Sei sulla pagina 1di 15

REVIEW

THE PATHOPHYSIOLOGY OF ALZHEIMER’S DISEASE


AND DIRECTIONS IN TREATMENT

Ann S. Morrison, PhD, RN, CS,* and Constantine Lyketsos, MD, MHS†

ABSTRACT placement. Other treatments (vitamins E and/or


C, nonsteroidal anti-inflammatory drugs, and
The pathophysiology of Alzheimer’s disease statins [cholesterol-lowering drugs]) show some
(AD) is complex, involving several neurotransmit- promise in preventing or delaying AD progres-
ter systems and pathophysiologic processes. The 3 sion, although more data are needed. There are
hallmarks of AD—β-amyloid plaques, neurofibril- now several published guidelines for the treat-
lary tangles, and neuronal cell death—are well ment of AD. Although some may argue that the
known and central factors in AD pathology. These effect of these drugs is modest, any preservation
hallmarks, combined with our information on neu- of function and delay of nursing home placement
rotransmitter involvement, are specific to AD has important and permanent consequences for
based on the timing, sequence, and location of the patient and caregiver. The profound and
these changes. This article reviews the pathophys- sometimes rapid loss of memory and function
iology and course of brain damage seen during with AD underscores the importance of trying at
AD progression, from preclinical to severe stages. least 1 of these treatments in a patient with AD.
Understanding these processes is fundamental in (Adv Stud Nurs. 2005;3(8):256-270)
understanding AD progression and symptom
manifestation, and in assessing the potential risks
and benefits of therapeutic interventions. There
are now 5 approved drugs for the cognitive symp-
toms of AD—4 acetylcholinesterase inhibitors
(approved for mild to moderate AD) and 1 non- ALZHEIMER’S DISEASE PATHOPHYSIOLOGY
competitive glutamate receptor antagonist
(approved for moderate to severe AD). Recent CHANGES IN BRAIN STRUCTURE
studies with these drugs are focusing on longer- Alzheimer’s disease (AD) pathology can be charac-
term follow-up (1 year and beyond), in addition to terized on a macro level as the progressive loss of brain
more “clinically relevant” outcomes, such as tissue. As the disease progresses, neurons die in a par-
measures of function and nursing home ticular pattern over time. One of the earliest signs of
AD is memory loss, particularly short-term recall. The
*Clinical Nurse Specialist Coordinator, Department of brain areas involved in memory include the cortex,
Psychiatry, Alzheimer’s Disease Research Center, The Johns especially the hippocampus (Figure 1).1 Preclinical AD
Hopkins Hospital, Baltimore, Maryland.
†Professor of Psychiatry and Behavioral Sciences, begins in the entorhinal cortex, which connects the
Codirector, Division of Geriatric Psychiatry and hippocampus, the structure responsible for memory
Neuropsychiatry, The Johns Hopkins Hospital, Baltimore, formation (short- and long-term memory; Figure 2),
Maryland.
Address correspondence to: Ann S. Morrison, PhD, RN,
to the cerebral cortex.1 Several studies, such as ones
CS, Clinical Nurse Specialist Coordinator, Department of including magnetic resonance imaging, suggest that
Psychiatry, Alzheimer’s Disease Research Center, The Johns neuronal loss (measured by atrophy in select regions)
Hopkins Hospital, 550 Broadway Avenue, Baltimore, MD may start years before signs of memory loss emerge.2,3
21205. E-mail: amorris7@jhmi.edu.

256 Vol. 3, No. 8 n October 2005


REVIEW

Figure 1. Major Areas of the Brain Figure 2. Brain Changes in Preclinical Alzheimer’s
Disease
Corpus Callosum Cerebral Cortex
Cerebral
Cortex
Thalamus

Hypothalamus
Cerebellum
Hippocampus
Brain Stem

Hippocampus Entorhinal
Cortex
Side view of the brain Reprinted with permission from Alzheimer’s Disease Education and Referral
Center Web site. Available at: http://www.alzheimers.org/unraveling/04.htm.
Accessed June 17, 2005.1
Copyright ©2002, Christy Krames.

As the brain atrophies, cerebrospinal fluid fills in the


space previously occupied by brain tissue. Figure 3. Brain Changes in Mild to Moderate
In mild to moderate AD, patients experience more Alzheimer’s Disease
prominent memory loss (eg, difficulty recalling well-
known names and confusion about familiar places), a Moderately
Enlarged
decline in the ability to process complex thoughts (eg, Cortical Ventricles
Shrinkage
difficulty with balancing the checkbook or preparing a
meal), and mood and personality changes. In the
brain, atrophy extends to other areas of the cerebral
cortex (Figure 3).1
At the later stages of AD progression, the cortex has
atrophied in areas that control speech, reasoning, senso- Shrinkage of
Hippocampus
ry processing, and conscious thought (Figure 4).1,4 As
Reprinted with permission from Alzheimer’s Disease Education and Referral
expected with this degree of brain atrophy, the symp- Center Web site. Available at: http://www.alzheimers.org/unraveling/04.htm.
toms of severe AD increase in severity (ie, impaired long- Accessed June 17, 2005.1
term memory, seizures, incontinence, weight loss, no
recognition of loved ones, inability to sit up, and groan-
ing/moaning/grunting). The progression of atrophy Figure 4. Brain Changes in Severe Alzheimer’s
throughout the brain is diagrammed in Figure 5.1 Disease
DEGENERATIVE PROCESSES IN ALZHEIMER’S DISEASE
Extreme Shrinkage of
On a micro level, AD is characterized by 3 neu- Cerebral Cortex Severely
ropathologic hallmarks: extracellular plaques of β-amy- Enlarged
Ventricles
loid protein (amyloid plaques), intracellular
neurofibrillary tangles (NFTs), and neuronal degenera-
tion. Plaques and NFTs were first discovered by Alois
Alzheimer in a 1906 autopsy of a demented patient.
Although they are a defining component of AD, they
Extreme
are not unique to AD. Plaques and NFTs occur with Shrinkage of
Hippocampus
normal aging and in some other neurodegenerative dis-
orders. In AD, plaques and NFTs are localized to areas Reprinted with permission from Alzheimer’s Disease Education and Referral
Center Web site. Available at: http://www.alzheimers.org/unraveling/04.htm.
in the brain that correspond to clinical symptoms. Accessed June 17, 2005.1

Advanced Studies in Nursing n 257


REVIEW

develop AD at a younger age than the general popula-


Figure 5. Brain Damage and Atrophy During Alzheimer’s
tion.6 PS1 and PS2 are genes that code for presenilin,
Disease Progression
the catalytic subunit of γ-secretase. There are at least
20 known mutations of APP, more than 140 known
A B C mutations in PS1, and approximately 10 in PS2.6,7
Immunization against Aβ is under investigation as a
treatment in animals and humans.8-11
Neurofibrillary tangles are seen as dying or dead
neurons when viewed after histologic staining. NFTs
A, preclinical Alzheimer’s disease; B, mild to moderate Alzheimer’s disease; and result from the destruction of neuronal microtubules
C, severe Alzheimer’s disease. caused by the modification of their supporting pro-
Reprinted with permission from Alzheimer’s Disease Education and Referral Center
Web site. Available at: http://www.alzheimers.org/unraveling/04.htm. Accessed June tein, tau (Figure 9).1 Microtubules are essential com-
17, 2005.1 ponents of neuronal cell structure; they act as tracks
along which nutrients are delivered and neuronal
transmission is propagated in the neuronal axon.
During AD pathogenesis, tau proteins become hyper-
β-amyloid plaques are thought to play the central phosphorylated, disrupting their bonds to micro-
role in AD pathogenesis (the “amyloid hypothesis”), tubules, thus collapsing microtubule structure and
with several diverse pathophysiologic processes that
follow (Figure 6), described as an “amyloid cascade.”5
The AD pathophysiologic cascade is complicated, but
each discovery of a neurotransmitter or pathologic
process leads to the potential discovery of new thera-
peutic targets. Figure 6. Diagram of the Amyloid Cascade

The β-amyloid hypothesis Cytoplasm


β-amyloid plaques are clumps of insoluble peptides
that result from the abnormal cleavage of amyloid pre- γ-secretase γ-secretase

cursor protein (APP), the exact function of which is Cell


membrane p3 α-secretase
β-secretase Aβ
unknown. APP is normally cleaved by 3 enzymes—β- APP

secretase, γ-secretase, and α-secretase. Cleavage by β- Extracellular


space α-APP
β-APP

secretase, followed by γ-secretase, yields a soluble 40 Aβ generation

amino acid peptide (Figure 7).1 In AD, a variant form


of the γ-secretase cleaves APP at an incorrect place,
Oxidation Excitotoxicity Aβ aggregation Inflammation Tau hyperphosphorylation
creating a 42 amino acid peptide called Aβ42 or Aβ,
which is not soluble and aggregates into identifiable
clumps termed β-amyloid plaques (Figure 8).1 α-secre-
tase actually serves a protective function as it cleaves Neuron
APP at a site that prevents Aβ formation. Senile plaque with Neurofibrillary
Genetic studies of AD support the role of γ-amy- microglial activation tangles

loid plaques and β-secretase in AD pathogenesis.


Three genes have been identified in familial AD (APP,
PS1 [presenilin 1], and PS2) and all are known to be Cell Cognitive
involved with the formation of Aβ. (The use of italics death and behavioral
abnormalities
indicates a gene, whereas normal font indicates the Neuro-
transmitter
protein made from the gene.) APP is the gene that deficit

codes for APP (the protein) and is located on chromo-


some 21. Interestingly, people with Down’s syndrome APP = amyloid precursor protein.
(trisomy 21—3 copies of chromosome 21) ultimately Reprinted with permission from Cummings. N Engl J Med. 2004;351:56-67.5

258 Vol. 3, No. 8 n October 2005


REVIEW

destroying the neuron’s transport and communication


Figure 7. The Structure of Amyloid Precursor
system. Neuronal cell death ensues. Although the
Protein Relative to the Cell Membrane
causal relationship is unclear, hyperphosphorylation of
tau is thought to occur after plaque formation.12,13

The cholinergic hypothesis β-amyloid


Loss of cholinergic neurons is another well-estab-
lished pathology of AD. By late-stage AD, the number
of cholinergic neurons is dramatically reduced; in
some parts of the brain there is more than 75% loss.14
Acetylcholine is an important neurotransmitter in Enzymes
brain regions involving memory, and loss of choliner-
gic activity correlates with some aspects of cognitive
impairment. Cholinergic abnormalities are the most Amyloid precursor protein is a transmembrane protein. Cleavage by β-sec-
retase, followed by γ-secretase, yields a soluble 40 amino acid peptide. This
prominent neurotransmitter changes in AD. peptide is shown here in green.
Acetylcholine binds to 2 postsynaptic receptor Copyright ©2002, Christy Krames.
types: muscarinic and nicotinic. Presynaptic nicotinic
receptors influence the release of neurotransmitters
Figure 8. Insoluble Aggregates of Aβ42
important for memory and mood—acetylcholine, glu-
tamate, serotonin, and norepinephrine—all of which
have been implicated in AD pathology.
β-amyloid
Plaque
The glutamatergic/excitotoxicity hypothesis
Glutamate is the primary excitatory neurotrans-
mitter in the brain. In the central nervous system, it is
virtually ubiquitous and is estimated to be involved in
roughly 66% of all brain synapses.15,16 Glutamatergic
neurons are also critical because they form projections
to other areas of the brain, including cholinergic neu-
rons, thus influencing cognition. In AD, the patholo-
gy associated with glutamatergic neurons is not with Copyright ©2002, Christy Krames.
glutamate itself but with the levels of pre- and postsy-
naptic glutamate receptors. Of the 3 types of postsy-
naptic glutamate receptors, AD pathology has only
been linked to 1 type, the N-methyl-D-aspartate Figure 9. The Role of Tau Protein in Neuronal Cell
(NMDA) receptor, which appears to undergo sus- Death
tained low-level activation in AD brains, causing sus-
tained low-level neurotransmission. This dysregulation
at the glutamate NMDA receptor is thought to per- Stabilizing
Tau Molecules
petuate a vicious cycle of neuronal damage, in which
Healthy Neuron
continuous activation of the receptor leads to chronic
calcium influx within the neuron that interferes with
Microtubules
normal signal transduction.17 It also leads to increased Disintegrating
Microtubule Subunits
Fall Apart
production of APP, which, as noted earlier in this arti- Diseased Neuron
Microtubule
Tangled Clumps
of Tau Proteins
cle, is associated with higher rates of plaque develop-
ment and hyperphosphorylation of tau protein (thus
NFT formation) followed by neuronal toxicity.18-21 Disintegrating
Microtubules
Glutamate is cleared from the synapse by reuptake into
the nerve terminal and surrounding glial cells. Copyright ©2002, Christy Krames.

Advanced Studies in Nursing n 259


REVIEW

Dementia patients also appear to have reduced num- nases COX-1 and COX-2, in the AD brain.28 It is inter-
bers of glutamate reuptake sites or abnormal expres- esting to note that the inflammation in AD is chronic
sion of a glutamate transporter, which may be linked and localized to discrete areas of the brain, as are the
to NFT formation.22,23 other types of cellular damage with AD.29

The oxidative stress hypothesis Other neurotransmitter deficiencies


In AD brains, Aβ induces lipid peroxidation and During the AD process, brain regions in which
generates reactive oxygen and nitrogen species; these acetylcholine, serotonin, and norepinephrine are
are oxygen or nitrogen molecules containing an prominent become altered, affecting widespread
unpaired extra electron (termed species) that reacts areas of the cerebral cortex. Serotonin is known to be
with other molecules to achieve a stable configuration. important in affective illness (eg, depression and anx-
During this process, the reactive species forms a mole- iety), and depression is a common comorbidity with
cular bond with another molecule, while a high-ener- AD. The number of serotonin receptors and trans-
gy electron (termed a “free radical”) is thrown off. The porters are altered in AD brains, corresponding to
reaction is permanent, thus structurally and function- decline in measures of cognition and presence of anx-
ally altering the molecule to which the reactive species iety.30-32 Also, norepinephrine levels are reduced and
is attached. The free radical is left to cause cellular and norepinephrine neurons are lost in AD.33
molecular damage. This oxidative damage can occur in Norepinephrine may play an important role in some
virtually all types of neuronal macromolecules (eg, of the behavioral and psychological symptoms of
lipids, carbohydrates, proteins, and nucleic acids). The dementia (aggression, agitation, and psychosis), in
brain is especially vulnerable to damage from oxidative addition to memory loss.34
stress because of its high oxygen consumption rate,
abundant lipid content, and relative paucity of antiox- Cholesterol
idant enzymes compared to other organs. In neurons, Cholesterol is also now thought to be involved in
oxidation can result in numerous problems, including AD pathogenesis. The brain contains the highest
upregulation of proinflammatory cytokines and amount of cholesterol of any human organ.35
irreparable DNA damage.24,25 Oxidative stress is Elevated cholesterol levels appear to increase Aβ pro-
thought to be important early in AD progression duction, whereas reduced cholesterol synthesis (eg, as
because it is temporally linked to the development of a result of statin drugs) reduces Aβ levels, thus the
plaques and NFTs.26 risk of AD progression.36 Cholesterol reduction may
also reduce the risk or severity of dementia through
The chronic inflammation hypothesis protection from vascular risk factors; vascular demen-
β-amyloid deposits, NFTs, and damaged neurons tia often coexists in patients with AD. In the
may stimulate inflammation as a natural response to Honolulu-Asia Aging Study, cholesterol levels of 218
cell damage. Microglia (1 of 4 types of brain cells, Japanese-American men were measured at mid-life
including neurons, astrocytes, and oligodendrocytes) and late-life. At autopsy, there was a strong linear
are involved in immune and inflammatory responses association between mid-life and late-life high-densi-
to injury or infection within the brain. During AD ty lipoprotein cholesterol and the number of plaques
pathogenesis, microglia are activated to release poten- and NFTs in the cortex.37
tially cytotoxic molecules, such as proinflammatory
cytokines, reactive oxygen species, proteinases, and SUMMARY
complement proteins.27 This is a normal response to Our understanding of AD pathology is now
cellular damage that, in AD, appears to proceed unin- extending beyond the details of pathophysiologic
hibited, causing more harm than protection. Cytokines processes to a broader view of the sequence and tim-
stimulate inflammatory processes that may promote ing of these events in the development of symptoms
apoptosis (programmed cell death) of neurons and oligo- and illness progression. Aβ generation is considered
dendrocytes and induce myelin damage. Inflammation to be the first step of an “amyloid cascade,” which
as part of AD pathology has been observed as increased continues with oxidation, excitotoxicity, inflamma-
prostaglandins, which are produced by the cyclooxyge- tion, and the hyperphosphorylation of tau. Each of

260 Vol. 3, No. 8 n October 2005


REVIEW

these processes contributes to neurotransmitter TREATMENTS FOR ALZHEIMER’S DISEASE


abnormalities and neuronal cell death, which then
manifest as cognitive and behavioral abnormalities.5 OVERVIEW
Pharmacologic treatments for the primary symp-
KEY POINTS toms of AD, namely memory loss and cognitive
• In AD, brain atrophy and neuronal loss occur in impairment, have only become available within the
a predictable pattern beginning with the entorhi- past decade. Four of 5 approved therapies (tacrine,
nal cortex and hippocampus, ultimately progress- donepezil, rivastigmine, and galantamine) act by
ing throughout the brain. inhibiting the enzyme acetylcholinesterase, which is
• AD is characterized by 3 neuropathologic hall- responsible for breaking down acetylcholine postsy-
marks: plaques of β-amyloid protein (amyloid naptically, thus keeping acetylcholine in the synapse
plaques), NFTs, and neuronal degeneration, all longer. As described earlier in this article, cholinergic
of which occur during normal aging, but in AD neuron loss is the primary type of neurodegeneration
occur in specific locations in the brain. in AD. The fifth drug, memantine, acts as a noncom-
• β-amyloid plaques are thought to play the central petitive antagonist to the NMDA receptor, curtailing
role in AD pathogenesis, described as the “amy- the low-level but steady NMDA receptor activation
loid cascade.” thought to occur in AD and contributing to excito-
• The β-amyloid hypothesis maintains that forma- toxicity.17 Several other treatments that address not
tion of β-amyloid plaques is the first step in AD only the symptoms but also the pathophysiologic
pathology, and genetic and histopathologic stud- processes in AD are also under investigation.
ies support this hypothesis. NFTs occur after
plaque formation. CHOLINESTERASE INHIBITORS
• The cholinergic hypothesis is supported by the Although there are 4 approved cholinesterase
drastic and widespread loss of cholinergic neu- inhibitors, only 3 are commonly used in the United
rons in AD, the benefits with acetylcholinesterase States. The fourth, tacrine, is still available but rarely
inhibitors, and the role of the nicotinic choliner- prescribed because of the potential hepatotoxicity
gic receptor. associated with its use; it also has a complicated titra-
• The glutamate/excitotoxicity hypothesis main- tion schedule. Tacrine should probably never be rec-
tains that slow but steady activation of a gluta- ommended as first-line therapy for AD. Although it is
mate receptor leads to cellular damage. still on the market, there is no practical use for tacrine
• The oxidative stress hypothesis suggests that β- in the treatment of AD. However, nurses may still
amyloid induces oxidative stress causing perma- encounter prescriptions for it. A summary of the rec-
nent damage to some neuronal macromolecules ommended dosing and titration schedule, in addition
(especially lipids) and creating reactive species, to common adverse events with each drug, are listed in
which further propagate neuronal toxicity. Table 1.38-41 Donepezil, rivastigmine, and galantamine
• The chronic inflammation observed with AD is are indicated for mild to moderate AD; memantine is
thought to be the result of all of these mecha- indicated for moderate to severe AD.
nisms causing cell damage and death. In AD, Donepezil was the first drug to be approved by the
inflammatory cytokines respond to these process- US Food and Drug Administration (FDA) for the
es and contribute to neuronal destruction symptoms of mild to moderate AD. Its efficacy was
because they appear to act uninhibited. established in 2 randomized, double-blind, placebo-
• Serotonin and norepinephrine are also involved controlled studies.42,43 Donepezil was followed by
in AD pathology. Serotonin is an important fac- rivastigmine, the clinical efficacy of which was shown
tor in depression and anxiety, both of which are in 3 randomized, placebo-controlled studies in the
common in patients with AD. United States and internationally.44-46 The efficacy of
• Cholesterol also appears to have a role in AD galantamine was demonstrated in 4 randomized, dou-
progression, perhaps through a reduction in vas- ble-blind, placebo-controlled studies—2 in the United
cular risk factors, which are common in patients States and 2 in other countries.47-50 The initial safety
with AD. and efficacy studies were typically shorter than 6

Advanced Studies in Nursing n 261


REVIEW

Table 1. The Currently Available Treatments for AD Symptoms—A Summary*

Drug AD Indication Dosing Common Adverse Events†

Donepezil Mild to moderate 5 mg or 10 mg; treatment with a 10-mg dose Nausea, diarrhea, insomnia, vomiting, muscle
should not be considered until patients have been cramps, fatigue, and anorexia
on a daily dose of 5 mg for 4 to 6 weeks to avoid
adverse events
Rivastigmine Mild to moderate 3–6 mg twice daily; starting dose: 1.5 mg twice daily; Nausea, vomiting, loss of appetite, dyspepsia,
increase to 3 mg twice daily after 2 weeks; other asthenia, and weight loss
dose increases should be separated by at least 2 weeks
Galantamine Mild to moderate 8–12 mg twice daily, preferably with morning and Nausea and vomiting
evening meals; 24 mg/d may provide additional
benefit for some patients, but it is associated with
more adverse events; starting dose: 4 mg twice daily;
increase to initial maintenance dose after at least 4 weeks
Memantine Moderate to severe Target dose: 20 mg/day; starting dose: 5 mg/day; Confusion, dizziness, headache, and constipation
titration: increase in 5-mg increments to 10 mg/d (5 mg
twice daily), 15 mg/d (5 mg and 10 mg as separate
doses), 20 mg/d (10 mg twice daily); minimum
recommended interval between dose increases is
1 week; can be taken with or without food
*Tacrine continues to remain on the market, but it is rarely used because of its potential hepatotoxicity and other adverse events.
†Based on clinical studies.
AD = Alzheimer’s disease.
Data from Donepezil US prescribing information38; Galantamine US prescribing information39; Memantine US prescribing information40; Rivastigmine US prescribing information.41

months. Current studies of AD drugs are focusing on sus 0.82 ± 0.71 (rivastigmine) and -8.0 (historical place-
longer follow-up, other AD-related indications (eg, bo) versus -3.8 (rivastigmine).52,53 In the 2-year study, cog-
mild cognitive impairment [MCI] and more severe nitive decline slowed by 35% in those patients receiving
stages of AD), nursing home placement, vascular risk rivastigmine compared to a historical placebo group.54
factors, and the pathophysiology of AD. Improvement with rivastigmine was also observed in a
subgroup of moderately severe patients.55 The long-term
Long-term follow-up efficacy of galantamine was shown in 3 studies. In an
There has been 1 long-term study of donepezil that 18.5-month study, patients taking galantamine enjoyed
extended to 4.9 years, an extension of an original 14- sustained cognitive benefits during the entire study dura-
week study.42 The results showed evidence of clinical tion.56 In two 3-year studies, galantamine was associated
benefit (ie, maintenance of baseline scores for cogni- with a roughly 50% slower cognitive decline compared to
tion measures) during the first 6 to 9 months of open- predicted rates of decline in a clinically similar historical
label treatment, with gradual cognitive decline after 9 control sample of patients with AD.57,58
months. The treatment group was compared to histor-
ical controls, which was a serious methodological Disease severity
problem for this study. The rate of cognitive decline Donepezil has shown significant benefit on mea-
throughout the nearly 5-year study was slower for sures of cognition compared to placebo in patients
those patients receiving donepezil than the estimates with “early stage” (mild) AD.59 An important observa-
for patients who had not been treated.51 The benefits tion in this study population is that the placebo group
with rivastigmine have been shown in studies of 1- and showed virtually no signs of cognitive decline (as mea-
2-years’ duration. In the 1-year studies, differences in sured by the Mini-Mental State Examination
cognitive decline as measured by the AD assessment [MMSE]) during the study period, whereas in studies
scale-cognitive subscale were -4.45 ± 0.8 (placebo) ver- of patients with mild to moderate AD, patients receiv-

262 Vol. 3, No. 8 n October 2005


REVIEW

ing placebo showed cognitive decline in MMSE scores donepezil tended to stabilize over time, rather than to
during the study.59 Therefore, the extent of observed improve over baseline, “suggesting that clinicians
donepezil benefit may depend on the level of AD should aim for stability rather than actual improve-
severity. At the other end of the severity spectrum, ment in ADLs.”64 This is an important message to con-
donepezil was compared to placebo in patients with vey to patients and their caregivers as AD progresses
moderate to severe AD and showed significant benefit and they consider treatment with a cholinesterase
in cognition measures at all time points during the 6- inhibitor. Another study showed not only slower
month study.60 decline in ADLs and IADLs with donepezil treatment
Two studies have analyzed the effect of rivastigmine but also less decline in the components required for
based on disease severity, but from a different approach. completing each ADL (initiation, planning and orga-
In a 1-year study, patients were classified as slowly pro- nization, and effective performance).65 Caregiver bur-
gressive or rapidly progressive, based on the magnitude den was also measured as time required with the
of response seen in the first 6 months of the study. The patient for ADLs/IADLs and stress levels. Caregivers
final results showed notable and significant differences in reported spending a mean of 52.4 fewer minutes/day
response to rivastigmine between these 2 groups of on ADLs and 17.2 fewer minutes/day on IADLs with
patients, with patients who were rapidly progressive donepezil-treated patients. Almost all components of
obtaining greater benefit from rivastigmine treatment.61 the caregiver stress scale favored donepezil. The
In a 12-month study of patients with mild AD or MCI, authors also noted the importance of stabilizing cogni-
cognitive function was slightly improved or maintained tive function because the placebo group did not show
in the patients with mild AD and unchanged or slightly a floor effect (ie, patients never reached a point at
worsened in the patients with MCI (both groups receiv- which further decline was not measurable).65
ing rivastigmine); cognitive function was markedly wors- Rivastigmine has beneficial effects on ADLs, as
ened in untreated patients with AD.62 shown in a retrospective analysis of 3 studies.66 Also,
ADL impairment differed across the stages of AD.
Functional measures Table 2 lists the ADLs with a significant change from
Researchers are now turning to more “clinically rel- baseline based on 3 levels of severity.66
evant” outcomes, such as functional decline (ie, ability A secondary analysis of a 5-month study showed that
to perform activities of daily living [ADLs] or instru- galantamine treatment resulted in little or no decline
mental ADLs [IADLs]), nursing home placement, and from baseline in ADL measures compared to significant
effect on caregiver burden. ADLs focus on basic func- decline in the placebo group.48,67 Galantamine treatment
tioning, such as toileting, feeding, dressing, bathing, resulted in maintenance or improvement in ADL and
walking, and grooming. IADLs are more complex IADL measures, with significant improvement over
activities, which most adults of normal mental capaci- placebo in 3 of 6 basic ADLs (toileting, bathing, and
ty can perform, such as operating household appli- grooming) and 6 of 17 IADLs (conversation, managing
ances, paying bills and managing money, shopping, personal belongings, current events, writing, hobbies,
preparing meals, and participation in hobbies. The and operating household appliances).67 ADL stabiliza-
effect on ADLs is important for several reasons. ADLs tion was seen throughout the range of AD severity, but
instill and maintain a sense of independence and self- the greatest differences were observed in those patients
efficiency, which is critical for patients’ and caregivers’ with severe disease.67
quality of life. In AD, loss of ADLs is enduring and is Regarding caregiver time, galantamine also has
a significant predictor of institutionalization.63 notable effects. Pooled data from two 6-month studies
Therefore, preservation of ADLs is a critical outcome show that caregivers reported spending a mean of 32
with important and permanent ramifications. fewer minutes per day with galantamine-treated
A 1-year study of donepezil versus placebo suggest- patients than with placebo-treated patients, a savings
ed that donepezil use may have effects on delaying loss of approximately 3.5 hours per week. In patients with
of ADLs. Patients taking donepezil were 31% less like- moderate AD, the benefits were even greater—a mean
ly to experience functional decline (ie, losing the abil- daily time savings of 53 minutes or more than 6 hours
ity to perform an ADL or IADL) than those taking per week. Similarly, galantamine-treated patients could
placebo.64 The authors noted that patients on spend 27 more minutes per day unsupervised and, in the

Advanced Studies in Nursing n 263


REVIEW

moderate AD group, the benefit was extended to spend- Jakimovich, in which donepezil was administered to a
ing 68 more minutes per day unsupervised.68 patient with severe AD, behavioral problems, and sev-
The profound consequences of maintaining ADLs eral comorbidities following admission to a long-term
is illustrated in a case study from Tariot and care facility.69 The positive effects of donepezil treat-

Table 2. ADLs Affected Based on Degree of AD Severity with Rivastigmine or Placebo Treatment

GDS ≤3 GDS = 4 GDS ≥5


PDS Item Mean change Improvement, % Mean change Improvement, % Mean change Improvement, %

Cannot handle money -0.1 vs -4.1* 49 vs 33*


Cannot tell time 0.5 vs -2.2 52 vs 40
Dials telephone numbers 2.1 vs -2.1 56 vs 44
Increased time doing hobbies/pursuits 2.4 vs -1.0 62 vs 43
Takes normal precautions 1.0 vs -1.7 56 vs 40
Unable to drive alone 1.3 vs -3.5 55 vs 42
Has a clear concept of time 0.1 vs -3.7 53 vs 40
Discusses politics -0.1 vs -3.3 51 vs 38
Dresses properly -0.5 vs -4.1 50 vs 39 -1.6 vs -6.9 45 vs 32
Drives car safely -1.9 vs -5.4 44 vs 33
Forgets things 0.9 vs -3.3* 52 vs 32* -1.5 vs -5.3* 40 vs 28*
Rearranges objects 0.7 vs -2.3 51 vs 38 -1.1 vs -5.8* 45 vs 30*
Stops participation in family finances -1.4 vs -4.7 45 vs 32
Cannot use telephone -3.1 vs -7.6* 42 vs 30*
Confusion in different settings -2.3 vs -6.5* 42 vs 28*
Good eating manners -2.5 vs -6.7* 42 vs 28*
Makes mistakes doing hobbies/pursuits -1.1 vs -6.8* 44 vs 27*
Does hobbies/pursuits
Stops household chores
Walks/travels alone
Tells time
Uses tools properly
Cannot discuss family finances
Walks safely
Cannot accurately care for finances
Stops social attendance
Drives own car
Works at his/her job
Meaningfully discusses TV, movies, and
similar events with spouse, family, or caregiver

This table shows the ADLs as listed in the PDS that were significantly changed from baseline during the study in the observed cases dataset. AD severity was measured
by the GDS and divided into 3 groups. As the GDS score increases, the severity of AD increases.
*Indicates P <.05 for rivastigmine versus placebo in the intent-to-treat dataset.
AD = Alzheimer’s disease; ADL = activity of daily living; GDS = Global Deterioration Scale; PDS = Progressive Deterioration Scale.
Reprinted with permission from Potkin et al. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26:713-720.66

264 Vol. 3, No. 8 n October 2005


REVIEW

ment included engagement in meaningful conversa- behavior. If no change is observed, a different drug in
tions about family events and current events seen on this class can be tried.
television, although he was amnesic for details; coher-
ent periods of (factually correct) reminiscence on his Head-to-head comparisons
life; and improved ability to perform ADLs, including Direct comparisons of cholinesterase inhibitors are
independent toileting, reduced belligerent and sexual- rare in the published literature. A comparison of galan-
ly inappropriate behavior, discontinuation of haloperi- tamine to donepezil revealed no significant differences
dol, ability to travel to a family event, and restoration between the 2 treatment groups at study end (52
of walking ability; none of which were possible when weeks) in measures of ADLs and neuropsychiatric
he was admitted to the nursing facility.69 symptoms.72 However, cognition measures showed a
greater benefit with galantamine treatment, maintain-
Nursing home placement ing baseline levels with galantamine but worsening
Nursing home placement is another important mile- with donepezil.72
stone in AD progression. The desire and decision to
place patients with AD in full-time nursing care varies Cholinesterase inhibitors and anticholinergic drugs
with each patient (based on severity of disease and neu- Anticholinergic drugs are part of many therapeutic
ropsychiatric symptoms) and the caregiver’s circum- drug classes and are commonly prescribed. They are
stances, including financial resources and caregiver notorious for producing undesirable cognitive effects,
ability. In general, families probably prefer to keep the to which patients with AD are especially sensitive.
patient at home as long as possible. Conversely, some Their concurrent use is likely to negate any of the cog-
caregivers would prefer to place a patient under full- nitive benefit with cholinesterase inhibition. A cross-
time professional care, but cannot afford to do so. If the sectional study of Iowa Medicaid beneficiaries aged 50
patient is a widow/er and the adult children live far years and older indicated that a surprising 35.4% of
away, the preference may be for earlier placement these patients received concurrent anticholinergic
because geographic constraints prohibit at-home care. agents with cholinesterase inhibitors. Roughly 33% of
Studies of cholinesterase inhibitors’ effect on nursing patients received an anticholinergic agent within 90
home placement have yielded mixed results, possibly days (before or after) of starting the cholinesterase
because of these varying influential factors. inhibitor.73 Nurses should evaluate their patients’ drug
One study of donepezil versus placebo estimated profiles at each visit, especially when a cholinesterase
(conservatively) that proper use of donepezil over 9 to inhibitor is prescribed, to avoid this unfortunate situ-
12 months (ie, therapeutic doses and high compliance ation. Negating the effects of cholinesterase inhibitors
rates) resulted in a delay of nursing home placement of incurs significant burden and cost to the patient and
21.4 months for a first-time dementia-related place- the healthcare industry.
ment and 17.5 months for permanent placement.70
The authors note that “doctors and caregivers need to NMDA ANTAGONISTS
be educated that, in the same way as the actual bene- Memantine is the only NMDA antagonist
fits of treating hypertension or hyperlipidemia are seen approved for the treatment of AD. Its efficacy was
only after years of treatment, treatment of AD with demonstrated in 2 randomized, double-blind, place-
donepezil needs to be maintained to see important bo-controlled studies in the United States.74,75 In addi-
long-term benefits.”70 A study in the United Kingdom tion to those studies, memantine was compared to
of donepezil showed no difference in institutionaliza- placebo in patients already receiving stable doses of
tion after 3 years of follow-up, but the decision to donepezil and showed an added benefit when coad-
place patients in nursing homes or even to treat with a ministered with this cholinesterase inhibitor, with
cholinesterase inhibitor was based on different factors regard to measures of cognition, ADLs, global out-
from those in the US healthcare system.71 In clinical come, and behavior.75 An oral solution of memantine
practice, a typical drug trial for a cholinesterase was recently approved by the FDA, which offers an
inhibitor would be approximately 4 months (for titra- alternative that may make administration easier for
tion up to the therapeutic and best-tolerated dose) to patients who have trouble swallowing tablets or prefer
see some stabilization in memory, function, and/or taking medication in liquid form.

Advanced Studies in Nursing n 265


REVIEW

In all of the studies of AD drugs, it is important to of vitamin E for treatment of AD at a recommended


remember that the results are a summation of effect on dose of 2000 IU, except in patients with vitamin K
the entire study group. Individual patients respond to a deficiency (in whom this large dose may exacerbate
drug to varying degrees, with some patients improving coagulation defects).
markedly and others showing mild or even no improve-
ment. The only way to know whether a particular patient ANTI-INFLAMMATORY AGENTS
will respond to any of these drugs is to try. A drug trial is Over the past 5 to 10 years, several studies have sug-
worth the cost because the loss in ADLs and memory and gested that anti-inflammatory drugs (nonsteroidal anti-
effects on behavior as a result of AD are devastating. inflammatory drugs [NSAIDs]) are associated with a
decreased incidence of AD. A 2003 meta-analysis of the
VITAMINS E AND C existing literature (using 9 studies) showed that the rel-
Vitamin E is found in all cell membranes and helps ative risk of AD among users of NSAIDs was 28%
to maintain cell membrane structure. Vitamin E also lower than among nonusers of NSAIDs.81 Among
acts as an antioxidant by neutralizing the effects of short-term users (ie, <1 month), the risk was 5% lower,
oxygen free radicals. Vitamin E is able to donate a increasing to 17% reduced risk among intermediate
hydrogen atom, thus stabilizing the reactive species users (<24 months) and further still to 73% lower risk
with the unpaired electron. Vitamin C may be among long-term users (mostly >24 months).
involved in “recycling” the vitamin E from its oxidized Therefore, the benefits with NSAID use may increase
form for further use and is also a known antioxidant.76 with longer use. In studies of aspirin as the only
The recommended daily allowance of vitamin E is NSAID, the relative risk of AD was reduced 13%.81
22 IU for adults, which can typically be obtained A 2004 meta-analysis of 11 studies assessing the
through the diet.76 As recently reviewed, the few studies benefits of exposure to nonaspirin NSAIDs showed
evaluating vitamin E and/or C for prevention of AD that the risk for development of AD was cut 50% with
(through diet or nutritional supplements) do not pro- NSAID exposure.82 Among the prospective studies,
vide any clear direction, although there is a lot of inter- the risk was reduced 26% for lifetime NSAID expo-
est in their use. Some studies show a benefit with 1 or sure and 58% for use of at least 2 years.82 Therefore,
both vitamins, whereas other studies show no effect.76,77 NSAID use may have a preventive benefit, but the
In a recent study in patients with MCI, the effects of appropriate timing of exposure remains to be studied.
2000 IU vitamin E, 10 mg donepezil, and placebo were Studies of prednisone and a comparison of rofe-
compared over 3 years of treatment. There was no sig- coxib (a COX-2 inhibitor) and naproxen (an NSAID)
nificant difference in the rate of progression to AD showed no benefit in cognition measures in patients
among the 3 groups, although donepezil showed some with AD.83,84 The benefits of NSAIDs vary and do not
benefit during the first 12 months and in apolipopro- seem to extend after the onset of dementia. Therefore,
tein E (APOE)-«4 allele carriers.78 NSAIDs are currently not considered to be an appro-
It is difficult to identify the most appropriate role priate treatment for dementia.
of vitamin E for AD. Variations in study design and
source of the vitamins (diet, daily supplement, and STATINS
multivitamin) make an overall interpretation of vita- Cholesterol is synthesized only in the liver and brain.
min E/C efficacy difficult.76,77 Another confounding One of the enzymes involved in cholesterol synthesis is
factor is whether the role of vitamins E and/or C inhibited by the class of drugs used to treat dyslipi-
should be used as treatment with AD diagnosis or as a demia—statins. In the brain, cholesterol is synthesized
preventive measure. The recent meta-analysis regard- primarily in astrocytes and is transported elsewhere
ing high-dose vitamin E supplementation and risk of through apolipoproteins, including APOE (a gene in
all-cause mortality and the strong response from the which a type 4 allele is a risk factor for AD). The exact
medical community underscore the ambiguous role of relationship between cholesterol levels and AD risk or
vitamin E for chronic diseases. The study raised sever- progression is unclear. Serum cholesterol levels do not
al issues regarding vitamin E that need to be investi- appear to correlate with risk of AD, and statin levels do
gated.79,80 Nonetheless, leading medical organizations not affect production of β amyloid in humans, although
(as discussed later in this article) endorse consideration they do have an effect in vitro and in mice.85 However,

266 Vol. 3, No. 8 n October 2005


REVIEW

some recent case-control studies suggest decreased risk of the meantime, we have 4 drugs with proven efficacy,
AD with statins, but this is not fully supported by albeit modest, in at least some patients with AD.
prospective studies.86-88 Statins may offer a neuroprotec- Although some may argue, based on clinical studies,
tive or anti-inflammatory effect, rather than prevent that the effect of cholinesterase inhibitors or meman-
accumulation of β-amyloid plaques.85 tine is minimal, in reality any preservation of function
There are limited clinical studies of statins for AD. and delay of nursing home placement in an individual
Atorvastatin showed some clinical benefit in cognition patient has important and permanent consequences.
after 1 year of treatment (compared to placebo), but The primary care nurse, in identifying and managing
the difference was not statistically significant.89 It a patient with AD, needs to understand each of the
should be noted that this was a pilot study; the final following aspects of AD treatment: the expected out-
participant numbers at 12 months were low (n = 46).89 comes with these drugs and their impact on the
Another study of simvastatin (6 months) showed sig- patient and caregiver, the importance of reiterating
nificant reductions in cerebrospinal fluid levels of these expectations to the patient and caregiver, the
some but not all AD-related proteins (eg, tau, phos- common concurrent prescribing of cholinesterase
phorylated tau, β amyloid, and other proteins) and inhibitors with anticholinergic drugs in older adults,
slight improvement in cognition.90 Clearly, more stud- the use of newer agents as possible preventive treat-
ies are needed, but thus far data do not support a role ments for AD, and any updates in recommended treat-
for statins in the treatment of AD. ment strategies from leading medical organizations.
Because of the profound and sometimes rapid loss of
THERAPEUTIC CHOICES memory and function with AD, the importance of try-
The American Psychiatric Association and the ing at least 1 of these treatments in a patient with AD
American Academy of Neurology (AAN) have published cannot be overstated.
consensus guidelines on the treatment of patients with
dementia.91-93 The American Geriatrics Society has
endorsed the AAN guidelines, and guidelines for family
REFERENCES
physicians have also been published.94,95 All of these pro-
fessional organizations recommend the use of acetyl- 1. Alzheimer’s Disease Education and Referral Center Web
cholinesterase inhibitors for treatment of mild to site. Alzheimer’s disease—unraveling the mystery. Available
moderate AD, noting comparable efficacy among the 3 at: http://www.alzheimers.org/unraveling/01.htm.
drugs and the importance of clearly conveying expected Accessed June 17, 2005.
2. Fox NC, Crum WR, Scahill RI, et al. Imaging of onset and
outcomes to patients and family members/caregivers. All progression of Alzheimer’s disease with voxel-compression
3 guidelines recommend vitamin E or support consider- mapping of serial magnetic resonance images. Lancet.
ation of its use to slow or delay disease progression, but 2001;358:201-205.
they cite insufficient evidence to support the use of anti- 3. Kaye JA, Swihart T, Howieson D, et al. Volume loss of the
hippocampus and temporal lobe in healthy elderly per-
inflammatory medications in patients with AD.91-93,95 sons destined to develop dementia. Neurology. 1997;
However, these guidelines were established before many 48:1297-1304.
of the NSAID or statin study publications. In fact, 4. Forstl H, Zerfass R, Geiger-Kabisch C, et al. Brain atrophy
statins and memantine are not mentioned in any of the in normal aging and Alzheimer’s disease. Volumetric dis-
crimination and clinical correlations. Br J Psychiatry.
current guidelines. 1995;167:739-746.
5. Cummings JL. Alzheimer’s disease. N Engl J Med.
CONCLUSIONS 2004;351:56-67.
The emerging details of AD pathophysiology pro- 6. Selkoe DJ. Defining molecular targets to prevent Alzheimer
disease. Arch Neurol. 2005;62:192-195.
vide possible therapeutic targets. β-amyloid plaques 7. Bertram L, Tanzi RE. Alzheimer’s disease: one disorder, too
and NFTs are the long-held hallmarks of AD, but it many genes? Hum Mol Genet. 2004;13:R135-R141.
appears that they (along with the reduced neuronal 8. Walsh DM, Selkoe DJ. Deciphering the molecular basis of
function and neurotransmitter availability) occur years memory failure in Alzheimer’s disease. Neuron. 2004;
44:181-193.
before clinical symptoms emerge. Future treatment 9. Walker LC, Ibegbu CC, Todd CW, et al. Emerging
strategies will probably focus on the pathophysiologic prospects for the disease-modifying treatment of Alzheimer’s
changes in AD and address multiple mechanisms. In disease. Biochem Pharmacol. 2005;69:1001-1008.

Advanced Studies in Nursing n 267


REVIEW

10. Gilman S, Koller M, Black RS, et al. Clinical effects of Aβ 30. Mossner R, Schmitt A, Syagailo Y, et al. The serotonin trans-
immunization (AN1792) in patients with AD in an interrupt- porter in Alzheimer’s and Parkinson’s disease. J Neural
ed trial. Neurology. 2005;64:1553-1562. Transm Suppl. 2000;60:345-350.
11. Fox NC, Black RS, Gilman S, et al. Effects of Aβ immuniza- 31. Lai MK, Tsang SW, Alder JT, et al. Loss of serotonin 5-
tion (AN1792) on MRI measures of cerebral volume in HT2A receptors in the postmortem temporal cortex corre-
Alzheimer disease. Neurology. 2005;64:1563-1572. lates with rate of cognitive decline in Alzheimer’s disease.
12. Selkoe DJ. Alzheimer’s disease is a synaptic failure. Psychopharmacology. 2005;179:673-677.
Science. 2002;298:789-791. 32. Tsang SW, Lai MK, Francis PT, et al. Serotonin transporters
13. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s are preserved in the neocortex of anxious Alzheimer’s dis-
disease: progress and problems on the road to therapeu- ease patients. Neuroreport. 2003;14:1297-1300.
tics. Science. 2002;297:353-356. 33. Heneka MT, Galea E, Gavriluyk V, et al. Noradrenergic
14. Perry EK, Tomlinson BE, Blessed G, et al. Correlation of depletion potentiates beta-amyloid-induced cortical inflam-
cholinergic abnormalities with senile plaques and mental mation: implications for Alzheimer’s disease. J Neurosci.
test scores in senile dementia. BMJ. 1978;2:1457-1459. 2002;22:2434-2442.
15. Francis PT. Glutamatergic systems in Alzheimer’s disease. Int 34. Herrmann N, Lanctot KL, Khan LR. The role of norepinephrine
J Geriatr Psychiatry. 2003;18(suppl 1):S15-S21. in the behavioral and psychological symptoms of dementia.
16. Rogawski MA, Wenk GL. The neuropharmacological basis J Neuropsychiatry Clin Neurosci. 2004;16:261-276.
for the use of memantine in the treatment of Alzheimer’s dis- 35. Dietschy JM, Turley SD. Cholesterol metabolism in the brain.
ease. CNS Drug Rev. 2003;9:275-308. Curr Opin Lipidol. 2001;12:105-112.
17. Danysz W, Parsons CG, Möbius HJ, et al. Neuroprotective 36. Reiss AB. Cholesterol and apolipoprotein E in Alzheimer’s dis-
and symptomological action of memantine relevant for ease. Am J Alzheimers Dis Other Demen. 2005;20:91-96.
Alzheimer’s disease—a unified glutamatergic hypothesis on 37. Launer LJ, White LR, Petrovitch H, et al. Cholesterol and
the mechanism of action. Neurotox Res. 2000;2:85-97. neuropathologic markers of AD: a population-based autop-
18. Parsons CG, Danysz W, Quack G. Glutamate in CNS dis- sy study. Neurology. 2001;57:1447-1452.
orders as a target for drug development: an update. Drug 38. Donepezil US prescribing information. Available at:
News Perspect. 1998;11:523-569. http://www.aricept.com/pi/aricept_pi.htm#clinicalpharm.
19. Abramov AY, Canevari L, Duchen MR. Calcium signals Accessed July 15, 2005.
induced by amyloid beta peptide and their consequences 39. Galantamine US prescribing information. Available at:
in neurons and astrocytes in culture. Biochim Biophys Acta. http://www.us.reminyl.com/active/janus/en_US/assets/
2004;1742:81-87. rem/reminyl.pdf. Accessed July 15, 2005.
20. Gordon-Krajcer W, Gajkowska B. Excitotoxicity-induced 40. Memantine US prescribing information. Available at:
expression of amyloid precursor protein (beta-APP) in the http://www.namenda.com/pdf/namenda_pi.pdf.
hippocampus and cortex of rat brain: an electron- Accessed July 15, 2005.
microscopy and biochemical study. Folia Neuropathol. 41. Rivastigmine US prescribing information. Available at:
2001;39:163-173. http://www.pharma.us.novartis.com/product/pi/pdf/
21. Harkany T, Abraham I, Timmerman W, et al. β-amyloid exelon.pdf. Accessed July 21, 2005.
neurotoxicity is mediated by a glutamate-triggered excitotox- 42. Rogers SL, Farlow MR, Doody RS, et al. A 24-week, dou-
ic cascade in rat nucleus basalis. Eur J Neurosci. ble-blind, placebo-controlled trial of donepezil in patients
2000;12:2735-2745. with Alzheimer’s disease. Donepezil Study Group.
22. Scott HL, Tannenberg AG, Dodd PR. Variant forms of neu- Neurology. 1998;50:136-145.
ronal glutamate transporter sites in Alzheimer disease cere- 43. Rogers SL, Doody RS, Mohs RC, Friedhoff LT. Donepezil
bral cortex. J Neurochem. 1995;64:2193-2202. improves cognition and global function in Alzheimer dis-
23. Scott HL, Pow DV, Tannenberg AE, Dodd PR. Aberrant ease: a 15-week, double-blind, placebo-controlled study.
expression of the glutamate transporter excitatory amino Arch Intern Med. 1998;158:1021-1031.
acid transporter 1 (EAAT1) in Alzheimer’s disease. 44. Corey-Bloom J, Anand R, Veach J. A randomized trial evalu-
J Neurosci. 2002;22:RC206. ating the efficacy and safety of ENA 713 (rivastigmine tar-
24. Butterfield DA, Griffin S, Munch G, Pasinetti GM. Amyloid trate), a new acetylcholinesterase inhibitor, in patients with
beta-peptide and amyloid pathology are central to the mild to moderately severe Alzheimer’s disease. Int J Geriatr
oxidative stress and inflammatory cascades under which Psychopharmacol. 1999;1:55-65.
Alzheimer’s disease brain exists. J Alzheimers Dis. 45. Anand R, Messina J, Hartmann R. Dose-response effect of
2002:4:193-201. rivastigmine in the treatment of Alzheimer’s disease. Int J
25. Zhu X, Raina AK, Smith MA. Cell cycle events in neurons. Geriatr Psychopharmacol. 2000;2:68-72.
Proliferation or death? Am J Pathol. 1999;155:327-329. 46. Rosler M, Anand R, Cicin-Sain A, et al. Efficacy and safety of
26. Zhu X, Lee HG, Casadesus G, et al. Oxidative imbalance in rivastigmine in patients with Alzheimer’s disease: international
Alzheimer’s disease. Mol Neurobiol. 2005;31:205-218. randomised controlled trial. BMJ. 1999;318:633-638.
27. Kim SU, de Vellis J. Microglia in health and disease. 47. Raskind MA, Peskind ER, Wessel T, et al. Galantamine in
J Neurosci Res. 2005;81:302-313. AD: a 6-month randomized, placebo-controlled trial with a
28. McGeer P, McGeer EG. Inflammation, cytotoxicity, and 6-month extension. Neurology. 2000;54:2261-2268.
Alzheimer disease. Neurobiol Aging. 2001;22:799-809. 48. Tariot PN, Solomon PR, Morris JC, et al. A 5-month, ran-
29. Akiyama H, Barger S, Barnum S, et al. Inflammation and domized, placebo-controlled trial of galantamine in AD.
Alzheimer’s disease. Neurobiol Aging. 2000;21:383-421. Neurology. 2000;54:2269-2276.

268 Vol. 3, No. 8 n October 2005


REVIEW

49. Wilcock GK, Lilienfeld S, Gaens E. Efficacy and safety of the effect on caregiver burden. J Am Geriatr Soc.
galantamine in patients with mild to moderate Alzheimer’s 2003;51:737-744.
disease: multicenter randomized controlled trial. BMJ. 66. Potkin SG, Anand R, Hartman R, et al. Impact of
2000;321:1445-1449. Alzheimer’s disease and rivastigmine on activities of daily
50. Rockwood K, Mintzer J, Truyen L, et al. Effects of a flexible living over the course of mild to moderately severe disease.
galantamine dose in Alzheimer’s disease: a randomized, Prog Neuropsychopharmacol Biol Psychiatry.
controlled trial. J Neurol Neurosurg Psychiatry. 2002;26:713-720.
2000;71:589-595. 67. Galasko D, Kershaw PR, Schneider L, et al. Galantamine
51. Rogers SL, Doody RS, Pratt RD, Ieni JR. Long-term efficacy maintains ability to perform activities of daily living in
and safety of donepezil in the treatment of Alzheimer’s dis- patients with Alzheimer’s disease. J Am Geriatr Soc.
ease: final analysis of a US multicenter open-label study. 2004;52:1070-1076.
Eur Neuropsychopharmacol. 2000;10:195-203. 68. Sano M, Wilcock GK, van Baelen B, Kavanagh S. The effects
52. Karaman Y, Erdogan F, Koseoglu E, et al. A 12-month study of galantamine treatment on caregiver time in Alzheimer’s dis-
of the efficacy of rivastigmine in patients with advanced ease. Int J Geriatr Psychiatry. 2003;18:942-950.
moderate Alzheimer’s disease. Dement Geriatr Cogn 69. Tariot PN, Jakimovich L. Donepezil use for advanced
Disord. 2005;19:51-56. Alzheimer’s disease-a case study from a long-term care facil-
53. Farlow M, Anand R, Messina J Jr, et al. A 52-week study of ity. J Am Med Dir Assoc. 2003;4:216-219.
the efficacy of rivastigmine in patients with mild to moder- 70. Geldmacher DS, Provenzano G, McRae T, et al. Donepezil
ately severe Alzheimer’s disease. Eur Neurol. is associated with delayed nursing home placement in
2000;44:236-241. patients with Alzheimer’s disease. J Am Geriatr Soc.
54. Grossberg G, Irwin P, Satlin A, et al. Rivastigmine in 2003;51:937-944.
Alzheimer disease. Efficacy over two years. Am J Geriatr 71. AD2000 Collaborative Group. Long-term donepezil treatment
Psychiatry. 2004;12:420-431. in 565 patients with Alzheimer’s disease (AD2000): random-
55. Dorwaiswamy PM, Krishnan KR, Anand R, et al. Long-term ized double-blind trial. Lancet. 2004;363:2105-2115.
effects of rivastigmine in moderately severe Alzheimer’s dis- 72. Wilcock G, Howe I, Coles H, et al. A long-term compari-
ease. Does early initiation of therapy offer sustained bene- son of galantamine and donepezil in the treatment of
fits? Prog Neuropsychopharmacol Biol Psychiatry. Alzheimer’s disease. Drugs Aging. 2003;20:777-789.
2002;26:705-712. 73. Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The con-
56. Lyketsos CG, Reichman WE, Kershaw P, Zhu Y. Long-term out- current use of anticholinergics and cholinesterase inhibitors:
comes of galantamine treatment in patients with Alzheimer dis- rare event or common practice? J Am Geriatr Soc.
ease. Am J Geriatr Psychiatry. 2004;12:473-482. 2005;52:2082-2087.
57. Raskind MA, Pesking ER, Truyen L, et al. The cognitive ben- 74. Reisberg B, Doody R, Stoffler A, et al. Memantine in mod-
efits of galantamine are sustained for at least 36 months: a erate-to-severe Alzheimer’s disease. N Engl J Med.
long-term extension trial. Arch Neurol. 2004;61:252-256. 2003;348:1333-1341.
58. Pirttila T, Wilcock G, Truyen L, Damaraju CV. Long-term effi- 75. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine
cacy and safety of galantamine in patients with mild to treatment in patients with moderate to severe Alzheimer dis-
moderate Alzheimer’s disease: multicenter trial. Eur J ease already receiving donepezil. A randomized controlled
Neurol. 2004;11:734-741. trial. JAMA. 2004;291:317-324.
59. Seltzer B, Zolnouni P, Nunez M, et al. Efficacy of donepezil in 76. Berman K, Brodaty H. Tocopherol (vitamin E) in Alzheimer’s
early stage Alzheimer disease: a randomized placebo-con- disease and other neurodegenerative disorders. CNS
trolled study. Arch Neurol. 2004;61:1852-1856. Drugs. 2004;18:807-825.
60. Feldman H, Gauther S, Hecker J, et al. A 24-week, random- 77. Kontush K, Schekatolina S. Vitamin E in neurodegenerative
ized, double-blind study of donepezil in moderate to severe disorders: Alzheimer’s disease. Ann N Y Acad Sci.
Alzheimer’s disease. Neurology. 2001;57:613-620. 2004;1031:249-262.
61. Farlow M, Hake A, Messina J, et al. Response of patients 78. Petersen RC, Thomas RG, Grundman M, et al. Vitamin E
with Alzheimer disease to rivastigmine treatment is predict- and donepezil for the treatment of mild cognitive impair-
ed by the rate of disease progression. Arch Neurol. ment. N Engl J Med. 2005;352:2379-2388.
2001;58:417-422. 79. Miller ER III, Pastor-Barriuso R, Dalal D, et al. Meta-analysis:
62. Almkvist O, Darreh-Shori T, Stefanova E, et al. Preserved cog- high-dosage vitamin E supplementation may increase all-
nitive function after 12 months of treatment with rivastigmine in cause mortality. Ann Intern Med. 2005;142:37-46.
mild Alzheimer’s disease in comparison with untreated AD and 80. Letters to the editor. High dosage vitamin E supplementation
MCI patients. Eur J Neurol. 2004;11:253-261. and all-cause mortality. Ann Intern Med. 2005;143:150-158.
63. Hope T, Keene J, Gedling K, et al. Predictors of institutional- 81. Etminan M, Gill S, Samii A. Effect of non-steroidal anti-
ization for people with dementia living at home with a inflammatory drugs on risk of Alzheimer’s disease: systemat-
carer. Int J Geriatr Psychiatry. 1998;13:682-690. ic review and meta-analysis of observational studies. BMJ.
64. Mohs RC, Doody RS, Morris JC, et al. A 1-year placebo- 2003;327:128-132.
controlled preservation of function survival study of 82. Szekely CA, Thorne JE, Zandi PP, et al. Nonsteroidal anti-
donepezil in AD patients. Neurology. 2001;57:481-488. inflammatory drugs for the prevention of Alzheimer’s dis-
65. Feldman H, Gauther S, Hecker J, et al. Efficacy of ease: a systematic review. Neuroepidemiology.
donepezil on maintenance of activities of daily living in 2004;23:159-169.
patients with moderate to severe Alzheimer’s disease and 83. Aisen PS, Davis KL, Berg JD, et al. A randomized controlled

Advanced Studies in Nursing n 269


REVIEW

trial of prednisone in Alzheimer’s disease. Alzheimer’s Disease 91. American Psychiatric Association. Practice guideline for the
Cooperative Study. Neurology. 2000;54:588-593. treatment of patients with Alzheimer’s disease and other demen-
84. Aisen PS, Schafer KA, Grundman M, et al. Effects of rofe- tias of late life. Am J Psychiatry. 1997;154(suppl 5):1-39.
coxib or naproxen vs placebo on Alzheimer disease pro- 92. Rabins PV. Guideline watch. Practice guideline for the treat-
gression: a randomized controlled trial. JAMA. ment of patients with Alzheimer’s disease and other demen-
2003;289:2819-2826. tias of late life. American Psychiatric Association Web site.
85. Wolozin B. Cholesterol, statins and dementia. Curr Opin Available at: www.psych.org/psych_pract/treatg/pg/
Lipid. 2004;15:667-672. AlzWatch_081704.pdf. Accessed July 13, 2005.
86. Wolozin B, Kellman W, Ruosseau P, et al. Decreased 93. Doody RS, Stevens JC, Beck C, et al. Practice parameter:
prevalence of Alzheimer disease associated with 3-hydroxy- management of dementia (an evidence-based review).
3-methyglutaryl coenzyme A reductase inhibitors. Arch Neurology. 2001;56:1154-1166.
Neurol. 2000;57:1439-1443. 94. American Geriatrics Society Web site. Position statement-
87. Jick H, Zornberg GL, Jick SS, et al. Statins and the risk of clinical practice guidelines. Guidelines abstracted from the
dementia. Lancet. 2000;256:1627-1631. American Academy of Neurology’s Dementia Guidelines for
88. Zandi PP, Sparks DL, Khachaturian AS, et al. Do statins Early Detection, Diagnosis, and Management of Dementia.
reduce risk of incident dementia and Alzheimer disease? Available at: http://www.americangeriatrics.org/prod-
The Cache County Study. Arch Gen Psychiatry. ucts/positionpapers/aan_dementiaPF.shtml. Accessed June
2005;62:217-224. 11, 2005.
89. Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for 95. Cummings JL, Frank JC, Cherry D, et al. Guidelines for man-
the treatment of mild to moderate Alzheimer disease. Arch aging Alzheimer’s disease: Part II. Treatment. Am Fam
Neurol. 2005;62:753-757. Physician. 2002;65:2525-2534.
90. Sjogren M, Gustafsson K, Syversen S, et al. Treatment with
simvastatin in patients with Alzheimer’s disease lowers both
alpha- and beta-cleaved amyloid precursor protein. Dement
Geriatr Cogn Disord. 2003;16:25-30.

270 Vol. 3, No. 8 n October 2005

Potrebbero piacerti anche