You are on page 1of 15

Drugs Aging

DOI 10.1007/s40266-015-0266-9

SYSTEMATIC REVIEW

A Risk-Benefit Assessment of Dementia Medications: Systematic


Review of the Evidence
Jacob S. Buckley1 • Shelley R. Salpeter2,3

Ó Springer International Publishing Switzerland 2015

Abstract review. In pooled trial data, cholinesterase inhibitors


Background There is no cure for dementia, and no (ChEIs) produce small improvements in cognitive, func-
treatments exist to halt or reverse the course of the disease. tional, and global benefits in patients with mild to moderate
Treatments are aimed at improving cognitive and func- Alzheimer’s and Lewy body dementia, but the clinical
tional outcomes. significance of these effects are unclear. There is no sig-
Objective Our objective was to review the basis of nificant benefit seen for vascular dementia. The efficacy of
pharmacological treatments for dementia and to summarize ChEI treatment appears to wane over time, with minimal
the benefits and risks of dementia treatments. benefit seen after 1 year. There is no evidence for benefit
Methods We performed a systematic literature search of for those with advanced disease or those aged over
MEDLINE through November 2014, for English-language 85 years. Adverse effects are significantly increased with
trials and observational studies on treatment of dementia ChEIs, in a dose-dependent manner. A two- to fivefold
and mild cognitive impairment. Additional references were increased risk for gastrointestinal, neurological, and car-
identified by searching bibliographies of relevant publica- diovascular side effects is related to cholinergic stimula-
tions. Whenever possible, pooled data from meta-analyses tion, the most serious being weight loss, debility, and
or systematic reviews were obtained. Studies were included syncope. Those aged over 85 years have double the risk of
for review if they were randomized trials or observational adverse events compared with younger patients. Meman-
studies on dementia or mild cognitive impairment that tine monotherapy may provide some cognitive benefit for
evaluated efficacy outcomes or adverse outcomes associ- patients with moderate to severe Alzheimer’s and vascular
ated with treatment. Studies were excluded if they dementia, but the benefit is small and may wane over the
evaluated non-FDA approved treatments, or if they course of several months. Memantine exhibits no sig-
evaluated treatment in disorders other than dementia and nificant benefit in mild dementia or Lewy body dementia or
mild cognitive impairment. as an add-on treatment with ChEIs. Memantine has a
Results The literature search found 540 potentially rele- relatively favorable side-effect profile, at least under con-
vant studies, of which 257 were included in the systematic trolled trial conditions.
Conclusions ChEIs produce small, short-lived improve-
ments in cognitive function in mild to moderate dementia,
which may not translate into clinically meaningful effects.
Marginal benefits are seen with severe disease, long-term
treatment, and advanced age. Cholinergic side effects, in-
& Shelley R. Salpeter cluding weight loss, debility, and syncope, are clinically
salpeter@stanford.edu significant and could be especially detrimental in the frail
1
elderly population, in which the risks of treatment out-
Brown University, Providence, RI, USA
weigh the benefits. Memantine monotherapy may have
2
Stanford University School of Medicine, Stanford, CA, USA minimal benefits in moderate to severe dementia, balanced
3
1670 Amphlett Blvd, Suite 300, San Mateo, CA 94402, USA by minimal adverse effects.
J. S. Buckley, S. R. Salpeter

age, or frailty, would have been ineligible for these trials,


Key Points and the benefit of long-term treatment is unclear [13–18]. It
is also important to distinguish between statistical and
Cholinesterase inhibitors produce small clinical significance, as a small change in cognitive status
improvements in cognitive function in mild to may not translate into a meaningful change for the patient
moderate dementia, with marginal effects seen in [1, 2, 19]. In addition, a risk–benefit assessment must in-
severe disease, long-term treatment, and advanced clude quantification of adverse effects, especially in old
age. frail individuals in whom side effects may have serious
consequences for survival and quality of life [2, 11, 20].
Cholinesterase inhibitors are associated with
significant cholinergic side effects, including weight
loss, debility, and syncope, which can be detrimental 2 Methods
in the frail elderly population.
The available evidence indicates that memantine We performed a literature search of MEDLINE through
monotherapy produces small benefits in cognitive November 2014 for English-language studies evaluating
function in moderate to severe dementia, without the risks and benefits of ChEIs and memantine in the
significant adverse effects. treatment of dementia and mild cognitive impairment
(MCI). Additional references were identified by reviewing
the bibliographies of relevant papers. Whenever possible,
pooled trial data from meta-analyses or systematic reviews
1 Introduction were obtained. Search terms for cognitive disorders in-
cluded dementia, mild cognitive impairment, cognition
Dementia is a clinical syndrome characterized by acquired disorder, Alzheimer, Parkinson’s dementia, Lewy
global cognitive deficits that interfere with social and oc- body dementia, and vascular dementia. Search terms for
cupational functioning [1]. It typically presents in elderly treatment included cholinesterase inhibitor, acetyl-
individuals and involves a progressive decline in memory, cholinesterase inhibitor, donepezil, galantamine, rivastig-
reasoning, language, and executive function [2, 3]. The mine, and memantine. Studies were included for review if
estimates for dementia prevalence vary based on geo- they were randomized trials or observational studies that
graphic location and diagnostic criteria used, but it is evaluated efficacy outcomes (cognitive, neuropsychiatric,
thought that at least 5 % of people over the age of 60 years or global function, and disease progression) or adverse
live with dementia, and the total number of these cases is outcomes associated with treatment. Studies were excluded
expected to double every 20 years [4]. Incidence of de- if they evaluated non-FDA approved treatments for de-
mentia increases exponentially with advancing age, with mentia, such as ginkgo biloba, vitamin E, and hormonal
one population study observing it doubling every 5 years treatments [2, 10]. In addition, studies were excluded if
[5]. Due to the rapid expansion of the elderly population, they evaluated ChEIs or memantine in disorders other than
dementia is a serious public health concern, with an esti- dementia and MCI.
mated worldwide cost of US$600 billion in 2010 [6]. Two independent reviewers (JSB, SRS) completed data
Currently, there is no cure for dementia, and no treat- abstraction and evaluated the quality and risk of bias of
ments exist to halt or reverse its progression, so therapeutic included studies. The results were then presented in a
interventions are targeted to treat symptoms or to improve systematic review, following PRISMA (preferred reporting
cognitive function [7–9]. The two treatments with mar- items for systematic reviews and meta-analyses) guide-
keting approval for dementia are the cholinesterase in- lines. The primary scales used to measure cognition deficits
hibitors (ChEIs) donepezil, galantamine, and rivastigmine, were the mini-mental state examination (MMSE), the
and the low-affinity N-methyl-D-aspartate (NMDA) an- Alzheimer’s Disease Assessment Scale (ADAS) cognitive
tagonist memantine [1, 10]. The vast majority of published subscale (ADAS-cog), and the severe impairment battery
data available on dementia treatment are in short-term in- (SIB). Other domains assessed included the activities of
dustry-sponsored trials of ChEIs [11]. The trials have daily living (ADL), instrumental activities of daily living
mostly been limited to patient populations with a mean age (IADL), and the neuropsychiatric inventory (NPI). Global
of approximately 75 years, with relatively mild Alzhei- function and disease progression were also assessed. Ad-
mer’s dementia and few to no comorbidities [11, 12]. This verse events assessed included abdominal pain, nausea,
significantly decreases the clinical applicability of the re- vomiting, diarrhea, anorexia, headache, dizziness, insom-
search to real-world cases of dementia, as the vast majority nia, abnormal dreams, vertigo, agitation, muscle cramps,
of patients, such as those with severe disease, advanced tremor, bradycardia, syncope, pacemaker implantation,
Systematic Review of the Risks and Benefits of Dementia Medications

fatigue, asthenia, weight loss, falls, and drug–drug disorders, often collectively called Lewy body dementia
interactions. [30]. Lewy body dementia may account for 10–20 % of
The search identified approximately 6800 potentially global dementia cases, with the risk increasing with age
relevant articles, of which 540 were retrieved and evaluated. and years since Parkinson’s disease onset [31–34]. Many
Of those, 257 studies were included in the systematic review. other forms of clinically relevant dementia exist, but this
Study inclusion is represented in a flow diagram (Fig. 1). review focusses on these three main subgroups.
MCI is classified as a cognitive decline that exceeds the
typical aging process yet does not meet the diagnostic
3 Results criteria of dementia [35]. This cognitive decline usually
manifests as a loss in memory that is not significant enough
3.1 Epidemiology and Assessment of Dementia to alter ADL [35]. The prevalence estimates for MCI range
from 5 to 20 % in older individuals, with up to one-third of
There are several types and causes of dementia, with MCI cases progressing to dementia within 2 years [7].
confusion related to variations in diagnostic criteria and the Although MCI is a distinct entity, it is often considered to
fact that many forms of dementia exist concurrently [2, 21]. be a pre-clinical stage of dementia [7]. Studies have
Alzheimer’s disease is the most common cause, accounting evaluated whether pharmacological interventions are ca-
for roughly 50–70 % of all dementia cases [22]. Another pable of delaying the progression to dementia or improving
large subgroup is vascular dementia, which is often ob- the quality of life of individuals with MCI.
served concurrently with other forms, resulting in preva- Various assessment tools are available to measure cog-
lence estimates that vary considerably [23–28]. Mixed nitive function, which have been used to determine the
Alzheimer’s disease and vascular dementia is common, presence and severity of dementia as well as the progres-
accounting for approximately 25 % of all dementia cases sion of the disease [36]. One standard measurement is the
[29]. Vascular dementia becomes increasingly prevalent MMSE, which is easy to administer in the clinical setting
with advanced age, with at least 70 % of those over the age and measures cognitive function on a 30-point scale, with
of 90 years having some degree of vascular pathology [13]. higher scores corresponding to higher cognitive function
Finally, Parkinson’s disease dementia and dementia with [37, 38]. Although the MMSE has its limitations, including
Lewy bodies are two major types of dementia with similar decreased utility for detecting dementia in patients with
neuropathological presentations, indicating that they are minimal cognitive deficits or those with low education
part of the same condition or exist within a spectrum of levels, it has been widely used and validated globally, and

Fig.1 Flow chart of studies


Literature search of studies on
search
treatment of dementia (n = 6800)

Studies excluded: Not potentially


relevant (n = 6260)

Potentially relevant studies of


benefits or risks of medications
for cognition disorders (n = 540)

Studies excluded from systematic review:


On non-FDA approved dementia
treatments (n = 52)
Not on treatments for dementia or mild
cognitive impairment (n = 72)
No relevant information provided
(n = 159)

Randomized trials or observational


studies on acetylcholinesterase
inhibitors or memantine for dementia
or mild cognitive impairment (n = 257)
J. S. Buckley, S. R. Salpeter

is quite accurate for diagnosing and tracking mild to prevent excitotoxicity without interfering with necessary
moderate dementia [39–42]. Another common tool is the functions of glutamate in learning and memory [68–71].
ADAS-cog, a 70-question scale with higher scores indi- Despite very different pathogenic mechanisms, there are
cating worse cognitive performance, which was designed key similarities in the pathophysiology of Alzheimer’s
to detect and follow clinical changes in cognition [43, 44]. dementia, Lewy body dementia, and vascular dementia that
The ADAS-cog is most sensitive to changes in mild to result in cognitive decline, and these may indicate ChEIs
moderate dementia [44–48]. Another cognitive assessment and memantine as possible treatment options.
tool is the SIB, which is most helpful for those with severe In Alzheimer’s disease, the initiating event is thought to
dementia [49]. be the accumulation of amyloid beta-protein (Ab) plaques,
Other tests are commonly used to measure outcomes which in turn trigger hyperphosphorylation of tau protein
other than cognition in patients with dementia. An impor- and neuronal degeneration [72]. The Ab plaques cause a
tant measure in the determination of quality of life and decrease in acetylcholine release and synthesis, along with
degree of disability is the assessment of ADL, such as increased activity of acetylcholinesterase, impaired mus-
feeding and dressing, and IADL, such as handling money, carinic acetylcholine signaling pathways, and a decrease in
preparing food, or using the telephone [36, 50, 51]. In cholinergic signaling and function; these changes in turn
addition, behavioral and psychological symptoms can be indirectly result in disrupted NMDA-receptor activity and
assessed with the NPI, which focuses on behavioral dis- glutamate neurotoxicity [73, 74]. Since the plaques are
turbances such as delusions, anxiety, and disinhibition [51, concentrated in areas related to memory and executive
52]. Another research tool is the measurement of global function, such as the basal ganglia and parts of the tem-
dementia status, which integrates in-depth clinical assess- poral lobe and neocortex, the neural damage caused by
ments of patients with caregiver input to derive a basic cholinergic impairment and glutamate neurotoxicity is re-
quantitative measure of change [53]. lated to a loss of these cognitive functions [65].
There are no universally accepted cut-offs for defining Vascular dementia results from varied cerebrovascular
dementia severity, but it is reasonable to use MMSE scores to events such as ischemia, infarct, or hemorrhage, so its
divide dementia severity into mild (20–23), moderate pathogenesis is multifactorial and poorly defined [75]. The
(10–19), severe (1–9), and end-stage (0) [40, 41, 54–59]. cognitive deficits involved resemble those found in Alz-
MCI corresponds to an MMSE score of 24–27, although this heimer’s disease and other dementia types, as they involve
is not sufficient as a sole diagnostic criterion [58, 60–62]. cerebrovascular damage to similar neural regions, such as
Mild dementia can be detected by basic clinical assessment the temporal lobe, basal ganglia, and neocortex [75]. These
and is characterized by decreased memory of personal and vascular changes in the brain are associated with inflam-
current events and slightly diminished executive function mation, atrophy, and decreased cerebral perfusion, as well
[41]. In moderate dementia, the individual begins to lose as decreased inhibition of NMDA channels, leading to
independence and conversational skills and may forget sig- chronic overstimulation of glutamate receptors [71, 75].
nificant personal information [56, 63]. By the stage of severe There is also some evidence that these changes may result
dementia, there is a complete loss of independence in per- in impaired cholinergic transmission [67, 76].
forming ADLs associated with lack of orientation, but with Lewy body dementia is a multisystem disease with
some maintained ambulatory and verbal functioning [63, various presentations along a spectrum; if the parkinsonian
64]. End-stage dementia, in which the individual no longer extrapyramidal symptoms precede dementia, it is consid-
has the ability to walk or speak, is an important clinical stage ered to be Parkinson’s disease dementia, but if the order of
that may persist for years before death [63, 64]. presentation is reversed it is typically diagnosed as de-
mentia with Lewy bodies [77]. Both disorders eventually
3.2 Pathophysiological Basis of Dementia involve the production of proteinaceous cytoplasmic in-
with Rationale for Treatment clusions throughout the brain, called Lewy bodies, with
neurodegeneration of dopaminergic neurons in the sub-
Cognitive decline in dementia is due in part to deficient stantia nigra and denervation of cholinergic neurons in the
cholinergic activity in the basal ganglia and neocortex basal forebrain and cerebral cortex [77–79]. There is some
caused by destruction of cholinergic neurons. ChEIs evidence that these conditions may also induce glutamate
function by inhibiting acetylcholinesterase, thereby in- hyperactivity by altering the expression of glutamate re-
creasing available acetylcholine [65–67]. Another cause of ceptors, and this is often worsened by chronic L-dopa ad-
cognitive decline common to various types of dementia is ministration, which is commonly used in the treatment of
excitatory neurotoxicity caused by overstimulation of Parkinson’s disease [69, 80, 81].
NMDA-glutamate receptors, and memantine could serve Ultimately, the use of ChEIs may serve to compensate
by blocking NMDA activity at a low enough affinity to for decreased cholinergic activity by increasing the
Systematic Review of the Risks and Benefits of Dementia Medications

availability of acetylcholine released from limited cholin- one long-term trial by the Alzheimer’s Disease Col-
ergic neurons, and memantine may offer neuroprotective laborative Group [100, 101]. This raises the possibility of
effects from excitotoxic stimulation of overexpressed publication bias or selective reporting of the results. One
NMDA pathways, but neither medication can reverse or study evaluating the potential bias in reporting adverse
slow the underlying pathophysiological progression of the events found that most of the randomized trials stated that
disease. Therefore, the target of these therapies is to at- they examined harm, but many did not report mortality
tempt to reduce symptoms related to the disease, rather data or provide clear definitions and detailed analyses of
than address the neurodegenerative processes themselves. harm [102].
A recent clinical practice guideline by the American
3.3 Cholinesterase Inhibitors College of Physicians and the American Academy of
Family Physicians presented the available evidence on
3.3.1 Benefits of Cholinesterase Inhibitors for Mild ChEI treatments based on systematic evidence reviews
Cognitive Impairment sponsored by the Agency for Healthcare Research and
Quality [1, 11, 93]. They concluded that ChEI treatment for
No medication has marketing approval for the treatment of dementia results in a small statistically significant im-
MCI, but many investigators have evaluated whether provement in cognitive function that was not considered to
ChEIs could delay the progression to dementia or provide be clinically important [1]. Pooled data from ChEI trials in
clinically significant improvements in cognition for indi- Alzheimer’s disease, Lewy body dementia, and vascular
viduals with MCI. A few small studies have shown some dementia show average improvements in the MMSE
minimal cognitive benefits of ChEIs in the treatment of ranging from 0.8 to 1.6 points on a 30-point scale, while 3
MCI [56, 82–84], but pooled data analyses of the trials points or more was generally considered to be clinically
show that there is no significant difference in progression significant [1, 12, 30, 93, 94, 103]. Pooled data also showed
to dementia, nor in global, cognitive, or neuropsychiatric an average improvement in ADAS-cog of approximately
outcomes [7, 85, 86]. A significant increase in adverse 1.4–2.7 points on a 70-point scale, while several studies
events was observed for MCI patients treated with ChEIs, have used a change of 4 points or more to indicate clinical
mainly gastrointestinal side effects such as nausea, diar- relevance [1, 12, 93, 94, 103].
rhea, and vomiting, as well as cardiac syncope and head- The majority of available data has been in mild to
aches [7, 54, 84, 85, 87, 88]. Therefore, ChEIs are not moderate Alzheimer’s disease, with limited data on Lewy
recommended for the treatment of MCI [89, 90]. body dementia and vascular dementia [11]. The pooled
data from meta-analyses on ChEIs have shown relatively
3.3.2 Benefits of Cholinesterase Inhibitors for Dementia similar results for cognitive outcomes in Alzheimer’s de-
mentia and Lewy body dementia, with improvements on
Numerous studies have investigated the efficacy of differ- the order of 1.5 points in MMSE and 2.5 points in ADAS-
ent treatments for cognitive function in dementia, and cog compared with placebo [12, 30]. In comparison, trials
several systematic reviews and meta-analyses have worked on vascular dementia show marginal cognitive effects of
to pool the data and summarize the results (Table 1) [1, 2, ChEIs (0.8 points in MMSE and 1.5 points in ADAS-cog),
11, 20, 30, 56, 91–96]. The three commercially available with some studies finding no improvements at all [94].
ChEIs, donepezil, galantamine, and rivastigmine, have In addition to cognitive assessment, many studies also
marketing approval for the treatment of mild to moderate assessed global and functional outcomes of ChEI treat-
dementia in most countries [10, 56]. The first-generation ment. The results from studies on global outcomes have
ChEI, tacrine, was removed from the US market due to been mixed, with some studies concluding that there is no
poor efficacy and high incidence of serious adverse events, significant change and others finding slight improvements
and is not evaluated here [2, 20]. The available ChEIs [12, 30, 93, 94]. Pooled data show a mean improvement of
differ slightly in their pharmacological properties, but they 0.4–0.5 for treatment in Alzheimer’s and Lewy body de-
share the main mechanism of acetylcholinesterase inhibi- mentia in a 7-point interview-based global assessment of
tion [2, 97]. Studies have shown similar efficacies with change scale [30, 93, 104]. Functional measures of ADL
these medications so the data will be pooled for this review showed similarly mixed results for the treatment of Alz-
[2, 91, 98, 99]. A meta-analysis also concluded that results heimer’s and Lewy body dementia, with studies achieving
in efficacy for low and high doses of ChEIs were compa- improvements on the order of 0.1 standard deviations
rable, so results from studies using different doses will also compared with placebo, which was not considered to be
be pooled [20]. clinically significant. [1, 28, 87, 89, 93] Studies on ChEIs
Essentially all of the published trials on ChEI use in for vascular dementia have found negligible improvements
dementia were industry sponsored, with the exception of in global and functional measures [94]. A secondary
J. S. Buckley, S. R. Salpeter

Table 1 Benefit assessment of cholinesterase inhibitors for dementia by type


Condition The evidence Comments

Mild cognitive impairment


Mild cognitive Minimal cognitive benefits and possible delay of Pooled data show no significant delay in dementia
impairment progression to dementia suggested by a few small studies progression or in cognitive and global benefits
Dementia
Mild-moderate Improvements on the order of 1.5 MMSE (30-point scale), The most evidence exists for otherwise relatively young and
AD 2.5 ADAS-cog (70-point scale), 0.5 in interview-based healthy individuals with this dementia type. While these
global assessment of change (7-point scale), 2 points in improvements are statistically significant, they fall short
NPI (144-point scale), and 0.1 standard deviations of ADL of many standards of clinical significance, and most do
scales not assess for longer than 1 year
Mild–moderate Similar outcomes in cognitive, neuropsychiatric, and global While there is slightly less evidence available for Lewy
Lewy body domains as with mild–moderate AD body dementia than AD, pooled evidence indicates the
dementia benefit of ChEIs is similar for both conditions in the short
term
Mild–moderate Some small studies have shown more marginal benefit than Fewer large and long-term studies have been conducted on
vascular for AD and Lewy body dementia—on the order of 0.8 vascular dementia than on other dementia types, but
dementia MMSE, 1.5 ADAS-cog, and negligible improvements in pooled evidence indicates that benefits are small or
other domains nonexistent
Advanced and end- What limited evidence exists is mostly for AD, and shows A huge paucity of evidence for severe dementia of any type
stage dementia of only small benefits to cognition—4 to 7 points SIB (133- and separately for dementia patients with advanced age
any type point scale) and little to no benefits in other domains makes assessing data difficult. Limited pooled data
indicate that the effectiveness of ChEIs decreases with
increasing dementia severity, age, and duration of
treatment
AD Alzheimer’s dementia, ADAS-cog Alzheimer’s Disease Assessment Scale-cognitive subscale, ADL activities of daily living, ChEI choli-
nesterase inhibitors, MMSE mini-mental state examination, NPI neuropsychiatric inventory, SIB severe impairment battery

outcome often studied in dementia trials is the incidence There is also limited evidence for efficacy of ChEIs in
and severity of neuropsychiatric symptoms, including be- long-term treatment, with most randomized trials lasting
havioral and psychiatric disturbances, common in patients only 3–6 months despite the fact that patients often con-
with dementia [104]. As with global and functional scores, tinue therapy indefinitely [12, 112]. Only four trials have
pooled data showed a marginal improvement of about 2 of lasted 1 year or longer, all in mild to moderate Alzheimer’s
144 points in NPI for Alzheimer’s and Lewy body de- disease, and they provide some evidence for a waning of
mentia, and no improvement for vascular dementia [91, 94, effect over time [100, 113–115]. One 1-year trial of
103]. donepezil found an improvement in MMSE of 1.5 points
Very few studies have evaluated severe dementia by 1 year [113], while another trial showed improvement at
(MMSE 1–9), and none have looked at end-stage dementia 6 months but no significant effect by 1 year [114]. One
(MMSE 0). In fact, only one published study with 248 2-year trial of galantamine found an improvement in
participants examined monotherapy of ChEIs in patients MMSE of 0.7 points by 2 years [115]. However, another
with severe dementia (MMSE 1–10) [105]. If the criteria 2-year trial of donepezil, which had a complicated protocol
were expanded to define severe dementia as MMSE 1–12, with two randomization periods, found an improvement in
and then post hoc subgroup analyses of larger studies were MMSE of 0.9 points by 12 weeks that did not significantly
included, that would allow for the inclusion of a few more change in the second randomization period for the duration
small trials of short duration [106–109]. One other study of the trial [100]. The two primary outcomes of that trial,
evaluated severe dementia but did not classify patients time to institutionalization and loss of critical ADLs, did
according to MMSE [110]. These trials had variable results not significantly differ between treatment and placebo by
using the SIB, with some showing minimal cognitive 2 years [100].
benefit (4–7 points of the 133-point SIB compared with Finally, a paucity of data exist for the effectiveness of
placebo) and others showing a trend to worse scores ChEIs in the treatment of those with advanced age; in
compared with placebo [105–111]. Treatment of severe pooled trial data, the mean age was approximately
dementia resulted in minimal to no benefit in ADLs or in 75 years, with few participants included over the age of
NPI scores [105–109]. 85 years [11, 12, 94, 116, 117]. A single trial examined the
Systematic Review of the Risks and Benefits of Dementia Medications

effectiveness of donepezil in a population of nursing home In addition, similar effects have been seen with Alzhei-
residents with a mean age of 85 years and found no sig- mer’s disease, vascular dementia, Lewy body dementia,
nificant improvement in MMSE or neuropsychiatric out- and MCI [7, 11, 12, 30, 94].
comes at 24 weeks [118]. Pooled data from dementia trials show that ChEI treat-
ment is associated with a twofold increase in drop-outs due
3.3.3 Risks of Cholinesterase Inhibitors to adverse events compared with placebo [12, 103]. The
absolute increase in risk is 10 %, indicating that the
ChEIs can cause adverse effects due to increased cholin- number needed to treat over 3–6 months to cause one
ergic stimulation, both centrally and peripherally [119, significant drug reaction is 10 [12]. For patients with ad-
120]. Acetylcholine-containing neurons exist in the brain, vanced age, frailty, and comorbidities, and those using
mainly in the cerebral cortex, and in the periphery, both in concomitant medications, there is heightened concern for
the autonomic parasympathetic nervous system and skele- clinically relevant adverse effects associated with ChEIs.
tal muscle [121]. The central effects result in gastroin- These patients, in general, have not been included in the
testinal and neurological disturbances, while peripheral trial data, as less than 10 % of patients treated in clinical
cholinergic activity related to vagal stimulation results in practice would have been eligible for the published trials
cardiovascular and gastrointestinal events, as well as gen- [15]. Therefore, the exact magnitude of risk for real-world
eral effects such as weakness or fatigue (Table 2). The patients is uncertain. Another major difficulty in evaluating
ChEIs donepezil, galantamine, and rivastigmine have the risks of treatment is that the vast majority of random-
slightly different mechanisms of action but they share the ized trials are industry sponsored [102]. One study
common effect of reversible acetylcholinesterase inhibition evaluated 27 ChEI trials that stated they reported on the
with relative selectivity for central nervous system effects; safety and tolerability of the medication; however, 90 % of
they have been found to have similar adverse effects, so the these trials did not report standard regulatory agency-de-
data for these drugs will be reported together [2, 121, 122]. fined serious adverse events, making interpretation of the
data very difficult [102]. We provide best estimates from
randomized trial data as well as real-world experiences.
Table 2 Adverse effects of cholinesterase inhibitors compared with
placebo in Alzheimer’s disease
3.3.4 Risks of Cholinesterase Inhibitors: Gastrointestinal
Adverse outcome OR (95 % CI)
Effects
Gastrointestinal
Abdominal pain 1.95 (1.46–2.61) Procholinergic drugs such as ChEIs significantly increase
Anorexia 3.75 (2.89–4.87) gastrointestinal side effects, which can be attributed to both
Diarrhea 1.91 (1.59–2.3) central and peripheral cholinergic pathways [119, 122].
Nausea 4.87 (4.13–5.74) Abdominal pain, nausea, vomiting, diarrhea, and anorexia
Vomiting 4.82 (3.91–5.94) are all increased by two- to fivefold compared with placebo
Neurological in pooled data of Alzheimer’s trials lasting at least
Abnormal dreams 5.38 (1.34–21.55) 6 months (Table 2) [12]. These effects may be particularly
Dizziness 1.99 (1.64–2.42) prominent in the initial dose escalation phase but can re-
Headache 1.56 (1.27–1.91) main throughout long-term treatment, often resulting in
Insomnia 1.49 (1.12–2.00)
discontinuation of the drug [12, 119]. The degree of ad-
Tremor 6.82 (1.99–23.37)
verse gastrointestinal side effects appears to be dose de-
Vertigo 3.95 (1.08–14.46)
pendent [122].
In a meta-analysis of ChEI treatment of Alzheimer’s
Cardiovascular
dementia, a threefold increase in clinically significant
Syncope 1.90 (1.09–3.33)
weight loss compared with placebo was observed, related
Edema 2.08 (1.01–4.28)
at least in part to these gastrointestinal side effects [12].
General
Little inter-study heterogeneity was observed in the results,
Asthenia 2.47 (1.27–4.81)
indicating a consistent class effect on weight loss for all
Fatigue 4.39 (1.21–15.85)
ChEIs [12]. Evaluation of trial data indicates a dose de-
Muscle cramps 13.32 (1.71–103.74)
pendence related to weight loss, with a loss of C7 % of
Weight loss 2.99 (1.89–4.75)
body weight occurring in 10–25 % of patients receiving
At least one adverse event 2.51 (2.14–2.95)
high-dose ChEIs [122–125]. In one randomized trial of
Based on Birks [12] ChEI use in nursing home patients with dementia, a two-
CI confidence interval, OR odds ratio fold increase in significant weight loss was observed for
J. S. Buckley, S. R. Salpeter

treatment compared with placebo, with those aged over ChEIs in patients with MCI also found a twofold increase
85 years having almost twice the risk of those aged under in risk of syncope [7]. Inter-study heterogeneity in these
85 years [118]. Weight loss can be quite detrimental in the pooled analyses was low, indicating a consistent increased
frail elderly population and has been associated with de- risk for syncope associated with ChEIs [7, 12]. Similar
creased quality of life, functional decline, institutionaliza- results have been found in a large population-based ob-
tion, and increased mortality [126–130]. Observational servational study of community-dwelling subjects with
studies have indicated that subsequent weight gain can dementia, which, after adjusting for potential confounding
occur when ChEIs are discontinued [131, 132]. variables, found that ChEI use was associated with a sig-
nificantly increased risk for hospital visits for syncope and
3.3.5 Risks of Cholinesterase Inhibitors: Neurological for syncope-related events such as pacemaker insertion and
Effects falls with hip fracture [136]. The absolute increase in risk
for syncope in the study was 13 per 1000 person-years,
Central nervous system effects are frequently reported with indicating that the number needed to treat for 1 year to
ChEIs [119]. In pooled data from Alzheimer’s trials, a cause one hospitalization for syncope was 76 [136]. The
statistically significant increase, on the order of two- to absolute increase in hip fracture was 3 per 1000 person-
fivefold, was observed for headache, dizziness, insomnia, years, indicating a number needed to treat of 333 patients
abnormal dreams, and vertigo (Table 2) [12]. Observa- for 1 year to cause one hip fracture [136].
tional evidence also exists that agitation may be increased
with ChEI use [119, 133]. Muscle cramps with ChEI use, 3.3.7 Risks of Cholinesterase Inhibitors: General Effects
which may be related to direct cholinergic stimulation at
the neuromuscular junction, is increased 13-fold for ChEIs Acetylcholinesterase inhibitors can also cause some gen-
compared with placebo, and could result in muscle weak- eral adverse effects through cholinergic stimulation of
ness or falls [12, 119]. central and peripheral pathways [122]. These include fa-
Tremor and Parkinsonian symptoms are also sig- tigue and asthenia, each of which have been shown to be
nificantly increased by ChEIs, in those with and without increased two- to fourfold compared with placebo. Weight
Parkinson’s disease, related to central cholinergic activa- loss is also increased threefold; this is discussed above
tion [12, 30, 103, 122]. Tremor is increased sevenfold with under gastrointestinal effects but is probably also related to
ChEI treatment in patients with Alzheimer’s dementia, general effects of ChEIs on fatigue and debility [12, 122].
while tremor and Parkinsonian symptoms increase two- to In older patients with frailty, these effects can be quite
threefold in patients with Parkinson’s disease being treated serious and may precipitate further decline. As mentioned
with dopamine antagonists compared with placebo [12, above, the risk for significant weight loss with ChEIs
103, 117]. essentially doubles for those aged over 85 years compared
with younger patients [118]. Data from pharmacovigilance
3.3.6 Risks of Cholinesterase Inhibitors: Cardiovascular studies have also suggested the risk for adverse drug re-
Effects actions related to ChEIs are twofold higher for those aged
over 85 years than for younger individuals with dementia
Procholinergic medications such as ChEIs can produce [137].
bradycardia through vagal stimulation of the heart [119]. The effect of ChEIs on falls is unclear, as the definition
The degree of bradycardia is not an outcome generally of what constitutes a fall varies considerably among studies
measured in randomized trials of ChEIs. Only one trial and fall-related events are significantly under-reported
reported this outcome and found a statistically significant [54]. As mentioned above, ChEIs are associated with an
50 % increase in bradycardia in patients with MCI [84]. increased risk for syncopal episodes, with some evidence
Several large observational studies have assessed the risk for an increase in syncope-related injuries such as hip
for bradycardia and found a significant association between fractures [12, 54, 136]. However, most falls are not due to
ChEI use and hospital visits for symptomatic bradycardia, syncope and many do not result in injury [138–140]. One
with higher doses of medication associated with higher risk meta-analysis of randomized ChEI trials found no sig-
[134–136]. Those with symptomatic bradycardia were nificant effect on falls, despite a 50 % increase in syncopal
significantly more likely to fall, experience syncope, or episodes compared with placebo [54]. Some observational
need a pacemaker implantation [134–136]. studies of dementia populations have found ChEIs to be
Pooled data from randomized trials in dementia have associated with an increased risk for syncope, falls, and
found a statistically significant increase, by approximately fall-related injuries such as hip fractures [136, 141], while
twofold, in the risk of syncope for ChEI treatment com- other studies have found no effect or reduced risk
pared with placebo (Table 2) [12, 54]. A meta-analysis of [142–144].
Systematic Review of the Risks and Benefits of Dementia Medications

3.3.8 Risks of Cholinesterase Inhibitors: Drug–Drug addition, no statistically significant gain from memantine
Interactions monotherapy on behavioral outcomes in moderate to sev-
ere dementia was observed as measured by the NPI at
Cholinesterase inhibitors could be involved in several 24–28 weeks [56]. When the meta-analysis pooled data on
drug–drug interactions, both because of their complex combination therapy with memantine and ChEIs, no sta-
pharmacodynamic and pharmacokinetic properties, and tistically significant benefit on cognitive, functional, be-
also because elderly patients with dementia are typically havioral, or global outcomes was observed when
receiving multiple concomitant medications [2, 119, 145– memantine was added to ChEI treatment [56].
147]. The use of medications with anticholinergic proper- Data on the effect of memantine in the treatment of mild
ties, such as some antidepressants, antipsychotics, antihis- to moderate Alzheimer’s dementia are minimal. Two meta-
tamines, and bronchodilators, is quite common in the analyses found that memantine treatment for up to
elderly, and the action of either the ChEI or the anti- 6 months in moderate Alzheimer’s disease produced a
cholinergic agent could be inhibited [145, 146]. In addition, marginal benefit in cognitive function (1 point on the
medications with anticholinergic effects could cause cog- 70-point ADAS-cog scale) and in global outcomes (0.1
nitive worsening or delirium in patients with dementia points on a 7-point global assessment of change scale) but
[147]. Observational studies indicate that over one-third of had no effect on mood and behavior [95, 153]. However,
patients on ChEIs also receive at least one anticholinergic when evaluating trials of mild Alzheimer’s disease, no
drug [148, 149]. Drugs with additional cholinesterase in- significant effect of memantine on any outcome was ob-
hibitory activity, such as ranitidine, could theoretically served, either in any single trial or when the trial data were
enhance cholinergic activity and cause greater adverse pooled together [153].
toxicity [145, 146]. The use of ChEIs together with an- Data on the effect of memantine on vascular and Lewy
tiparkinsonian medications could limit the efficacy of ei- body dementia are even more scarce [1, 30, 93–95, 154–
ther drug when treating Parkinson’s disease and dementia 156]. Memantine treatment in vascular dementia for
[147]. In addition, as mentioned above, ChEIs have been 6 months showed minimal improvement in cognitive status
shown to increase tremor in patients with Parkinson’s (approximately 2 points on the ADAS-cog scale) without a
disease two- to threefold [12, 103, 119]. Medications with significant effect on global outcomes or neuropsychiatric
bradycardic effects, such as beta-blockers, digoxin, amio- scores [94, 154, 155]. Pooled trial data of memantine
darone, and calcium-channel blockers, cause vagal treatment in Lewy body dementia found no statistically
stimulation or sympathetic blockade, and may increase the significant effect on cognition, global outcome, or behav-
risk for syncope or heart block [145–147, 150]. ioral symptoms [30, 156–158].

3.4 Memantine 3.4.2 Risks of Memantine

3.4.1 Benefits of Memantine In pooled trial data of memantine, the most common side
effects seen were constipation, dizziness, headache, hy-
Memantine has marketing approval for the treatment of pertension, and somnolence, but there was no statistically
moderate to severe Alzheimer’s dementia in most countries significant difference in the number of withdrawals or pa-
[10, 56, 151, 152]. Studies that have assessed memantine, tients with at least one adverse event between memantine
either as monotherapy or combination therapy with ChEIs, and placebo [93, 95, 159, 160]. A small reduction was
are limited and all are short-term (3–6 months) industry- observed in the incidence of agitation recorded as an ad-
sponsored trials [56, 95]. A recent meta-analysis from the verse event, with a number needed to treat of 17 patients
UK National Institute for Health and Care Excellence for 6 months to prevent one occurrence of agitation [95].
pooled data on trials of memantine monotherapy versus Agitation was not a primary outcome for efficacy of me-
placebo in moderate to severe Alzheimer’s dementia and mantine in these trials, and there was no evidence that
found a small benefit at 3 months of treatment (4 points on agitation or other behavioral problems could be treated
the 133-point SIB scale), but no significant change was with memantine [95]. Memantine had no significant effect
seen by 6 months of treatment [56]. In addition, memantine on falls, weight loss, or confusion [95, 159, 160]. One trial
monotherapy was found to produce a small improvement found a threefold increase in hypertension for memantine
of 0.3 points in a 7-point global assessment of change scale compared with placebo, but other studies did not evaluate
at 24–28 weeks of treatment [56]. However, pooled data this effect [95].
from the meta-analysis showed no significant benefit in Minimal drug–drug interactions have been seen with
functional outcomes as measured by ADL, or by the memantine use [160]. However, little is known about the
Functional Assessment Staging instrument [56]. In use of memantine with other NMDA antagonists, such as
J. S. Buckley, S. R. Salpeter

amantadine, ketamine, and dextromethorphan, so caution is Gastrointestinal effects are common with ChEI treat-
recommended [160, 161]. ment, with a significant increased risk for abdominal
pain, nausea, vomiting, diarrhea, and anorexia, and
clinically significant weight loss. In addition, the rate of
4 Conclusions headache, dizziness, tremor, insomnia, abnormal dreams,
fatigue, asthenia, vertigo, and muscle cramps are higher
This systematic review summarizes the available data on than with placebo. Finally, hospitalization for bradycar-
the risks and benefits of dementia treatments in the elderly. dia and syncope is doubled, and is associated with an
Treatment with ChEIs for mild to moderate Alzheimer’s increased risk for syncope-related conditions such as hip
and Lewy body dementia for durations of up to 1 year fracture. There appears to be a dose-dependence related
produces small statistically-significant improvements in to adverse effects but not efficacy, so treatment with
cognitive, functional, and global outcomes, but whether lower doses may be preferable. It is clear that real-world
these improvements result in clinically relevant effects is conditions are quite different from the controlled trial
unclear. The three available ChEIs have similar efficacy, environment, involving an older, frailer population with
with minimal differences seen between low and high doses more severe disease.
of these agents. Evidence exists that the beneficial effects Memantine monotherapy in moderate to severe Alz-
of ChEIs wane over the course of 1–2 years, and essen- heimer’s and vascular dementia has been shown to have
tially no data is available to evaluate treatment beyond small beneficial effects on cognitive and global function
2 years. No evidence exists for any beneficial effect of in trials of 3–6 months duration, with some waning of
ChEIs in the treatment of vascular dementia, and their use effect by 6 months. In limited pooled trial data, me-
is not recommended for this disease [94]. mantine has not been shown to be effective in the
Trial data on ChEI treatment for severe Alzheimer’s treatment of mild dementia or Lewy body dementia, or
dementia are limited and have variable results, and no as an add-on treatment to ChEIs. From the available
studies have evaluated severe end-stage dementia. This evidence, memantine seems to have a favorable side-
lack of data may indicate a publication bias on the part of effect profile.
the pharmaceutical industry, with the assumption that pa- In summary, ChEIs may provide some cognitive, func-
tients with more severe dementia are less likely to benefit tional, and global benefits in mild to moderate Alzheimer’s
from treatment [11]. Over half of community-dwelling and and Lewy body dementia, but at the risk of significant
roughly 90 % of institutionalized Alzheimer’s patients cholinergic side effects, the most serious being weight loss,
have dementia rated as moderate to severe, so further re- debility, and syncope. These effects could be especially
search on this population is clearly needed [55, 162]. The detrimental in the frail elderly population, in which the
present evidence indicates that ChEIs have minimal benefit risks of treatment outweigh the benefits. The effectiveness
in patients with severe dementia, and discontinuation of of ChEI treatment may be short lived, with minimal evi-
treatment should be considered once the disease becomes dence for benefit over 1 year or in those with more ad-
severe [163, 164]. vanced disease. No significant efficacy of treatment is seen
The limited trial data indicate that ChEIs do not sig- for those with vascular dementia or for those aged over
nificantly improve cognitive or neuropsychiatric outcomes in 85 years. Memantine monotherapy may provide some
patients aged over 85 years. Of note, the prevalence of vas- benefit for patients with moderate to severe Alzheimer’s
cular dementia increases with advancing age, with 70–90 % and vascular dementia, but the benefit is small and may
of those aged over 90 years showing vascular pathology at wane over the course of several months. Memantine has a
autopsy [13]. As ChEIs showed marginal efficacy for the relatively favorable side-effect profile, at least under con-
treatment of vascular dementia, this could explain why ChEIs trolled trial conditions.
have decreased efficacy in older populations. This systematic review provides the available evidence
ChEIs produce side effects through increased choliner- on efficacy and risk of dementia treatments, but leaves a lot
gic stimulation of central and peripheral neural pathways. of uncertainty on how to use these drugs in clinical prac-
Pooled trial data show a twofold increase in withdrawals tice. Good diagnostic criteria for identifying various de-
due to adverse effects compared with placebo, with a mentia types are lacking, and no well-defined markers for
number needed to harm of ten. Individuals aged over evaluating therapeutic response across the spectrum of
85 years have almost double the risk for an adverse event memory disorders exists. How efficacy should be assessed
compared with younger patients. For this older population, for individual patients in order to understand whether
with decreased efficacy of treatment and increased occur- treatment is providing continued benefit or should be
rence of adverse effects, we believe the risks of treatment stopped is unclear. These decisions will need to be made on
outweigh the benefits. a case-by-case basis.
Systematic Review of the Risks and Benefits of Dementia Medications

Compliance with ethical standards No outside funding was used 20. Lanctot KL, Herrmann N, Yau KK, et al. Efficacy and safety of
to complete this research. The authors, Jacob Buckley and Shelley cholinesterase inhibitors in Alzheimer’s disease: a meta-analy-
Salpeter, have no conflicts of interest that are directly relevant to the sis. CMAJ. 2003;169(6):557–64.
content of this study. 21. Chui HC, Mack W, Jackson JE, et al. Clinical criteria for the
diagnosis of vascular dementia: a multicenter study of compa-
rability and interrater reliability. Arch Neurol. 2000;57(2):191–6.
22. Qiu C, Kivipelto M, von Strauss E. Epidemiology of Alzhei-
References mer’s disease: occurrence, determinants, and strategies toward
intervention. Dialogues Clin Neurosci. 2009;11(2):111–28.
1. Qaseem A, Snow V, Cross JT Jr, et al. Current pharmacologic 23. Hebert R, Brayne C. Epidemiology of vascular dementia. Neu-
treatment of dementia: a clinical practice guideline from the roepidemiology. 1995;14(5):240–57.
American College of Physicians and the American Academy of 24. Rocca WA, Kokmen E. Frequency and distribution of vascular
Family Physicians. Ann Intern Med. 2008;148(5):370–8. dementia. Alzheimer Dis Assoc Disord. 1999;13(Suppl
2. Evans JG, Wilcock G, Birks J. Evidence-based pharmacotherapy 3):S9–14.
of Alzheimer’s disease. Int J Neuropsychopharmacol. 2004;7(3): 25. Knopman DS, Parisi JE, Boeve BF, et al. Vascular dementia in a
351–69. population-based autopsy study. Arch Neurol. 2003;60(4):569–
3. American Psychiatric Association. Diagnostic and statistical 75.
manual of mental disorders: DSM-IV-TR. 4th ed. Washington, 26. Skoog I, Nilsson L, Palmertz B, Andreasson LA, Svanborg A. A
DC; 2000. population-based study of dementia in 85-year-olds. N Engl J
4. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. Med. 1993;328(3):153–8.
The global prevalence of dementia: a systematic review and 27. Lobo A, Launer LJ, Fratiglioni L, et al. Prevalence of dementia
metaanalysis. Alzheimers Dement. 2013;9(1):63–75.e2. and major subtypes in Europe: a collaborative study of
5. Bachman DL, Wolf PA, Linn RT, et al. Incidence of dementia population-based cohorts. Neurologic Diseases in the Elderly
and probable Alzheimer’s disease in a general population: the Research Group. Neurology. 2000;54(11 Suppl 5):S4–9.
Framingham Study. Neurology. 1993;43(3 Pt 1):515–9. 28. Fitzpatrick AL, Kuller LH, Ives DG, et al. Incidence and
6. Wimo A, Jonsson L, Bond J, Prince M, Winblad B, Alzheimer prevalence of dementia in the Cardiovascular Health Study.
Disease I. The worldwide economic impact of dementia 2010. J Am Geriatr Soc. 2004;52(2):195–204.
Alzheimers Dement. 2013;9(1):1–11.e3. 29. Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain
7. Russ TC, Morling JR. Cholinesterase inhibitors for mild cog- pathologies account for most dementia cases in community-d-
nitive impairment. Cochrane Database Syst Rev. 2012;9:CD00 welling older persons. Neurology. 2007;69(24):2197–204.
9132. 30. Wang HF, Yu JT, Tang SW, et al. Efficacy and safety of cho-
8. O’Brien JT, Burns A, BAP Dementia Consensus Group. Clinical linesterase inhibitors and memantine in cognitive impairment in
practice with anti-dementia drugs: a revised (second) consensus Parkinson’s disease, Parkinson’s disease dementia, and demen-
statement from the British Association for Psychopharmacology. tia with Lewy bodies: systematic review with meta-analysis and
J Psychopharmacol. 2011;25(8):997–1019. trial sequential analysis. J Neurol Neurosurg Psychiatry.
9. Cummings JL. Challenges to demonstrating disease-modifying 2015;86(2):135–43. doi:10.1136/jnnp-2014-307659.
effects in Alzheimer’s disease clinical trials. Alzheimers De- 31. Aarsland D, Zaccai J, Brayne C. A systematic review of
ment. 2006;2(4):263–71. prevalence studies of dementia in Parkinson’s disease. Mov
10. Drugs for cognitive loss and dementia. Treat Guidel Med Lett. Disord. 2005;20(10):1255–63.
2013;11(134):95–100. 32. Mayeux R, Stern Y, Rosenstein R, et al. An estimate of the
11. Santaguida PS, Raina P, Booker L, et al. Pharmacological prevalence of dementia in idiopathic Parkinson’s disease. Arch
treatment of dementia. Evid Rep Technol Assess (Summ). 2004; Neurol. 1988;45(3):260–2.
97:1–16. 33. Shergil S, Mullan E, D’ath P, Katona C. What is the clinical
12. Birks J. Cholinesterase inhibitors for Alzheimer’s disease. prevalence of Lewy body dementia? Int J Geriatr Psychiatry.
Cochrane Database Syst Rev. 2006;(1):CD005593. 1994;9:907–12.
13. Bullain SS, Corrada MM. Dementia in the oldest old. Con- 34. Vann Jones SA, O’Brien JT. The prevalence and incidence of
tinuum (Minneap Minn). 2013;19(2 Dementia):457–69. dementia with Lewy bodies: a systematic review of population
14. Gill SS, Bronskill SE, Mamdani M, et al. Representation of and clinical studies. Psychol Med. 2014;44(4):673–83.
patients with dementia in clinical trials of donepezil. Can J Clin 35. Chertkow H, Massoud F, Nasreddine Z, et al. Diagnosis and
Pharmacol. 2004;11(2):e274–85. treatment of dementia: 3. Mild cognitive impairment and cognitive
15. Schneider LS, Olin JT, Lyness SA, Chui HC. Eligibility of impairment without dementia. CMAJ. 2008;178(10):1273–85.
Alzheimer’s disease clinic patients for clinical trials. J Am 36. Sheehan B. Assessment scales in dementia. Ther Adv Neurol
Geriatr Soc. 1997;45(8):923–8. Disord. 2012;5(6):349–58.
16. Asahina Y, Sugano H, Sugiyama E, Uyama Y. Representation of 37. Folstein MF, Folstein SE, McHugh PR. ‘‘Mini-mental state’’. A
older patients in clinical trials for drug approval in Japan. J Nutr practical method for grading the cognitive state of patients for
Health Aging. 2014;18(5):520–3. the clinician. J Psychiatr Res. 1975;12(3):189–98.
17. Killin LO, Russ TC, Starr JM, Abrahams S, Della Sala S. The 38. Crutch S. The mini mental state examination (MMSE). Fact
effect of funding sources on donepezil randomised controlled sheet. 2013. http://www.alzheimers.org.uk/site/scripts/download_
trial outcome: a meta-analysis. BMJ Open. 2014;4(4):e004083. info.php?downloadID=1116. Accessed 12 Apr 2015.
18. Gilstad JR, Finucane TE. Results, rhetoric, and randomized 39. Tombaugh TN, Hubley AM, McDowell I, Kristjansson B. Mini-
trials: the case of donepezil. J Am Geriatr Soc. 2008;56(8): mental state examination (MMSE) and the modified MMSE
1556–62. (3MS): a psychometric comparison and normative data. Psychol
19. Rockwood K, MacKnight C. Assessing the clinical importance Assess. 1996;8(1):48–59.
of statistically significant improvement in anti-dementia drug 40. Kahle-Wrobleski K, Corrada MM, Li B, Kawas CH. Sensitivity
trials. Neuroepidemiology. 2001;20(2):51–6. and specificity of the mini-mental state examination for
J. S. Buckley, S. R. Salpeter

identifying dementia in the oldest-old: the 90? study. J Am 59. Schmitt FA, Wichems CH. A systematic review of assessment
Geriatr Soc. 2007;55(2):284–9. and treatment of moderate to severe Alzheimer’s disease. Prim
41. Galea M, Woodward M. Mini-mental state examination Care Companion J Clin Psychiatry. 2006;8(3):158–9.
(MMSE). Aust J Physiother. 2005;51(3):198. 60. Benson AD, Slavin MJ, Tran TT, Petrella JR, Doraiswamy PM.
42. Lourenco RA, Veras RP. Mini-mental state examination: psy- Screening for early Alzheimer’s disease: is there still a role for
chometric characteristics in elderly outpatients. Rev Saude the mini-mental state examination? Prim Care Companion J Clin
Publica. 2006;40(4):712–9. Psychiatry. 2005;7(2):62–9.
43. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alz- 61. Petersen RC. Mild cognitive impairment as a diagnostic entity.
heimer’s disease. Am J Psychiatry. 1984;141(11):1356–64. J Intern Med. 2004;256(3):183–94.
44. Doraiswamy PM, Kaiser L, Bieber F, Garman RL. The Alz- 62. Pozueta A, Rodriguez-Rodriguez E, Vazquez-Higuera JL, et al.
heimer’s Disease Assessment Scale: evaluation of psychometric Detection of early Alzheimer’s disease in MCI patients by the
properties and patterns of cognitive decline in multicenter combination of MMSE and an episodic memory test. BMC
clinical trials of mild to moderate Alzheimer’s disease. Alz- Neurol. 2011;11:78.
heimer Dis Assoc Disord. 2001;15(4):174–83. 63. Voisin T, Vellas B. Diagnosis and treatment of patients with
45. Skinner J, Carvalho JO, Potter GG, et al. The Alzheimer’s dis- severe Alzheimer’s disease. Drugs Aging. 2009;26(2):135–44.
ease assessment scale-cognitive-plus (ADAS-Cog-Plus): an ex- 64. Reisberg B, Jamil IA, Khan S, et al. Staging dementia. In: Abou-
pansion of the ADAS-Cog to improve responsiveness in MCI. Saleh MT, Katona CLE, Kumar A, editors. Principles and
Brain Imaging Behav. 2012;6(4):489–501. practice of geriatric psychiatry. 3rd ed. Chichester, Hoboken:
46. Khan A, Yavorsky C, DiClemente G, et al. Reliability of the Wiley; 2011. p. 162–169.
Alzheimer’s disease assessment scale (ADAS-Cog) in longitu- 65. Frolich L. The cholinergic pathology in Alzheimer’s disease:
dinal studies. Curr Alzheimer Res. 2013;10(9):952–63. discrepancies between clinical experience and pathophysio-
47. Weyer G, Erzigkeit H, Kanowski S, Ihl R, Hadler D. Alz- logical findings. J Neural Transm. 2002;109(7–8):1003–13.
heimer’s Disease Assessment Scale: reliability and validity 66. Francis PT, Perry EK. Cholinergic and other neurotransmitter
in a multicenter clinical trial. Int Psychogeriatr. 1997;9(2): mechanisms in Parkinson’s disease, Parkinson’s disease de-
123–38. mentia, and dementia with Lewy bodies. Mov Disord.
48. Mohs RC, Knopman D, Petersen RC, et al. Development of 2007;22(Suppl 17):S351–7.
cognitive instruments for use in clinical trials of antidementia 67. Grantham C, Geerts H. The rationale behind cholinergic drug
drugs: additions to the Alzheimer’s Disease Assessment Scale treatment for dementia related to cerebrovascular disease.
that broaden its scope. The Alzheimer’s Disease Coop- J Neurol Sci. 2002;203–204:131–6.
erative Study. Alzheimer Dis Assoc Disord. 1997;11(Suppl 2): 68. Molinuevo JL, Llado A, Rami L. Memantine: targeting gluta-
S13–21. mate excitotoxicity in Alzheimer’s disease and other dementias.
49. Panisset M, Roudier M, Saxton J, Boller F. Severe impairment Am J Alzheimers Dis Other Demen. 2005;20(2):77–85.
battery. A neuropsychological test for severely demented pa- 69. Ahmed I, Bose SK, Pavese N, et al. Glutamate NMDA receptor
tients. Arch Neurol. 1994;51(1):41–5. dysregulation in Parkinson’s disease with dyskinesias. Brain.
50. Lawton MP, Brody EM. Assessment of older people: self- 2011;134(Pt 4):979–86.
maintaining and instrumental activities of daily living. Geron- 70. Sabbagh MN, Hake AM, Ahmed S, Farlow MR. The use of
tologist. 1969;9(3):179–86. memantine in dementia with Lewy bodies. J Alzheimers Dis.
51. Robert P, Ferris S, Gauthier S, Ihl R, Winblad B, Tennigkeit F. 2005;7(4):285–9.
Review of Alzheimer’s disease scales: is there a need for a new 71. Mobius HJ. Pharmacologic rationale for memantine in chronic
multi-domain scale for therapy evaluation in medical practice? cerebral hypoperfusion, especially vascular dementia. Alzhei-
Alzheimers Res Ther. 2010;2(4):24. mer Dis Assoc Disord. 1999;13(Suppl 3):S172–8.
52. Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, 72. Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease.
Carusi DA, Gornbein J. The neuropsychiatric inventory: com- Lancet. 2006;368(9533):387–403.
prehensive assessment of psychopathology in dementia. Neu- 73. Nyakas C, Granic I, Halmy LG, Banerjee P, Luiten PG. The
rology. 1994;44(12):2308–14. basal forebrain cholinergic system in aging and dementia. Res-
53. Reisberg B. Global measures: utility in defining and measuring cuing cholinergic neurons from neurotoxic amyloid-beta42 with
treatment response in dementia. Int Psychogeriatr. 2007;19(3): memantine. Behav Brain Res. 2011;221(2):594–603.
421–56. 74. Danysz W, Parsons CG. Alzheimer’s disease, beta-amyloid,
54. Kim DH, Brown RT, Ding EL, Kiel DP, Berry SD. Dementia glutamate, NMDA receptors and memantine–searching for the
medications and risk of falls, syncope, and related adverse connections. Br J Pharmacol. 2012;167(2):324–52.
events: meta-analysis of randomized controlled trials. J Am 75. Iemolo F, Duro G, Rizzo C, Castiglia L, Hachinski V, Caruso C.
Geriatr Soc. 2011;59(6):1019–31. Pathophysiology of vascular dementia. Immun Ageing. 2009;6:13.
55. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. 76. Ni JW, Matsumoto K, Li HB, Murakami Y, Watanabe H.
Alzheimer disease in the US population: prevalence estimates Neuronal damage and decrease of central acetylcholine level
using the 2000 census. Arch Neurol. 2003;60(8):1119–22. following permanent occlusion of bilateral common carotid ar-
56. National Institute for Health and Care Excellence. Donepezil, teries in rat. Brain Res. 1995;673(2):290–6.
galantamine, rivastigmine and memantine for the treatment of 77. Tsuboi Y, Uchikado H, Dickson DW. Neuropathology of
Alzheimer’s disease. NICE technology appraisal guidance 217. Parkinson’s disease dementia and dementia with Lewy bodies
NICE; London; 2011. with reference to striatal pathology. Parkinsonism Relat Disord.
57. Anthony JC, LeResche L, Niaz U, von Korff MR, Folstein MF. 2007;13(Suppl 3):S221–4.
Limits of the ‘Mini-Mental State’ as a screening test for de- 78. Bohnen NI, Albin RL. The cholinergic system and Parkinson
mentia and delirium among hospital patients. Psychol Med. disease. Behav Brain Res. 2011;221(2):564–73.
1982;12(2):397–408. 79. Grothe MJ, Schuster C, Bauer F, Heinsen H, Prudlo J, Teipel SJ.
58. O’Bryant SE, Humphreys JD, Smith GE, et al. Detecting de- Atrophy of the cholinergic basal forebrain in dementia with
mentia with the mini-mental state examination in highly Lewy bodies and Alzheimer’s disease dementia. J Neurol.
educated individuals. Arch Neurol. 2008;65(7):963–7. 2014;261(10):1939–48.
Systematic Review of the Risks and Benefits of Dementia Medications

80. Dalfo E, Albasanz JL, Martin M, Ferrer I. Abnormal 100. Courtney C, Farrell D, Gray R, et al. Long-term donepezil
metabotropic glutamate receptor expression and signaling in the treatment in 565 patients with Alzheimer’s disease (AD2000):
cerebral cortex in diffuse Lewy body disease is associated with randomised double-blind trial. Lancet. 2004;363(9427):
irregular alpha-synuclein/phospholipase C (PLCbeta1) interac- 2105–15.
tions. Brain Pathol. 2004;14(4):388–98. 101. Schneider LS, Sano M. Current Alzheimer’s disease clinical
81. Starr MS. Glutamate/dopamine D1/D2 balance in the basal trials: methods and placebo outcomes. Alzheimers Dement.
ganglia and its relevance to Parkinson’s disease. Synapse. 2009;5(5):388–97.
1995;19(4):264–93. 102. Lee PE, Fischer HD, Rochon PA, et al. Published randomized
82. Petersen RC, Thomas RG, Grundman M, et al. Vitamin E and controlled trials of drug therapy for dementia often lack com-
donepezil for the treatment of mild cognitive impairment. plete data on harm. J Clin Epidemiol. 2008;61(11):1152–60.
N Engl J Med. 2005;352(23):2379–88. 103. Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase
83. Lu PH, Edland SD, Teng E, et al. Donepezil delays progression inhibitors for dementia with Lewy bodies, Parkinson’s disease
to AD in MCI subjects with depressive symptoms. Neurology. dementia and cognitive impairment in Parkinson’s disease.
2009;72(24):2115–21. Cochrane Database Syst Rev. 2012;(3):CD006504.
84. Winblad B, Gauthier S, Scinto L, et al. Safety and efficacy of 104. Schneider LS. Assessing outcomes in Alzheimer disease. Alz-
galantamine in subjects with mild cognitive impairment. Neu- heimer Dis Assoc Disord. 2001;15(Suppl 1):S8–18.
rology. 2008;70(22):2024–35. 105. Winblad B, Kilander L, Eriksson S, et al. Donepezil in pa-
85. Tricco AC, Soobiah C, Berliner S, et al. Efficacy and safety of tients with severe Alzheimer’s disease: double-blind, parallel-
cognitive enhancers for patients with mild cognitive impair- group, placebo-controlled study. Lancet. 2006;367(9516):
ment: a systematic review and meta-analysis. CMAJ. 2013;185 1057–65.
(16):1393–401. 106. Black SE, Doody R, Li H, et al. Donepezil preserves cognition
86. Cooper C, Li R, Lyketsos C, Livingston G. Treatment for mild and global function in patients with severe Alzheimer disease.
cognitive impairment: systematic review. Br J Psychiatry. Neurology. 2007;69(5):459–69.
2013;203(3):255–64. 107. Homma A, Imai Y, Tago H, et al. Donepezil treatment of pa-
87. Doody RS, Ferris S, Salloway S, et al. Safety and tolerability of tients with severe Alzheimer’s disease in a Japanese population:
donepezil in mild cognitive impairment: open-label extension results from a 24-week, double-blind, placebo-controlled, ran-
study. Am J Alzheimers Dis Other Demen. 2010;25(2):155–9. domized trial. Dement Geriatr Cogn Disord. 2008;25(5):
88. Feldman HH, Ferris S, Winblad B, et al. Effect of rivastigmine 399–407.
on delay to diagnosis of Alzheimer’s disease from mild cogni- 108. Burns A, Bernabei R, Bullock R, et al. Safety and efficacy of
tive impairment: the InDDEx study. Lancet Neurol. 2007;6(6): galantamine (Reminyl) in severe Alzheimer’s disease (the
501–12. SERAD study): a randomised, placebo-controlled, double-blind
89. Russ TC. Cholinesterase inhibitors should not be prescribed for trial. Lancet Neurol. 2009;8(1):39–47.
mild cognitive impairment. Evid Based Med. 2014;19(3):101. 109. Feldman H, Gauthier S, Hecker J, et al. Efficacy and safety of
90. Gauthier S, Touchon J. Mild cognitive impairment is not a donepezil in patients with more severe Alzheimer’s disease: a
clinical entity and should not be treated. Arch Neurol. subgroup analysis from a randomized, placebo-controlled trial.
2005;62(7):1164–6 (discussion 1167). Int J Geriatr Psychiatry. 2005;20(6):559–69.
91. Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of choli- 110. Ballard C, Margallo-Lana M, Juszczak E, et al. Quetiapine and
nesterase inhibitors in the treatment of neuropsychiatric symp- rivastigmine and cognitive decline in Alzheimer’s disease:
toms and functional impairment in Alzheimer disease: a meta- randomised double blind placebo controlled trial. BMJ.
analysis. JAMA. 2003;289(2):210–6. 2005;330(7496):874.
92. Takeda A, Loveman E, Clegg A, et al. A systematic review of 111. Cummings J, Jones R, Wilkinson D, et al. Effect of donepezil on
the clinical effectiveness of donepezil, rivastigmine and galan- cognition in severe Alzheimer’s disease: a pooled data analysis.
tamine on cognition, quality of life and adverse events in Alz- J Alzheimers Dis. 2010;21(3):843–51.
heimer’s disease. Int J Geriatr Psychiatry. 2006;21(1):17–28. 112. Froelich L, Andreasen N, Tsolaki M, et al. Long-term treatment
93. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cho- of patients with Alzheimer’s disease in primary and secondary
linesterase inhibitors and memantine for treating dementia: care: results from an international survey. Curr Med Res Opin.
evidence review for a clinical practice guideline. Ann Intern 2009;25(12):3059–68.
Med. 2008;148(5):379–97. 113. Winblad B, Engedal K, Soininen H, et al. A 1-year, randomized,
94. Kavirajan H, Schneider LS. Efficacy and adverse effects of placebo-controlled study of donepezil in patients with mild to
cholinesterase inhibitors and memantine in vascular dementia: a moderate AD. Neurology. 2001;57(3):489–95.
meta-analysis of randomised controlled trials. Lancet Neurol. 114. Mohs RC, Doody RS, Morris JC, et al. A 1-year, placebo-con-
2007;6(9):782–92. trolled preservation of function survival study of donepezil in
95. McShane R, Areosa Sastre A, Minakaran N. Memantine for AD patients. Neurology. 2001;57(3):481–8.
dementia. Cochrane Database Syst Rev. 2006;(2):CD003154. 115. Hager K, Baseman AS, Nye JS, et al. Effects of galantamine in a
96. Hansen RA, Gartlehner G, Webb AP, Morgan LC, Moore CG, 2-year, randomized, placebo-controlled study in Alzheimer’s
Jonas DE. Efficacy and safety of donepezil, galantamine, and ri- disease. Neuropsychiatr Dis Treat. 2014;10:391–401.
vastigmine for the treatment of Alzheimer’s disease: a systematic 116. Birks J, McGuinness B, Craig D. Rivastigmine for vascular
review and meta-analysis. Clin Interv Aging. 2008;3(2):211–25. cognitive impairment. Cochrane Database Syst Rev. 2013;5:
97. Ellis JM. Cholinesterase inhibitors in the treatment of dementia. CD004744.
J Am Osteopath Assoc. 2005;105(3):145–58. 117. Pagano G, Rengo G, Pasqualetti G, et al. Cholinesterase in-
98. Hogan DB, Goldlist B, Naglie G, Patterson C. Comparison hibitors for Parkinson’s disease: a systematic review and meta-
studies of cholinesterase inhibitors for Alzheimer’s disease. analysis. J Neurol Neurosurg Psychiatry. 2014.
Lancet Neurol. 2004;3(10):622–6. 118. Tariot PN, Cummings JL, Katz IR, et al. A randomized, double-
99. Nordberg A, Svensson AL. Cholinesterase inhibitors in the blind, placebo-controlled study of the efficacy and safety of
treatment of Alzheimer’s disease: a comparison of tolerability donepezil in patients with Alzheimer’s disease in the nursing
and pharmacology. Drug Saf. 1998;19(6):465–80. home setting. J Am Geriatr Soc. 2001;49(12):1590–9.
J. S. Buckley, S. R. Salpeter

119. Thompson S, Lanctot KL, Herrmann N. The benefits and risks 137. Pariente A, Sanctussy DJ, Miremont-Salame G, et al. Factors
associated with cholinesterase inhibitor therapy in Alzheimer’s associated with serious adverse reactions to cholinesterase in-
disease. Expert Opin Drug Saf. 2004;3(5):425–40. hibitors: a study of spontaneous reporting. CNS Drugs.
120. Inglis F. The tolerability and safety of cholinesterase inhibitors 2010;24(1):55–63.
in the treatment of dementia. Int J Clin Pract Suppl. 138. Brignole M. Distinguishing syncopal from non-syncopal causes
2002;127:45–63. of fall in older people. Age Ageing. 2006;35(Suppl 2):ii46–50.
121. Wilkinson DG, Francis PT, Schwam E, Payne-Parrish J. Cho- 139. McIntosh S, Da Costa D, Kenny RA. Outcome of an integrated
linesterase inhibitors used in the treatment of Alzheimer’s dis- approach to the investigation of dizziness, falls and syncope in
ease: the relationship between pharmacological effects and elderly patients referred to a ‘syncope’ clinic. Age Ageing.
clinical efficacy. Drugs Aging. 2004;21(7):453–78. 1993;22(1):53–8.
122. Gauthier S. Cholinergic adverse effects of cholinesterase in- 140. Shaw FE, Kenny RA. The overlap between syncope and falls in
hibitors in Alzheimer’s disease: epidemiology and management. the elderly. Postgrad Med J. 1997;73(864):635–9.
Drugs Aging. 2001;18(11):853–62. 141. French DD, Campbell R, Spehar A, Cunningham F, Bulat T,
123. Corey-Bloom J, Anand R, Veach J. A randomized trial Luther SL. Drugs and falls in community-dwelling older people:
evaluating the efficacy and safety of ENA 713 (rivastigmine a national veterans study. Clin Ther. 2006;28(4):619–30.
tartrate), a new acetylcholinesterase inhibitor, in patients with 142. Tamimi I, Ojea T, Sanchez-Siles JM, et al. Acetylcholinesterase
mild-to-moderately severe Alzheimer’s disease. Int J Geriatr inhibitors and the risk of hip fracture in Alzheimer’s disease
Psychopharmacol. 1998;1:55–65. patients: a case-control study. J Bone Miner Res. 2012;27(7):
124. Rosler M, Anand R, Cicin-Sain A, et al. Efficacy and safety of 1518–27.
rivastigmine in patients with Alzheimer’s disease: international 143. Chung KA, Lobb BM, Nutt JG, Horak FB. Effects of a central
randomised controlled trial. BMJ. 1999;318(7184):633–8. cholinesterase inhibitor on reducing falls in Parkinson disease.
125. Tariot PN, Solomon PR, Morris JC, Kershaw P, Lilienfeld S, Neurology. 2010;75(14):1263–9.
Ding C. A, 5-month, randomized, placebo-controlled trial of 144. Kallin K, Gustafson Y, Sandman PO, Karlsson S. Drugs and
galantamine in AD. The Galantamine USA-10 Study Group. falls in older people in geriatric care settings. Aging Clin Exp
Neurology. 2000;54(12):2269–76. Res. 2004;16(4):270–6.
126. White H, Pieper C, Schmader K. The association of weight 145. Tavassoli N, Sommet A, Lapeyre-Mestre M, Bagheri H, Mon-
change in Alzheimer’s disease with severity of disease and trastruc JL. Drug interactions with cholinesterase inhibitors: an
mortality: a longitudinal analysis. J Am Geriatr Soc. analysis of the French pharmacovigilance database and a com-
1998;46(10):1223–7. parison of two national drug formularies (Vidal, British National
127. Keller HH, Ostbye T, Goy R. Nutritional risk predicts quality of Formulary). Drug Saf. 2007;30(11):1063–71.
life in elderly community-living Canadians. J Gerontol A Biol 146. Bentue-Ferrer D, Tribut O, Polard E, Allain H. Clinically sig-
Sci Med Sci. 2004;59(1):68–74. nificant drug interactions with cholinesterase inhibitors: a guide
128. Crogan NL, Pasvogel A. The influence of protein-calorie mal- for neurologists. CNS Drugs. 2003;17(13):947–63.
nutrition on quality of life in nursing homes. J Gerontol A Biol 147. Seritan AL. Prevent drug-drug interactions with cholinesterase
Sci Med Sci. 2003;58(2):159–64. inhibitors: avoid adverse events when prescribing medications
129. Payette H, Coulombe C, Boutier V, Gray-Donald K. Nutrition for patients with dementia. Curr Psychiatry. 2008;7(2):57–67.
risk factors for institutionalization in a free-living functionally 148. Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The concurrent
dependent elderly population. J Clin Epidemiol. use of anticholinergics and cholinesterase inhibitors: rare event or
2000;53(6):579–87. common practice? J Am Geriatr Soc. 2004;52(12):2082–7.
130. Sullivan DH, Morley JE, Johnson LE, et al. The GAIN (Geri- 149. Roe CM, Anderson MJ, Spivack B. Use of anticholinergic
atric Anorexia Nutrition) registry: the impact of appetite and medications by older adults with dementia. J Am Geriatr Soc.
weight on mortality in a long-term care population. J Nutr 2002;50(5):836–42.
Health Aging. 2002;6(4):275–81. 150. Paulison B, Leos CL. Potential cardiotoxic reaction involving
131. Stewart JT, Gorelik AR. Involuntary weight loss associated with rivastigmine and beta-blockers: a case report and review of the
cholinesterase inhibitors in dementia. J Am Geriatr Soc. literature. Cardiovasc Toxicol. 2010;10(4):306–10.
2006;54(6):1013–4. 151. Agency EM. Ebixa. European Public Assessment Report.2004.
132. Gallini A, Sommet A, Salandini AM, Veyssiere P, Montastruc www.emea.eu.int/humandocs/Humans/EPAR/ebixa/ebixa.htm.
JL, Montastruc JL. Weight-loss associated with anti-dementia Accessed 12 Apr 2015.
drugs in a patient with Parkinson’s disease. Mov Disord. 152. FDA approves memantine drug for treating AD. Am J Alzhei-
2007;22(13):1980–1. mers Dis Other Demen. 2003;18(6):329–30.
133. Dunn NR, Pearce GL, Shakir SA. Adverse effects associated 153. Schneider LS, Dagerman KS, Higgins JP, McShane R. Lack of
with the use of donepezil in general practice in England. J Psy- evidence for the efficacy of memantine in mild Alzheimer dis-
chopharmacol. 2000;14(4):406–8. ease. Arch Neurol. 2011;68(8):991–8.
134. Hernandez RK, Farwell W, Cantor MD, Lawler EV. Cho- 154. Wilcock G, Mobius HJ, Stoffler A, Group MMM. A double-
linesterase inhibitors and incidence of bradycardia in patients blind, placebo-controlled multicentre study of memantine in
with dementia in the veterans affairs New England healthcare mild to moderate vascular dementia (MMM500). Int Clin Psy-
system. J Am Geriatr Soc. 2009;57(11):1997–2003. chopharmacol. 2002;17(6):297–305.
135. Park-Wyllie LY, Mamdani MM, Li P, Gill SS, Laupacis A, 155. Orgogozo JM, Rigaud AS, Stoffler A, Mobius HJ, Forette F.
Juurlink DN. Cholinesterase inhibitors and hospitalization for Efficacy and safety of memantine in patients with mild to
bradycardia: a population-based study. PLoS Med. 2009;6(9): moderate vascular dementia: a randomized, placebo-controlled
e1000157. trial (MMM 300). Stroke. 2002;33(7):1834–9.
136. Gill SS, Anderson GM, Fischer HD, et al. Syncope and its 156. Emre M, Tsolaki M, Bonuccelli U, et al. Memantine for patients
consequences in patients with dementia receiving cholinesterase with Parkinson’s disease dementia or dementia with Lewy
inhibitors: a population-based cohort study. Arch Intern Med. bodies: a randomised, double-blind, placebo-controlled trial.
2009;169(9):867–73. Lancet Neurol. 2010;9(10):969–77.
Systematic Review of the Risks and Benefits of Dementia Medications

157. Leroi I, Overshott R, Byrne EJ, Daniel E, Burns A. Randomized 161. Stahl SM. Essential psychopharmacology: the prescriber’s
controlled trial of memantine in dementia associated with guide. Cambridge: Cambridge University Press; 2005.
Parkinson’s disease. Mov Disord. 2009;24(8):1217–21. 162. Lindsay J. Patterns of caring for people with dementia in Canada. The
158. Aarsland D, Ballard C, Walker Z, et al. Memantine in patients Canadian study of health and aging. Can J Aging. 1994;13:470–87.
with Parkinson’s disease dementia or dementia with Lewy 163. Molino I, Colucci L, Fasanaro AM, Traini E, Amenta F. Effi-
bodies: a double-blind, placebo-controlled, multicentre trial. cacy of memantine, donepezil, or their association in moderate-
Lancet Neurol. 2009;8(7):613–8. severe Alzheimer’s disease: a review of clinical trials. Scien-
159. Jones RW. A review comparing the safety and tolerability of tificWorldJournal. 2013;2013:925702.
memantine with the acetylcholinesterase inhibitors. Int J Geriatr 164. Shega JW, Ellner L, Lau DT, Maxwell TL. Cholinesterase in-
Psychiatry. 2010;25(6):547–53. hibitor and N-methyl-D-aspartic acid receptor antagonist use in
160. Mimica N, Presecki P. Side effects of approved antidementives. older adults with end-stage dementia: a survey of hospice
Psychiatr Danub. 2009;21(1):108–13. medical directors. J Palliat Med. 2009;12(9):779–83.

You might also like