively safe in the treatment of patients with AD (218). treatment of problems/issues that we may not have ade-
The shunt is implanted in the lateral ventricle of the brain, quately addressed in the management of AD (Table 9).
MANAGEMENT OF ALZHEIMER’S DISEASE 349

Table 9. Further Recommended Reading Regarding Management 7. Hardy J. The Alzheimer family of diseases: many etiologies, one pa-
of Alzheimer’s Disease (AD) thogenesis? Proc Natl Acad Sci USA. 1997;94:2095–2097.
8. Dunkin JJ, Anderson-Hanley C. Dementia caregiver burden: a re-
1. Small GW, Rabins PV, Barry PP, et al. Diagnosis and treatment of view of the literature and guidelines for assessment and intervention.
Alzheimer’s disease and related disorders: consensus statement of the Neurology. 1998;51(suppl 1):S53–S60.
American Association for Geriatric Psychiatry, the Alzheimer’s 9. Goate A, Chartier-Harlin MC, Mullan M, et al. Segregation of a
Association, and the American Geriatrics Society. JAMA. missense mutation in the amyloid precursor protein gene with familial
1997;278:1363–1371. Alzheimer’s disease. Nature. 1991;349:704–706.
2. Doody RS, Stevens JC, Beck C, et al. Practice parameter: management 10. Sherrington R, Rogaev EI, Liang Y, et al. Cloning of a gene bearing
of dementia (an evidence-based review). Neurology. 2001;56:1154–1166. missense mutations in early-onset familial Alzheimer’s disease.
3. Suh Y, Checler F. Amyloid precursor protein, presenilins, and alpha- Nature. 1995;375:754–760.
synuclein: molecular pathogenesis and pharmacological applications 11. Levy-Lahad E, Wasco W, Poorkaj P, et al. Candidate gene for the
in Alzheimer’s disease. Pharmacol Rev. 2002;54:469–525. chromosome 1 familial Alzheimer’s disease locus. Science. 1995;269:

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
973–977.
4. Desai AK, Grossberg GT. Recognition and management of behavioral
12. Gauthier S. Advances in the pharmacotherapy of Alzheimer’s disease.
disturbances in dementia. Primary Care Companion J Clin Psychiatr.
CMAJ. 2002;166(5):616–623.
2001;3:93–109.
13. Wolfson C, Wolfson DB, Asgharian M, et al. A reevaluation of the
duration of survival after the onset of dementia. N Engl J Med. 2001;
344:1111–1116.
Patients with AD are at risk of becoming medical orphans 14. Salmon DP, Thomas RG, Pay MM, et al. Alzheimer’s disease can be
accurately diagnosed in very mildly impaired individuals. Neurology.
in a health system geared toward cure and in a culture where 2002;59:1022–1028.
it is ‘‘normal’’ for the elderly to suffer. AD patients and their 15. McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of
families have a right to receive competent, compassionate, Alzheimer’s disease: report of the NINCDS-ADRDA Work Group
and consistent care. With appropriate treatment, we can under the auspices of Department of Health and Human Services Task
Force on Alzheimer’s Disease. Neurology. 1984;34:939–944.
substantially reduce the number of AD patients receiving in- 16. Mace NL, Rabins PV. The 36-Hour Day. A Family Guide to Care for
appropriate medication, futile procedures, and hospitaliza- Persons With Alzheimer’s Dementia, Related Dementing Illnesses and
tion and surgeries. Memory Loss in Later Life. Revised ed. Baltimore, MD: John Hopkins
With active research in AD, some of the current re- University Press; 1991.
commendations may become outdated by the time of 17. Siegal AP, Siegal RS. Forget Me Not: Caring For and Coping With
Your Aging Parents. Berkeley, CA: Celestial Arts; 1993.
publication of this article. Also, physicians must decide to 18. American College of Medical Genetics/American Society of Human
adopt any particular recommendation in the light of available Genetics Working Group on ApoE and Alzheimer’s Disease.
resources and the circumstances of individual patients. Statement on use of apolipoprotein E testing for Alzheimer’s disease.
Patients and their families have reason to be optimistic. JAMA. 1995;274:1627–1629.
19. National Institute on Aging/Alzheimer’s Association Working Group.
The intense ongoing research in the area of etiology and Apolipoprotein E genotyping in Alzheimer’s disease. Lancet. 1996;347:
treatment of AD offers hope and confidence that treatments 1091–1095.
to delay onset of AD; to treat, inhibit, and reverse symp- 20. Green RC. Risk assessment for Alzheimer’s disease with genetic
toms; and, ultimately, to prevent AD, will indeed be dis- susceptibility testing: has the moment arrived? Alzheimer’s Care Q.
covered. Until then, patients and families can benefit from 2002;Summer:208–214.
21. Schulz R, O’Brien AT, Bookwala J, et al. Psychiatric and physical
the variety of pharmacologic and nonpharmacologic inter- morbidity effects of dementia caregiving: prevalence, correlates, and
ventions described here. causes. Gerontologist. 1995;35:771–791.
22. Schulz R, O’Brien AT, Bookwala J, Fleissner K. Psychiatric and
physical morbidity effects of dementia caregiving: prevalence, cor-
ACKNOWLEDGMENT relates, and causes. Gerontologist. 1995;35:771–791.
Address correspondence to Abhilash K. Desai, MD, 808 Delta Pine 23. Rabins P, Mace N, Lusas M. The impact of dementia on the family.
Lane, Sikeston, MO 63801-5735. E-mail: zachary@sbmu.net JAMA. 1982;248:333–335.
24. Mittelman MS, Ferris SH, Steinberg G, et al. An intervention that
delays institutionalization of Alzheimer’s disease patients: treatment
of spouse caregivers. Gerontologist. 1993;33:730–740.
25. Mittelman MS, Ferris SH, Shulman E, et al. A family intervention to
delay nursing home placement of patients with Alzheimer’s disease:
REFERENCES a randomized controlled trial. JAMA. 1996;276:1725–1731.
1. Hendrie HC. Epidemiology of dementia and Alzheimer’s disease. Am 26. Chiverton P, Caine E. Education to assist spouses in coping with
J Geriatr Psychiatr. 1998;6(2 suppl 1):S3–S18. Alzheimer’s disease: a controlled trial. J Am Geriatr Soc. 1989;37:
2. Evans DA, Funkenstein HH, Albert MS, et al. Prevalence of Alzhei- 593–598.
mer’s disease in a community population of older persons. JAMA. 27. Brodaty H, Gresham M. Effect of training program to reduce stress in
1989;262:2551–2556. carers of patients with dementia. BMJ. 1989;299:1375–1379.
3. Evans DA. Estimated prevalence of Alzheimer’s disease in the US. 28. Lawton MP, Brody EM, Saperstein AR. A controlled study of respite
Milbank Q. 1990;68:267–289. service for care givers of Alzheimer’s patients. Gerontologist. 1989;29:
4. Brookmeyer R, Gray S, Kawas C. Projections of Alzheimer’s disease 8–16.
in the United States and the public health impact of delaying disease 29. Mohide EA, Pringle DM, Streiner DL, et al. A randomized trial of
onset. Am J Public Health. 1998;88:1337–1342. family care giver support in the home management of dementia. J Am
5. Small GW, Rabins PV, Berry PP, et al. Diagnosis and treatment of Geriatr Soc. 1990;38:446–454.
Alzheimer’s disease and related disorders: consensus statement of 30. Doraiswamy PM, Leon J, Cummings JL, Marin D, Neumann PJ.
the American Association for Geriatric Psychiatry, the Alzheimer’s Prevalence and impact of medical comorbidity in Alzheimer’s disease.
Association, and the American Geriatrics Society. JAMA. 1997;278: J Gerontol Med Sci. 2002;57A:M173–M177.
1363–1371. 31. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults:
6. Khachaturian ZS. Diagnosis of Alzheimer’s disease. Arch Neurol. evidence for a phenotype. J Gerontol Med Sci. 2001;56A:M146–
1985;42:1097–1105. M156.
350 GROSSBERG AND DESAI

32. Maguire CP. Telling the diagnosis of dementia: consider each patient 60. Olin J, Schneider L. Galantamine for Alzheimer’s disease [Cochrane
individually. Int Psychogeriatr. 2002;14:123–126. Review]. In: The Cochrane Library. Issue 2. Oxford: Update Software;
33. Pinner G, Bouman WP. To tell or not to tell: on disclosing the 2002.
diagnosis of dementia. Int Psychogeriatr. 2002;14:127–137. 61. Birks J, Grimley Evans J, Iakovidou V, Tsolaki M. Rivastigmine for
34. Ross J. Rules over research on mental patients invalid. NY Law J. Alzheimer’s disease [Cochrane Review]. In: The Cochrane Library.
1996;10:25–30. Issue 2. Oxford: Update Software; 2002.
35. Overman W Jr, Stoudemire A. Guidelines for legal and financial 62. Cummings JL. Cholinesterase inhibitors: a new class of psychotropic
counseling of Alzheimer’s disease patients and their families. Am J agents. Am J Psychiatr. 2000;157:4–15.
Psychiatr. 1988;145:1495–1500. 63. Fillit H, Gutterman EM, Brooks RL. Impact of donepezil on caregiving
36. Valcour VG, Masaki KH, Blanchette PL, et al. Driving and dementia. burden for patients with Alzheimer’s disease. Int Psychogeriatr. 2000;
J Am Geriatr Soc. 2002;50:1265–1267. 12:389–401.
37. Porter MM, Whitton MJ. Assessment of driving with the global 64. Farlow M, Potkin S, Koumaras B. Analysis of outcome in retrieved
positioning system and video technology in young, middle-aged, and drop-out patients: a rivastigmine versus placebo, 26-week Alzheimer’s

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
older drivers. J Gerontol Med Sci. 2002;57:M578–M582. trial. Arch Neurol. In press.
38. Margolis KL, Kerani RP, McGovern P, Songer T, Cauley JA, Ensrud 65. Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of
KE. Risk factors for motor vehicle crashes in older women. J Gerontol tacrine administration in patients with Alzheimer’s disease. JAMA.
1994;271:992–998.
Med Sci. 2002;57A:M186–M191.
66. Waldemar G, Winblad B, Engedal K, et al. Donepezil benefits pa-
39. Lundberg, et al. Dementia and driving: an attempt at a consensus.
tients with either mild or moderate Alzheimer’s disease over one year.
Alzheimer’s Dis Assoc Disorders. 1997;11:28–37.
Neurology.
40. Dubinski, et al. Practice parameter: risk of driving and Alzheimer’s
67. Mohs R, Doody R, Morris J, et al. A 1-year, placebo-controlled
disease. Neurology. 2000;54:2205–2211. preservation of function survival study of donepezil in AD patients.
41. Spangenberg KB, Wagner MT, Hendrix S, Bachman DL. Firearm
Neurology. 2001;57:481–488.
presence in households of patients with Alzheimer’s disease and 68. Dooley M, Lamb HM. Donepezil. A review of its use in Alzheimer’s
related dementias. J Am Geriatr Soc. 1999;47:1183–1186. disease. Drugs Aging. 2000;16:199–226.
42. Hughes JC, Louw SJ. Electronic tagging of people with dementia who 69. Carrier L. Donepezil and paroxetine: possible drug interaction. J Am
wander. BMJ. 2002;325:847–848. Geriatr Soc. 1999;47:1037.
43. Smith GE, Kokmen E, O’Brien PC. Risk factors for nursing home 70. Verrico MM, Nace DA, Towers AL. Fulminant chemical hepatitis pos-
placement in a population-based dementia cohort. J Am Geriatr Soc. sibly associated with donepezil and sertraline therapy. J Am Geriatr
2000;48:519–525. Soc. 2000;48:1659–1663.
44. Yaffe K, Fox P, Newcomer R, et al. Patient and caregiver characteristics 71. Grossberg GT, Stahelin HB, Messina JC, et al. Lack of adverse
and nursing home placement in patients with dementia. JAMA. 2002; pharmacodynamic drug interactions with rivastigmine and twenty-two
287:2090–2097. classes of medications. Int J Geriatr Psychiatr. 2000;15:242–247.
45. Wilson SA. Transition to nursing home life: a comparison of planned 72. Farlow MR, Hake A, Messina J, et al. Response of patients with
and unplanned admissions. J Adv Nurs. 1997;26:864–871. Alzheimer’s disease to rivastigmine treatment is predicted by the rate
46. Coyne AC, Reichman WE, Berbig LJ. The relationship between de- of disease progression. Arch Neurol. 2001;58:417–422.
mentia and elder abuse. Am J Psychiatr. 1993;150:643–646. 73. Raskind MA, Peskind ER, Wessel T, Yuan W. Galantamine in AD:
47. Lachs MS, Williams C, O’Brien S, et al. Risk factors for reported a 6-month randomized, placebo-controlled trial with a 6-month ex-
elder abuse and neglect: a nine-year observational cohort study. tension: the Galantamine USA-1 Study Group. Neurology. 2000;54:
Gerontologist. 1997;37:469–474. 2261–2268.
48. Michel JP, Pautex S, Zekry D, Zulian G, Gold G. End-of-life care of 74. Giacobini E, ed. Cholinesterase inhibitors: from the Calabar bean to
persons with dementia. J Gerontol Med Sci. 2002;57A:M640–M644. Alzheimer therapy. In: Cholinesterases and Cholinesterase Inhibitors.
49. Reynish W, Andrieu S, Nourhashemi F, Vellas B. Nutritional factors London: Martin Dunitz; 2000:181–226.
and Alzheimer’s disease. J Gerontol Med Sci. 2001;56A:M675– 75. Auriacombe S, Pere JJ, Loria-Kanza Y, Vellas B. Efficacy and safety
M680. of rivastigmine in patients with Alzheimer’s disease who failed to
50. Young KWH, Greenwood CE. Shift in diurnal feeding patterns in benefit from treatment with donepezil. Curr Med Res Opin. 2002;18:
nursing home residents with Alzheimer’s disease. J Gerontol Med Sci. 129–138.
2001;56A:M700–M706. 76. Robillard A, Arts R, Nasreddine Z, Rasmussen L. Efficacy of
51. Morley JE. Decreased food intake with aging. J Gerontol Med Sci. galantamine in patients not responding to donepezil (18-week interim
2001;56A(Special Issue 2):81–88. results). J Neurol Sci. 2001;187(suppl 1):S59.
52. Van der Steen JT, Ooms ME, Mehr D, et al. Severe dementia and 77. Robillard A, McKelvey R, Nasreddine Z. Galantamine improves sleep
adverse outcomes of nursing home-acquired pneumonia: evidence for in patients withdrawn from donepezil (preliminary results). J Neurol
mediation by functional and pathophysiological decline. J Am Geriatr Sci. 2001;187(suppl 1):S55.
78. Emre M. Switching cholinesterase inhibitors in patients with
Soc. 2002;50:439–448.
Alzheimer’s disease. Int J Clin Pract Suppl. 2002;127:64–72.
53. Finucane TE, Christmas C, Travis K. Tube feeding in patients with
79. Morris JC. Therapeutic continuity in Alzheimer’s disease: switching
advanced dementia: a review of the evidence. JAMA. 1999;282:1365–
patients to galantamine. Clin Ther. 2001;23(suppl A):A1–A2.
1370. 80. Doody RS, Geldmacher DS, Gordon B, et al. Open-label, multicenter,
54. Gillick MR. Sounding board: rethinking the role of tube feeding in phase 3 extension study of the safety and efficacy of donepezil in pa-
patients with advanced dementia. N Engl J Med. 2000;342:206–210. tients with Alzheimer’s disease. Arch Neurol. 2001;58:427–433.
55. Volicer L, Rheaume Y, Riley ME, et al. Discontinuation of tube feed- 81. Farlow MR, Anand R, Messina J Jr, et al. A 52-week study of the
ing in patients with dementia of the Alzheimer type. Am J Alzheimer’s efficacy of rivastigmine in patients with mild to moderately severe
Care. 1990;5:22–25. Alzheimer’s disease. Eur Neurol. 2000;44:236–241.
56. Volicer L, McKee A, Hewitt S. Dementia. Palliative care. Neurol Clin. 82. Tariot PN, Solomon PR, Morris JC, et al. A 5-month, randomized,
2001;19:867–885. placebo-controlled trial of galantamine in AD. The Galantamine USA-
57. Volicer L, Rheaume Y, Brown J, et al. Hospice approach to the 10 Study Group. Neurology. 2000;54:2261–2268.
treatment of patients with advanced dementia of the Alzheimer type. 83. Feldman H, Gauthier S, Hecker J, et al. A 24-week, randomized,
JAMA. 1986;256:2210–2213. double-blind study of donepezil in moderate to severe Alzheimer’s
58. O’Brien JT, Ballard CG. Drugs for Alzheimer’s disease. BMJ. 2001; disease. Neurology. 2001;57:613–620.
323:123–124. 84. Tariot PN, Cummings JL, Katz IR, et al. A randomized, double-blind,
59. Birks JS, Melzer D, Beppu H. Donepezil for mild and moderate placebo-controlled study of the efficacy and safety of donepezil in
Alzheimer’s disease [Cochrane review]. In: The Cochrane Library. patients with Alzheimer’s disease in the nursing home setting. J Am
Issue 2. Oxford: Update Software; 2002. Geriatr Soc. 2001;49:1590–1599.
MANAGEMENT OF ALZHEIMER’S DISEASE 351

85. Anand R, Koumaras B, Hartman RD. The effects of rivastigmine on 107. Desai AK, Grossberg GT. Recognition and management of behavioral
behavioral symptoms in severe AD patients in a nursing home setting. disturbances in dementia. Primary Care Companion J Clin Psychiatr.
Neurobiol Aging. 2002;21(suppl 1):S220–S221. 2001;3:93–109.
86. Erkinjuntti T, Kurz A, Gauthier S, et al. Efficacy of galantamine in 108. Lyketsos CG, Lopez O, Jones B, et al. Prevalence of neuropsychiatric
probable vascular dementia and Alzheimer’s disease combined with symptoms in dementia and mild cognitive impairment. JAMA. 2002;
cerebrovascular disease: a randomized trial. Lancet. 2002;359:1283– 288:1475–1483.
1290. 109. Small GW, Rabins PV, Barry PP, et al. Diagnosis and treatment of
87. McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in Alzheimer’s disease and related disorders. JAMA. 1997;278:1363–
dementia with Lewy bodies: a randomized, double-blind, placebo- 1371.
controlled international study. Lancet. 2000;356:2031–2036. 110. Patterson CJS, Gauthier S, Bergman H, et al. The recognition, as-
88. Wesnes KA, McKeith IG, Ferrara R, et al. Effects of rivastigmine sessment and management of dementing disorders: conclusions from
on cognitive function in dementia with Lewy bodies: a randomized the Canadian Consensus Conference on Dementia. Can Med Assoc J.
placebo-controlled international study using the cognitive drug research 1999;160(suppl 12):S1–S15.

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
computerized assessment system. Dement Geriatr Cogn Disorders. 111. Mega MS, Masterman DM, O’Connor SM, et al. The spectrum of
2002;13:183–192. behavioral responses to cholinesterase inhibitor therapy in Alzheimer
89. Tiraboschi P, Hansen LA, Alford M, et al. Early and widespread disease. Arch Neurol. 1999;56:1388–1393.
cholinergic losses differentiate dementia Lewy bodies from Alzheimer 112. Cott CA, Dawson P, Sidani S, Wells D. The effects of a walking/
disease. Arch Gen Psychiatr. 2002;59:946–951. talking program on communication, ambulation, and functional status
90. Prasher VP, Huxley A, Haque MS. A 24-week, double-blind, placebo- in residents with Alzheimer’s disease. Alzheimer’s Dis Assoc Dis-
controlled trial of donepezil patients with Down syndrome and orders. 2002;16:81–87.
Alzheimer’s disease—pilot study. Int J Geriatr Psychol. 2002;17: 113. Fiatarone MA, Marks EC, Ryan ND, et al. High-intensity strength
270–278. training in nonagenarians. Effects on skeletal muscle. JAMA. 1990;
91. Farlow MR, Cyrus PA. Metrifonate therapy in Alzheimer’s disease: 263:3029–3034.
a pooled analysis of four randomized, double-blind, placebo-con- 114. Singh MAF. Exercise comes of age: rationale and recommendations
trolled trials. Dement Geriatr Cogn Disorders. 2000;11:202–211. for a geriatric exercise prescription. J Gerontol Med Sci. 2002;57A:
92. Winblad B, Poritis N. Memantine in severe dementia: results of the M262–M282.
9M-Best Study (benefit and efficacy in severely demented patients 115. Snyder M, Egan EC, Bruns KR. Efficacy of hand massage in de-
during treatment with memantine). Int J Geriatr Psychiatr. 1999;14: creasing agitation behaviors associated with care activities in persons
135–146. with dementia. Geriatr Nurs. 1995;16:60–63.
93. Oken BS, Storzbach DM, Kaye JA. The efficacy of Ginkgo biloba on 116. Rabins P, Bland W, Bright-Long L, et al. Practice Guideline for the
cognitive function in Alzheimer’s disease. Arch Neurol. 1998;55:1409– Treatment of Patients With Alzheimer’s Disease and Other Dementias
1415.
of Late Life. Practice Guidelines for the Treatment of Psychiatric
94. Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline,
Disorders Compendium. Washington, DC: American Psychiatric
alpha-tocopherol, or both as treatment for Alzheimer’s disease. N Engl
Association; 2002.
J Med. 1997;336:1216–1222.
117. Robichaud L, Hebert R, Desrosiers J. Efficacy of a sensory integration
95. Tabet N, Birks J, Grimley Evans J, et al. Vitamin E for Alzheimer’s
program on behaviors of inpatients with dementia. Am J Occup Ther.
disease [Cochrane Review]. In: The Cochrane Library. Issue 4. Oxford:
1994;48:355–360.
Update Software; 2002.
118. Woods P, Ashley J. Simulated presence therapy: using selected mem-
96. Wilcock GK, Birks J, Whitehead A, et al. The effect of selegiline in
the treatment of people with Alzheimer’s disease: a meta-analysis of ories to manage problem behaviors in Alzheimer’s disease patients.
published trials. Int J Geriatr Psychiatr. 2002;175–183. Geriatr Nurs. 1995;16:9–14.
97. Mulnard RA, Cotman CW, Kawas C, et al. Estrogen replacement 119. Baines S, Saxby P, Ehlert K. Reality orientation and reminiscence
therapy for treatment of mild to moderate Alzheimer’s disease. JAMA. therapy: a controlled cross-over study of elderly confused people. Br J
2000;283:1007–1015. Psychiatr. 1987;151:222–231.
98. Henderson VW, Paganini-Hill A, Miller BL, et al. Estrogen for 120. Kiernat JM. The use of life review activity with confused nursing
Alzheimer’s disease in women: randomized, double-blind, placebo- home residents. Am J Occup Ther. 1979;33:306–310.
controlled trial. Neurology. 2000;54:295–301. 121. Powell-Proctor L, Miller E. Reality orientation: a critical appraisal. Br
99. Kyomen HH, Hennen J, Gottlieb GL, Wei JY. Estrogen therapy and J Psychiatr. 1982;140:457–463.
noncognitive psychiatric signs and symptoms in elderly patients with 122. Tappen RM. The effect of skill training on functional abilities of
dementia. Am J Psychiatr. 2002;159:1225–1227. nursing home residents with dementia. Res Nurs Health. 1994;17:
100. Kyomen HH, Satlin A, Hennen J, Wei JY. Estrogen therapy and 159–165.
aggressive behavior in elderly patients with moderate-to-severe de- 123. Gerber GJ, Prince PN, Snider HG, et al. Group activity and cognitive
mentia: results from a short-term, randomized, double-blind trial. Am improvement among patients with Alzheimer’s disease. Hosp Com-
J Geriatr Psychiatr. 1999;7:339–348. munity Psychiatr. 1991;42:843–845.
101. Zandi PP, Carlson MC, Plassman BL, et al. Hormone replacement 124. Karlsson I, Brane G, Melin E, et al. Effects of environmental stimu-
therapy and incidence of Alzheimer’s disease in older women: the lation on biochemical and psychological variables in dementia. Acta
cache county study. JAMA. 2002;288:2123–2129. Psychiatr Scand. 1988;77:207–213.
102. Fillit HM. The role of hormone replacement therapy in the prevention 125. Rovner BW, Steel CD, Shmuely Y, Folstein MF. A randomized trial
of Alzheimer’s disease. Arch Intern Med. 2002;162:1934–1942. of dementia care in nursing homes. J Am Geriatr Soc. 1996;44:7–13.
103. Ott BR, Belazi D, Lapane KL. Cognitive decline among female 126. Dunkin JJ, Anderson-Hanley C. Dementia caregiver burden: a review
estrogen users in nursing homes. J Gerontol Med Sci. 2002;57A: of the literature and guidelines for assessment and intervention.
M594–M598. Neurology. 1998;51(suppl 1):S53–S60.
104. Petitti DB, Buckwalter G, Crooks VC, Chiu V. Prevalence of de- 127. Knight BG, Lutzky SM, Macofsky-Urban F. A meta-analytic review
mentia in users of hormone replacement therapy as defined by pre- of interventions for caregiver distress: recommendations for future re-
scription data. J Gerontol Med Sci. 2002;57A:M532–M538. search. Gerontologist. 1993;33:240–248.
105. Petitti DB, Buckwalter G, Crooks VC, Chiu V. Prevalence of demen- 128. King AC, Gaumann K, O’Sullivan P, Wilcox S, Castro C. Effects of
tia in users of hormone replacement therapy as defined by prescription moderate–intensity exercise on physiological, behavioral, and emo-
data. J Gerontol Med Sci. 2002;57A:M532–M538. tional responses to family caregiving: a randomized controlled trial.
106. Finkel SI, Costa e Silva, Cohen G, et al. Behavioral and psychological J Gerontol Med Sci. 2002;57A:M26–M36.
signs and symptoms of dementia: a consensus statement on current 129. Zarit SH, Stephens MA, Townsend A, Greene R. Stress reduction for
knowledge and implications for research and treatment. Int Psycho- family caregivers: effects of adult day care use. J Gerontol Sci Soc.
geriatr. 1996;8(suppl 3):497–500. 1998;53B:S267–S277.
352 GROSSBERG AND DESAI

130. McCurry SM, Logsdon RG, Teri L. Behavioral treatment of sleep 153. Alexopoulos G, et al. The course of geriatric depression with ‘‘rever-
disturbance in elderly dementia caregivers. Clin Gerontol. 1996;17: sible dementia’’: a controlled study. Am J Psychiatr. 1993;150:1693–
35–50. 1699.
131. Burgio LD, Engel BT, Hawkins A, et al. A staff management system 154. Blazer DG. Depression in late life: review and commentary. J Geron-
for maintaining improvements in continence with elderly nursing home tol Med Sci. 2003;58A:249–265.
residents. J Appl Behav Anal. 1990;23:111–118. 155. Wragg RE, Jeste DV. Overview of depression and psychosis in
132. Schnelle JF, MacRae PG, Ouslander JG, et al. Functional incidental Alzheimer’s disease. Am J Psychiatr. 1989;146:350–353.
training, mobility performance, and incontinence care with nursing 156. Sweet RA, Nimgaonkar VL, Devlin B, et al. Increased familial risk of
home residents. J Am Geriatr Soc. 1995;43:1356–1362. the psychotic phenotype of Alzheimer’s disease. Neurology. 2002;
133. Beck C, Heacock P, Mercer SO, et al. Improving dressing behavior 58:907–911.
in cognitively impaired nursing home residents. Nurs Res. 1997;46: 157. Alexoupoulous GS, Silver JM, Kahn DA, et al. The Expert Consensus
126–132. Guideline Series: agitation in older persons with dementia. Postgrad
134. Burgio L, Scilley K, Hardin JM, Hsu C, Yancey J. Environmental Med. [special report]. 1998: 1–88.

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
‘‘white noise’’: an intervention for verbally agitated nursing home 158. Lanctot KL, Best TS, Mittmann N, et al. Efficacy and safety of
residents. J Gerontol Psychol Sci. 1996;51B:P364–P373. neuroleptics in behavioral disorders associated with dementia. J Clin
135. Cohen-Mansfield J, Werner P. Management of verbally disruptive Psychiatr. 1998;59:550–561.
behaviors in nursing home residents. J Gerontol Med Sci. 1997;52A: 159. Schneider LS, Pollock VE, Lyness SA. A metaanalysis of controlled
M369–M377. trials of neuroleptic treatment in dementia. J Am Geriatr Soc. 1990;
136. Schnelle JF, Cruise PA, Rahman A, Ouslander JG. Developing 38:553–563.
rehabilitative behavioral interventions for long-term care: technology 160. Scott K, Lawrence RM, Duggal A, Brooks E. Prescribing patterns for
transfer, acceptance, and maintenance issues. J Am Geriatr Soc. 1998; psychotic and behavioral symptoms in dementia: a national survey.
46:771–777. Psychiatr Bull. 2002;26:288–290.
137. Katz IR. Diagnosis and treatment of depression in patients with 161. Katz IR, Jeste DV, Mintzer JE, et al. Comparison of risperidone and
Alzheimer’s disease and other dementias. J Clin Psychiatr. 1998; placebo for psychosis and behavioral disturbances associated with
59(suppl 9):38–44. dementia. A randomized, double-blind trial. Risperidone Study Group.
138. Landes AM, Sperry S, Strauss ME. Apathy in Alzheimer’s disease. J Clin Psychiatr. 1999;60:107–115.
J Am Geriatr Soc. 2001;49:1700–1707. 162. DeDeyn PP, Rabheru K, Rasmussen A, et al. A randomized trial
139. Olin JT, Schneider LS, Katz IR, et al. Provisional diagnostic criteria of risperidone, placebo, and haloperidol for behavioral symptoms of
for depression of Alzheimer’s disease. Am J Geriatr Psychiatr. 2002; dementia. Neurology. 1999;53:899–901.
10:125–128. 163. Goldberg RJ. Long-term use of risperidone for the treatment of demen-
140. Barak Y, Aizenberg D. Suicide amongst Alzheimer’s disease patients: tia-related behavioral disturbances in a nursing home population.
a 10-year survey. Dement Geriatr Cogn Disorders. 2002;14:101–103. Int J Geriatr Psychopharmacol. 1999;2:1–4.
141. Eccles M, Clarke J, Livingston M, et al. North of England evidence 164. Street JS, Clark WS, Gannon KS, et al. Olanzapine treatment of
psychotic and behavioral symptoms in patients with Alzheimer’s
based guidelines development project; guideline for the primary care
disease in nursing care facilities: a double-blind, randomized, placebo-
management of dementia. BMJ. 1998;317:802–808.
controlled trial. The HGEU Study Group. Arch Gen Psychiatr. 2000;
142. Teri L, Gallagher-Thompson D. Cognitive-behavioral interventions
57:968–976.
for treatment of depression in Alzheimer’s patients. Gerontologist.
165. Tariot PN, Schneider L, Katz I, et al. Quetiapine in nursing home
1991;3:413–416.
residents with Alzheimer’s dementia and psychosis. Am J Geriatr
143. Teri L, Uomoto J. Reducing excess disability in dementia patients:
Psychiatr. 2002;10(suppl 1):93.
training caregivers to manage patient depression. Clin Gerontol. 1991;
166. McManus DQ, Arvanitis LA, Kowalcyk BB. Quetiapine, a novel
10:49–63.
antipsychotic: experience in elderly patients with psychotic disorders.
144. Teri L, Logsdon RG, Uomoto, et al. Behavioral treatment of depres-
Seroquel Trial 48 Study Group. J Clin Psychiatr. 1999;60:292–298.
sion in dementia patients: a controlled clinical trial. J Gerontol Psychol 167. Lane H, Chang Y, Su M, et al. Shifting from haloperidol to ris-
Sci. 1997;52B:P159–P166. peridone for behavioral disturbances in dementia: safety, response
145. American Psychiatric Association. Practice guideline for the treatment predictors, and mood effects. J Clin Psychopharmacol. 2002;22:4–10.
of patients with Alzheimer’s disease and other dementias of late life. 168. Jeste DV, Okamoto A, Napolitano J, et al. Low incidence of persistent
Am J Psychiatr. 1997;154:1–39. tardive dyskinesia in elderly patients with dementia treated with ris-
146. Taragano FE, Lyketsos CG, Mangone CA, et al. A double-blind, peridone. Am J Psychiatr. 2000;157:1150–1155.
randomized, fixed-dose trial of fluoxetine vs. amitriptyline in the treat- 169. Pollock BG, Mulsant BH, Rosen J, et al. Comparison of citalopram,
ment of major depression complicating Alzheimer’s disease. Psycho- perphenazine, and placebo for the acute treatment of psychosis and
somatics. 1997;38:246–252. behavioral disturbances in hospitalized, demented patients. Am J
147. Lyketsos CG, Sheppard JM, Steel CD, et al. Randomized, placebo- Psychiatr. 2002;159:460–465.
controlled, double-blind clinical trial of sertraline in the treatment of 170. Shankle WR, Nielson KA, Cotman CW. Low dose propranolol
depression complicating Alzheimer’s disease: initial results from the reduces aggression and agitation resembling that associated with orbito-
Depression in Alzheimer’s Disease Study. Am J Psychiatr. 2000;157: frontal dysfunction in elderly demented patients. Alzheimer’s Dis Assoc
1686–1689. Disord. 1995;9:233–237.
148. Nyth AL, Gottfies CG. The clinical efficacy of citalopram in treatment 171. Porsteinsson AP, Tariot PN, Erb R, et al. Placebo-controlled study
of emotional disturbances in dementia disorders: a Nordic multicenter of divalproex sodium for agitation and dementia. Am J Geriatr
study. Br J Psychiatr. 1990;157:894–901. Psychiatr. 2001;9:58–66.
149. Nyth AL, Gottfries CG, Lyby K, et al. A controlled multicenter 172. Tariot PN, Erb R, Podgorski CA, et al. Efficacy and tolerability of
clinical study of citalopram and placebo in elderly depressed patients carbamazepine for agitation and aggression in dementia. Am J Psy-
with and without concomitant dementia. Acta Psychiatr Scand. 1992; chiatr. 1998;155:54–61.
86:138–145. 173. Teri L, Logsdon RG, Peskind E, et al. Treatment of agitation in AD:
150. Katona CLE, Hunter BN, Bray J. A double-blind comparison of the a randomized, placebo-controlled clinical trial. Neurology. 2000;55:
efficacy and safety of paroxetine and imipramine in the treatment of 1271–1278.
depression with dementia. Int J Geriatr Psychiatr. 1994;2:352–354. 174. Miller LJ. Gabapentin for treatment of behavioral and psychological
151. Rao V, Lyketsos CG. The benefits and risks of ECT for patients with symptoms of dementia. Ann Pharmacother. 2001;35:427–431.
primary dementia who also suffer from depression. Int J Geriatr 175. Desai AK, Grossberg GT. Buspirone in Alzheimer’s disease. Expert
Psychiatr. 2000;15:729–735. Rev Neurotherap. 2003;3:19–28.
152. Price TR, McAllister TW. Safety and efficacy of ECT in depressed 176. Nelson J, Chouinard G. Guidelines for the clinical use of benzo-
patients with dementia: a review of clinical experience. Convulsive diazepines: pharmacokinetics, dependency, rebound and withdrawal.
Ther. 1989;5:1–74. Can J Clin Pharmacol. 1999;6:69–83.
MANAGEMENT OF ALZHEIMER’S DISEASE 353

177. Stern RG, Duffelmeyer ME, Zemishlani Z, et al. The use of benzo- 200. Tariot PN, Podgorski CA, Blazina L, et al. Mental disorders in the
diazepines in the management of behavioral symptoms in dementia nursing home: another perspective. Am J Psychiatr. 1993;143:1446–
patients. Psychiatr Clin North Am. 1991;14:375–384. 1449.
178. Salzman C, Fisher J, Nobel K, et al. Cognitive improvement following 201. Mathews FE, Dening T. Prevalence of dementia in institutional care.
benzodiazepine discontinuation in elderly nursing home residents. Int Lancet. 2002;360:225–226.
J Geriatr Psychiatr. 1992;7:89–93. 202. Macdonald AJD, Carpenter I, Box O, et al. Dementia and use of
179. Satlin A. Sleep disorders in dementia. Psychiatr Ann. 1994;24:186– psychotropic medication in non-ÔElderly Mentally Infirm’ nursing
190. homes in South East England. Age Ageing. 2002;31:58–64.
180. Satlin A, Volicer L, Ross V, et al. Bright light treatment of behavioral 203. Avorn J, Gurwitz JH. Drug use in the nursing home. Arch Int Med.
and sleep disturbances in patients with Alzheimer’s disease. Am J 1995;123:195–204.
Psychiatr. 1992;149:1028–1032. 204. Rovner BW, Steel CD, Shmuely Y, Folstein MF. A randomized trial
181. Mishima K, Okawa M, Hishikawa Y, et al. Morning bright light of dementia care in nursing homes. J Am Geriatr Soc. 1996;44:7–13.
therapy for sleep and behavior disorders in elderly patients with 205. Judd S, Marshall M, Phippen P, eds. Design for Dementia. London:

Downloaded from biomedgerontology.oxfordjournals.org at The Australian National University on March 23, 2011
dementia. Acta Psychiatr Scand. 1994;89:1–7. Hawker; 1997.
182. Shaw SH, Curson H, Coquelin JP. A double-blind, comparative study 206. Horwitz GJ, Tariot PN, Mead K, Cox C. Discontinuation of anti-
of zolpidem and placebo in the treatment of insomnia in elderly psychotics in nursing home patients with dementia. Am J Geriatr
psychiatric inpatients. J Int Med Res. 1992;20:150–161; correction Psychiatr. 1995;3:290–299.
1992;20:494. 207. Banks MR, Banks WA. The effects of animal-assisted therapy
183. Knopman D, Schneider LS, Davis K, et al. Long-term tacrine on loneliness in an elderly population in long-term care facilities.
(Cognex) treatment effects on nursing home placement and mortality. J Gerontol Med Sci. 2002;57A:M428–M432.
The Tacrine Study Group. Neurology. 1996;47:166–177. 208. Coleman MT, Looney S, O’Brien J, Ziegler C, Pastorino CA, Turner
184. Lopez OL, Becker JT, Wisniewski S, et al. Cholinesterase inhibitors C. The Eden Alternative: findings after one year of implementation.
postpone nursing home placement for Alzheimer’s disease. J Neurol J Gerontol Med Sci. 2002;57A:M422–M427.
Neurosurg Psychiatr. 2002;72:310–314. 209. Morley JE, Flaherty JH. Putting the ‘‘home’’ back in nursing home.
185. Volicer L, Hurley AC. Comorbidity in Alzheimer’s disease. J Mental J Gerontol Med Sci. 2002;57A:M419–M421.
Health Aging. 1997;3:5–17. 210. Petersen RC, Stevens JC, Ganguli M, et al. Practice parameter: early
186. Leon J, Cheng CK, Neumann PJ. Alzheimer’s disease care: costs and detection of dementia: mild cognitive impairment (an evidence-based
potential savings. Health Aff (Millwood). 1998;17:206–216. review). Neurology. 2001;56:1133–1142.
187. Chandra V, Barucha NE, Schoenberg BS. Conditions associated with 211. Morris JC, Storandt M, Miller JP, et al. Mild cognitive impairment
Alzheimer’s disease at death: case-control study. Neurology. 1986;36: represents early-stage Alzheimer’s disease. Arch Neurol. 2001;58:
209–211. 397–405.
188. Hashimoto M, Imamura T, Tanimukai S, et al. Urinary incontinence; 212. Jhee SS, Shiovitz T, Crawford AW, Cutler NR. Beta-amyloid ther-
an unrecognized adverse effect of donepezil. Lancet. 2000;356:568. apies in Alzheimer’s disease. Expert Opin Investig Drugs. 2001;10:
189. Levkoff SE, Evans DA, Liptzin B, et al. Delirium: the occurrence and 593–605.
persistence of symptoms among elderly hospitalized patients. Arch 213. Morley JE, Farr SA, Flood JF. Antibody to amyloid beta protein
Intern Med. 1992;152:334–340. alleviates impaired acquisition, retention, and memory processing in
190. Tune L, Carr S, Hoag E, Cooper T. Anticholinergic effects of drugs SAMP8 mice. Neurobiol Learning Memory. 2002;78:125–138.
commonly prescribed for the elderly: potential means for assessing 214. Morley JE, Kumar VB, Bernardo AE, et al. Beta-amyloid precursor
risk of delirium. Am J Psychiatr. 1992;149:1393–1394. polypeptide in SAMP8 mice affects learning and memory. Peptides.
191. Beers MH. Explicit criteria for determining potentially inappropriate 2000;21:1761–1767.
medication use by the elderly. An update. Arch Intern Med. 1997;157: 215. Hock C, Konietzko U, Papassotiropoulos A, et al. Generation of anti-
1531–1536. bodies specific for beta-amyloid by vaccinating patients with Alzheimer
192. Mendez MF, Catanzaro P, Doss RC, et al. Seizures in Alzheimer’s disease. Nat Med. 2002;Oct 15.
disease: clinico-pathologic study. J Geriatr Psychiatr Neurol. 1994;7: 216. Kumar VB, Farr SA, Flood JF, et al. Site-directed antisense
230–233. oligonucleotide decreases the expression of amyloid precursor protein
193. Hauser WA, Morris ML, Heston LL, Anderson VE. Seizures and and reverses deficits in learning and memory in aged SAMP8 mice.
myoclonus in patients with Alzheimer’s disease. Neurology. 1986;36: Peptides. 2000;21:1769–1775.
1226–1230. 217. Verlinsky Y, Rechitsky S, Verlinsky O, et al. Preimplantation diag-
194. Jellinger KA. Alzheimer disease and cerebrovascular pathology: an nosis for early-onset Alzheimer disease caused by V717L mutation.
update. J Neural Transm. 2002;109:813–836. JAMA. 2002;287:1018–1021.
195. Torian LV, Davidson EJ, Sell L, et al. The effect of senile dementia on 218. Silverberg GD, Levinthal E, Sullivan EV, et al. Assessment of low-
acute medical care in a geriatric medicine unit. Int Psychogeriatr. flow CSF drainage as a treatment for AD. Neurology. 2002;59:1139–
1994;4:231–239. 1145.
196. Lyketsos CG, Sheppard JM, Rabins PV, et al. Dementia hospitaliza- 219. Reisberg B, Ferris SH, DeLeon MJ, Crook T. The Global Dete-
tion and psychiatry. Am J Psychiatr. 2000;157:704–707. rioration Scale for assessment of primary degenerative dementia. Am
197. Gillick MR, Serrell NA, Gillick LS. Adverse consequences of J Psychiatr. 1982;139:1136–1139.
hospitalization in the elderly. Soc Sci Med. 1982;16:1033–1038. 220. Folstein MF, Folstein SE, McHugh PR. Mini Mental State: a practical
198. Rabins P, Nicholson M. Acute psychiatric hospitalization for patients method for grading the cognitive state of patients for the clinician.
with irreversible dementia. Int J Geriatr Psychiatr. 1991;6:209–211. J Psychiatr Res. 1975;12:189–198.
199. Zubenko GS, Rosen J, Sweet RA, et al. Impact of psychiatric hos-
pitalization on behavioral complications of Alzheimer’s disease. Am J Received December 2, 2002
Psychiatr. 1992;149:1484–1491. Accepted December 17, 2002

Vous aimerez peut-être aussi