You are on page 1of 5

The Journal of Nutrition, Health & A ging©

V olume 14, Number 9, 2010

MEMA NTINE BENEFITS FUNCTIONA L A BILITIES IN MODERA TE TO SEV ERE A LZHEIMER’S DISEA SE

MEMANTINE BENEFITS FUNCTIONAL ABILITIES IN MODERATE


TO SEVERE ALZHEIMER’S DISEASE
B. WINBLAD1, S. GAUTHIER2, D. ÅSTRÖM3, K. STENDER3
1. Karolinska Institutet, Alzheimer Disease Research Center (KI-ADRC) NOVUM, Huddinge, Sweden; 2. McGill Center for Studies in Aging, Douglas Institute for Mental Health,
Montreal, Quebec, Canada; 3. H. Lundbeck A/S, Copenhagen, Denmark. Corresponding author: Bengt Winblad, Karolinska Institutet, Alzheimer Disease Research Center (KI-ADRC)
NOVUM, Floor 5, S-14157 Huddinge, Sweden, Phone: +46 8 585 85474, Fax: +46 8 585 83610, E-mail: bengt.winblad@ki.se

Abstract: Objective: Functional abilities are severely impacted in Alzheimer’s disease (AD). Loss of the ability to
perform complex (instrumental) and basic activities of daily living (ADL), leads to decreased independence and
increased caregiver burden. This post-hoc analysis investigated the effect of memantine (20 mg/day) on ADLs, as
measured by Alzheimer’s Disease Cooperative Study-Activities of Daily Living 19-item (ADCS-ADL19) and 23-
item (ADCS-ADL23) scales, in patients with moderate-to-severe AD. Design: Data were pooled from six
multicenter, randomized, placebo-controlled, double-blind, 6-month studies of memantine 20 mg/day.
Participants: Male and female patients aged ≥50 years at baseline with a Mini Mental State Examination (MMSE)
score <20. Measurements: ADCS-ADL19 and ADCS-ADL23 scales were pooled, and 14 shared items, with a
score range of 0-45, were identified and included in the analysis (ADL14). Basic ADLs (BADLs) were defined
as: eating, walking, toileting, bathing, and grooming. Instrumental ADLs (IADLs) were defined as: using a
telephone, watching television, conversing, clearing a table, finding belongings, obtaining a beverage, disposing
of household rubbish, travelling outside the house, and being left alone. Changes from baseline on single-item,
BADL (range: 0–15), IADL (range: 0–30), and total ADL14 scores were analysed for observed cases using
ANCOVA, with study, center and treatment as categorical explanatory variables and score at baseline as a
covariate. Results: 959 patients were treated with memantine and 867 patients received placebo. Memantine-
treated patients had less decline from baseline on the ADL14 total score, compared with placebo (p<0.001) at
study end. Memantine also showed lower reductions in BADLs (p<0.05) and IADLs (p<0.001), for observed
cases, compared with placebo. Memantine-treated patients showed less worsening than placebo recipients for the
ADL items: toileting (p<0.01), grooming (p<0.01), finding belongings (p<0.01), and travelling outside the house
(p<0.05). Conclusion: Compared with placebo, memantine shows benefits for both basic and instrumental ADLs
in patients with moderate-to-severe AD, suggesting that memantine treatment may lead to a more interactive and
dignified life for patients with moderate-to-severe AD.

Key words: Memantine, Alzheimer’s disease, functional ability.

Introduction towards the major burden for carers (6).


Treatment interventions that enable a patient with AD to
Alzheimer’s disease (AD) is a progressive neurodegenerative retain some ability to perform activities of daily living and to
condition characterized by memory impairment, deterioration of preserve their independence for longer, may prevent increases
language, other cognitive deficits, and behavioural problems in patient distress and caregiver burden that would ultimately
such as confusion, irritability, aggression, mood swings and lead to patient institutionalisation. Memantine, a specific,
general withdrawal (1). In addition, functional impairments uncompetitive N-methyl-d-aspartate (NMDA) receptor
include an inability to perform instrumental daily activities and antagonist with moderate affinity and rapid voltage-dependent
basic daily activities (2). It is the single most common cause of kinetics, acts by targeting the neurotransmitter (glutamate)
dementia, and in Europe, accounts for approximately 55-80% of system central to the pathology of AD (7, 8). Memantine has
all cases of dementia (3). been approved in Europe and the USA for the treatment of
Progression of AD is defined by increasing functional and patients with moderate-to-severe AD, and is the only treatment
behavioural impairments, which, in the moderate to severe in clinical use for AD that specifically targets the glutamatergic
stages of the disease, can result in total dependence on a system.
caregiver and a high risk of institutionalisation (4, 5). Thus, a In clinical studies of outpatients with AD, memantine had
patient’s functional performance gradually progresses from an significant clinical effects on global status, cognitive
inability to perform instrumental daily activities, such as (including language, memory, orientation, praxis, and
telephoning and travelling alone, to an eventual decline in the visuospatial ability), functional, and behavioural (including
basic daily activities of eating, walking or grooming (2). symptoms of delusions, agitation/aggression and
Deterioration in functional performance can be measured over irritability/lability) outcomes, and was well tolerated with an
the space of 6-12 months (2). This loss of functional ability is adverse event profile similar to placebo (9-12). Furthermore, in
also one of the biggest problems for caregivers and contributes nursing home patients with severe dementia (AD or vascular

770
Received December 23, 2009
A ccepted for publication January 21, 2010
The Journal of Nutrition, Health & A ging©
V olume 14, Number 9, 2010

JNHA : CLINICA L TRIA LS A ND A GING

dementia (VaD)), memantine improved activities of daily authorized representatives, provided written informed consent
living, care dependence and behavioural symptoms compared prior to participation.
with placebo (13). For these reasons, memantine has the Study design and patient populations have been described in
potential to improve the functional status of patients to a detail elsewhere (17-22). All patients were diagnosed with
greater extent than that seen with donepezil, for which there is probable AD according to the NINCDS-ADRDA criteria (23,
conflicting evidence in relation to its benefits on activities of 24). Briefly, study participants were male or female outpatients
daily living in severe AD (14-16). aged ≥50 years at baseline. In all studies, patients received an
Six randomized, placebo controlled trials have assessed the initial memantine dose of 5 mg/day (or placebo), which was
efficacy and safety of memantine 20 mg/day (17-22) and the titrated in 5 mg/day increments at weekly intervals to 20
data on cognitive outcomes, behavioural symptoms and safety mg/day. Concomitant psychotropic medications were permitted
and tolerability from these trials have been assessed in detail in in five studies (17, 18, 20-22), but not in one study (19).
meta-analyses (9, 10, 12). The information on functional
performance from these studies has not been assessed in the Efficacy outcome measures
same way, and therefore, in this post hoc analysis, data from Functional abilities were assessed in all six studies (17-22)
these six studies were pooled to evaluate the effects of treatment using the AD Cooperative Study-Activities of Daily Living 19-
on functional performance in patients with moderate-to-severe item (ADCS-ADL19) or 23-item (ADCS-ADL23) structured
AD. questionnaires at baseline, and after 4, 12 and 24 weeks. The
ADCS-ADL19 is used for patients with moderate to severe AD
Methods and the ADCS-ADL23 for patients with mild to moderate AD
Study design and patient population (11). Each item on these scales evaluates a patient’s ability to
Data were pooled from six multicenter, randomized, perform an activity of daily living. Caregivers assess a patient’s
placebo-controlled, double-blind, 6-month studies of performance during the previous four weeks; increasing scores
indicate an improvement in functional ability, with a total score
memantine 20 mg/day (Table 1). All English-language
of 54 (for ADCS-ADL19) or 78 (for ADCS-ADL23) indicating
published double-blind, randomized, placebo-controlled, phase
optimal performance. In the present pooled analysis, mean
III studies of memantine with a minimum duration of 24 weeks
change in ADL score from baseline was assessed for 14
were included in the analysis. All patients, or their legally
Table 1
Summary of the six memantine (20 mg/day) randomized, double-blind, placebo-controlled studies included in this analysis

Study MMSE inclusion range (mean) Duration and other details Patients Efficacy measures

Peskind et al (2006) 10-22 (17.3) – mild to moderate 24 weeks 403 patients ADAS-cog,
Twice-daily memantine P: 202 ADCS-ADL23,
M: 201 CIBIC-Plus, NPI
Porsteinsson et al (2008) 10-22 (16.9) – mild to moderate 24 weeks, patients already 433 patients ADCS-ADL23,
receiving a cholinesterase P: 216 CIBIC-Plus,
inhibitor M: 217 MMSE, NPI
Once-daily memantine
Bakchine and Loft (2008) 11-23 (18.7) – mild to moderate 24 weeks 470 patients ADAS-cog,
Twice-daily memantine P: 152 ADCS-ADL23,
M: 318 CIBIC-Plus, NPI
Reisberg et al (2003) 3-14 (7.9) – moderate to severe 28 weeks 252 patients SIB,
Twice-daily memantine P: 126 ADCS-ADL19,
M:126 CIBIC-Plus, FAST,
GDS, MMSE, NPI
Van Dyck et al (2007) 5-14 (10.1) – moderate to severe 24 weeks 350 patients SIB,
Twice-daily memantine P: 172 ADCS-ADL19,
M: 178 BGP, CIBIC-Plus,
FAST, NPI
Tariot et al (2004) 5-14 (10.0) – moderate to severe 24 weeks, patients already 403 patients ADCS-ADL19,
receiving donepezil P: 201 BGP, CIBIC-Plus,
Twice-daily memantine M: 202 NPI,
SIB

P = placebo; M = memantine; ADAS-cog = Alzheimer’s Disease Assessment Scale Cognitive Subscale; SIB = Severe Impairment Battery; ADCS-ADL19 = 19-item AD Cooperative
Study-Activities of Daily Living Inventory; ADCS-ADL23 = 23-item AD Cooperative Study-Activities of Daily Living Inventory; BGP = Behavioural Rating Scale for Geriatric Patients;
CIBIC-plus = Clinician’s Interview-Based Impression of Change Plus Caregiver Input; FAST = Functional Assessment Staging; GDS = Global Deterioration Scale; NPI =
Neuropsychiatric Inventory. Cholinesterase inhibitors were donepezil , rivastigmine, or galantamine.

771
The Journal of Nutrition, Health & A ging©
V olume 14, Number 9, 2010

MEMA NTINE BENEFITS FUNCTIONA L A BILITIES IN MODERA TE TO SEV ERE A LZHEIM ER’S DISEA SE

overlapping items on the ADCS-ADL 19 and ADCS-ADL 23 with memantine. Significantly less worsening of individual
scales (for the purposes of this paper, referred to as ADL14), ADL scores (two BADL single-item scores [toileting and
because three studies recruited patients with mild-to-moderate grooming], and two IADL single-item scores [finding
AD and therefore used the ADCS-ADL23 (17, 18, 22) and three belongings and travelling outside the house]) were also recorded
studies recruited patients with moderate-to-severe AD and at week 24 in memantine recipients, compared with placebo
therefore used the ADCS-ADL19 (19-21). The score range for (Figure 3). The LOCF analysis showed similar significant
the ADL14 was 0-45. The basic ADLs (BADLs) comprised the differences, except the difference between groups for the
items eating (score range 0–3), walking (0–3), toileting (0–3), travelling outside the house item was not significant (p=0.14).
bathing (0–3), and grooming (0–3), and the instrumental ADLs Conversing worsened more in the placebo-group than in the
(IADLs) were the remaining non-basic items, which were using memantine-treated group, but the difference did not reach
a telephone (0–5), watching television (0–3), conversing (0–3), statistical significance (p=0.06 in OC analysis and p=0.08 in
clearing a table (0–3), finding belongings (0–3), obtaining a LOCF analysis).
beverage (0–3), disposing of household rubbish (0–3),
travelling outside the house (0–4), and being left alone (0–3). Table 2
For each item, scores ranged from 0 = ‘least able to perform Patient demographics and clinical characteristics at baseline
task’ to 3 or 4 = ‘most able to perform task’. (FAS; MMSE <20)
Statistical analyses
Placebo (n=867) Memantine
Efficacy analyses were conducted on the patients from the (n=959)
full analysis set (FAS; all patients who received at least one
dose of study medication and had at least one post-baseline Women, n (%) 550 (63.4) 644 (67.2)
efficacy assessment) with Mini Mental State Examination Mean (±SD) age, years 76.2 ± 8.3 76.2 ± 8.1
(MMSE) score <20 (moderate-to-severe AD), using an Caucasian ethnicity, n (%) 788 (90.9) 865 (90.2)
MMSE score, mean ± SD 12.2 ± 4.1 12.3 ± 4.2
observed-case (OC) approach and a last-observation-carried ADL19 total score, mean ± SD 33.1 ± 10.7 (n=499) 32.5 ± 10.7 (n=506)
forward (LOCF) approach to support the OC analyses. The ADL23 total score, mean ± SD 52.7 ± 13.2 (n=368) 51.3 ± 14.7 (n=453)
efficacy endpoints were the change from baseline in ADL14 ADL14 total score, mean ± SD 29.3 ± 8.5 (n=850) 29.0 ± 8.7 (n=938)
total score, BADL score and IADL score, and change in ADL14 Basic ADLs 12.4 ± 2.9 (n=850) 12.4 ± 3.0 (n=938)
single-item scores. Differences between memantine and Eating 2.7 ± 0.5 (n=850) 2.7 ± 0.5 (n=938)
placebo were analyzed within the OC and LOCF populations Walking 2.9 ± 0.5 (n=849) 2.8 ± 0.5 (n=938)
Toileting 2.6 ± 0.8 (n=850) 2.6 ± 0.8 (n=938)
using analysis of covariance (ANCOVA), with study, center, Bathing 2.0 ± 1.0 (n=850) 2.0 ± 1.0 (n=938)
and treatment as categorical explanatory variables and score at Grooming 2.3 ± 1.0 (n=849) 2.2 ± 1.0 (n=937)
baseline as a covariate. All efficacy analyses were tested at the Instrumental ADLs 16.8 ± 6.2 (n=850) 16.6 ± 6.3 (n=938)
two-sided 95% level of significance. Using a telephone 2.1 ± 1.3 (n=850) 2.1 ± 1.3 (n=938)
Watching television 1.0 ± 1.0 (n=835) 1.0 ± 1.0 (n=925)
Results Conversing
Clearing a table
2.1 ± 1.2 (n=850)
2.3 ± 1.1 (n=850)
2.1 ± 1.1 (n=937)
2.2 ± 1.2 (n=938)
Finding belongings 1.9 ± 1.1 (n=850) 1.9 ± 1.1 (n=938)
Baseline characteristics Obtaining a beverage 1.6 ± 1.2 (n=850) 1.6 ± 1.2 (n=937)
The FAS consisted of the 959 memantine-treated patients Disposing of litter 2.4 ± 1.0 (n=850) 2.4 ± 1.1 (n=938)
and 867 placebo recipients who had baseline MMSE scores of Travelling outside the house 2.4 ± 0.9 (n=850) 2.3 ± 0.9 (n=938)
Being left alone 1.5 ± 1.2 (n=540) 1.6 ± 1.2 (n=619)
less than 20. Baseline demographic and clinical characteristics,
including scores for the ADCS-ADL19, ADCS-ADL23, and ADL19 and ADL23 = Alzheimer’s Disease Cooperative Study-Activities of Daily Living 19
ADL14, were similar in the two treatment groups (Table 2). and 23 item scales; ADL14 = overlapping items on the ADL19 and ADL23 scales; MMSE =
Mini Mental State Examination.

Functional efficacy
Memantine-treated patients had smaller reductions (i.e., less Discussion
worsening) in scores on the ADL14 than placebo recipients.
In this pooled post hoc analysis of six 6-month studies of
Differences in functional decline between memantine-treated
patients with moderate to severe AD (MMSE<20), memantine
and placebo-treated patients increased over time (Figure 1).
(20 mg/day) treatment provided statistically significant benefits
BADLs and IADLs decreased by less in the memantine- in overall functional ability and activities of daily living
treated patients than in placebo-treated patients (Figure 2). The compared with placebo. These results confirm the initial, less
results from the LOCF analysis were consistent with those of comprehensive, report of better functional ability with
the OC analysis: at week 4 there were no significant differences memantine than placebo (11), and suggest that memantine
in the BADL or IADL scores in the two LOCF patient groups, treatment may help patients maintain a more interactive life.
but by week 12 IADL scores were significantly less reduced in The ADCS-ADL19 and ADCS-ADL23 are reliable, sensitive
memantine-treated patients than placebo-treated patients, and by and validated instruments for assessing functional change in
week 24 both BADL and IADL were significantly less reduced patients with moderate to severe AD (2).

772
The Journal of Nutrition, Health & A ging©
V olume 14, Number 9, 2010

JNHA : CLINICA L TRIA LS A ND A GING

Figure 1 Figure 3
Mean change from baseline in ADL14 total score, at Weeks 4, 12 Mean change from baseline in ADL single-item scores at Week
and 24/28 (observed cases) and at Week 24/28 (last observation 24/28 (observed cases)
carried forward) in the pooled population of patients with
moderate to severe AD. P-values from ANCOVA a) BADL scores

Figure 2
Mean change from baseline in ADL14 BADL and IADL score
b) IADL scores

at Week 24/28 (observed cases) in the pooled population of


patients with moderate to severe AD. P-values from ANCOVA

An assessment of clinical effectiveness showed that


combination therapy with memantine plus cholinesterase
inhibitors slowed both cognitive and functional decline
Although the patients recruited in the clinical trials analysed (measured using the Weintraub ADL Scale) compared with
in this study were not selected for having functional cholinesterase inhibitor monotherapy or no treatment (25). In
impairments, mean ADL14 total scores in both memantine and the current analysis, differences in functional decline between
placebo treatment groups at baseline were indicative of patients memantine-treated and placebo-treated patients increased over
with impaired functional ability. At study end, mean total score time, and Atri et al showed similar increases with time that
significantly favoured memantine and a single-item analysis were sustained for at least four years (predicted frequency of
revealed the significant benefits of memantine, compared to dependence rising from 37% to 74% in untreated patients and
placebo, in improving patients’ abilities to perform basic daily from 32% to 57% in patients treated with memantine plus
activities, such as toileting and grooming, which are often cholinesterase inhibitors) (25).
impaired in the moderate to severe stages of the disease (2). AD is a complex disease, with a progressive course that
Analyses according to OC and LOCF approaches did not manifests as disturbances across functional, behavioural and
markedly influence the observed results in this analysis, cognitive domains (1). As the only approved treatment for AD
indicating the consistency of the functional benefits shown with that targets the glutamatergic system, memantine has been
memantine. Furthermore, significant benefits of memantine shown to confer statistically significant advantages compared
therapy were seen as early as week 12 (according to the ADL14 with placebo on cognitive, behavioural, and global endpoints
total scores and IADL scores). throughout the course of AD (10-12, 26). In addition, the current
The six trials included in this analysis all recruited analysis provides evidence of benefits for memantine on
outpatients, but the findings were also congruent with results functional outcomes based on six 6-month studies in moderate
from the subset analysis of nursing home patients with AD in to severe AD. Furthermore, in efficacy and safety studies,
the 9M-BEST study (13). memantine treatment was consistently well tolerated (17-22).

773
The Journal of Nutrition, Health & A ging©
V olume 14, Number 9, 2010

MEMA NTINE BENEFITS FUNCTIONA L A BILITIES IN MODERA TE TO SEV ERE A LZHEIMER’S DISEA SE

Indeed, the overall incidence of treatment-related adverse events 5. Yaffe K, Fox P, Newcomer R, Sands L, Lindquist K, Dane K, et al. Patient and

with memantine did not differ from placebo, in a published


caregiver characteristics and nursing home placement in patients with dementia.
JAMA. 2002;287:2090-7.
pooled analysis of six randomized trials of memantine 20 6. Georges J, Jansen S, Jackson J, Meyrieux A, Sadowska A, Selmes M. Alzheimer's
mg/day in patients with AD (11). disease in real life-the dementia carer's survey. Int J Geriatr Psychiatry. 2008;23:546-
51.
It is well established that caregiver burden increases, as does 7. Danysz W, Parsons CG. The NMDA receptor antagonist memantine as a
the risk of institutionalisation, in the more advanced stages of symptomatological and neuroprotective treatment for Alzheimer's disease: preclinical

AD (4, 5) – and functional decline plays a major part in the


evidence. Int J Geriatr Psychiatry. 2003;18,(Suppl 1):S23-32.
8. Lipton SA. Paradigm shift in NMDA receptor antagonist drug development:
burden and risk. The results from this post hoc analysis molecular mechanism of uncompetitive inhibition by memantine in the treatment of

demonstrate that memantine supports patient independence by


Alzheimer's disease and other neurologic disorders. J Alzheimers Dis.
2004;6(Suppl):S61-74.
reducing the decline in basic and instrumental skills and their 9. Farlow MR, Graham SM, Alva G. Memantine for the treatment of Alzheimer's
ability to perform everyday tasks. This could potentially ease disease: tolerability and safety data from clinical trials. Drug Saf. 2008;31:577-85.
10. Gauthier S, Loft H, Cummings J. Improvement in behavioural symptoms in patients
the burden of caregiving and improve patient and carer quality with moderate to severe Alzheimer's disease by memantine: a pooled data analysis.
of life, and one study has shown that patients treated with Int J Geriatr Psychiatry. 2008;23:537-45.

memantine required 52 hours less caregiver time per month


11. Winblad B, Jones RW, Wirth Y, Stoffler A, Mobius HJ. Memantine in moderate to
severe Alzheimer's disease: a meta-analysis of randomised clinical trials. Dement
(1.5 h/day), than those receiving placebo (27). In addition, Geriatr Cogn Disord. 2007;24:20-7.

patients with AD receiving memantine were three times more


12. Mecocci P, Bladstrom A, Stender K. Effects of memantine on cognition in patients
with moderate to severe Alzheimer's disease: post-hoc analyses of ADAS-cog and
likely to remain autonomous after 28 weeks of daily treatment SIB total and single-item scores from six randomized, double-blind, placebo-
than patients receiving placebo (28), and recently it was shown controlled studies. Int J Geriatr Psychiatry. 2009;24:532-8.
13. Winblad B, Poritis N. Memantine in severe dementia: results of the 9M-Best Study
that nursing home admission for patients with AD was (Benefit and efficacy in severely demented patients during treatment with
significantly delayed during memantine treatment, in memantine). Int J Geriatr Psychiatry. 1999;14:135-46.

combination with acetylcholinesterase inhibitors, compared


14. Black SE, Doody R, Li H, McRae T, Jambor KM, Xu Y, et al. Donepezil preserves
cognition and global function in patients with severe Alzheimer disease. Neurology.
with patients receiving acetylcholinesterase inhibitors alone or 2007;69:459-69.

receiving no cognitive enhancers (29).


15. Homma A, Imai Y, Tago H, Asada T, Shigeta M, Iwamoto T, et al. Long-term safety
and efficacy of donepezil in patients with severe Alzheimer's disease: results from a
The investigation of single-items may lead to the 52-week, open-label, multicenter, extension study in Japan. Dement Geriatr Cogn
identification of specific areas of treatment effects; however, it Disord. 2009;27:232-9.
16. Winblad B, Kilander L, Eriksson S, Minthon L, Båtsman L, Wetterholm A-L, et al.
can also increase the risk of identifying random performance Donepezil in patients with severe Alzheimer’s disease: double-blind, parallel-group,
fluctuations. The results of the single-item/sub-scale analyses placebo-controlled study. Lancet, 2006:367:1057–65

identified specific items for which memantine demonstrated a


17. Bakchine S, Loft H. Memantine treatment in patients with mild to moderate
Alzheimer's disease: results of a randomised, double-blind, placebo-controlled 6-
significant beneficial effect, and these results were consistent at month study. J Alzheimers Dis. 2007;11:471-9.

the analysed time-points. The consistent results indicate that the


18. Peskind ER, Potkin SG, Pomara N, Ott BR, Graham SM, Olin JT, et al. Memantine
treatment in mild to moderate Alzheimer disease: a 24-week randomized, controlled
single-item/sub-scale scores are not random fluctuation but trial. Am J Geriatr Psychiatry. 2006;14:704-15.
significant effects of memantine on specific sub-domains. 19. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. Memantine in
moderate-to-severe Alzheimer's disease. N Engl J Med. 2003;348:1333-41.
There are, however, limitations to this study as it is a post-hoc 20. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I.
study with no correction for multiple testing. Memantine treatment in patients with moderate to severe Alzheimer disease already

These results suggest that memantine treatment may lead to


receiving donepezil: a randomized controlled trial. JAMA. 2004;291:317-24.
21. van Dyck CH, Tariot PN, Meyers B, Malca Resnick E. A 24-week randomized,
a more interactive and dignified life for patients with moderate- controlled trial of memantine in patients with moderate-to-severe Alzheimer disease.

to-severe AD, than placebo. The functional improvements


Alzheimer Dis Assoc Disord. 2007;21:136-43.
22. Porsteinsson AP, Grossberg GT, Mintzer J, Olin JT. Memantine treatment in patients
observed in this analysis add to the growing evidence with mild to moderate Alzheimer's disease already receiving a cholinesterase
supporting the clinically meaningful benefits of memantine in inhibitor: a randomized, double-blind, placebo-controlled trial. Curr Alzheimer Res.
2008;5:83-9.
patients with moderate-to-severe AD. 23. Criteria for the clinical diagnosis of Alzheimer's disease. Excerpts from the
NINCDS-ADRDA Work Group report. J Am Geriatr Soc. 1985;33:2-3.
A cknowledgements: The authors would like to thank all patients and caregivers 24. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical
participating in the studies, and thank all investigators for their contributions to the studies. diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under
H. Lundbeck A/S, Merz Pharmaceuticals GmbH and Forest Research Institute Inc. funded the auspices of Department of Health and Human Services Task Force on
the studies used for this analysis. H. Lundbeck A/S conducted the pooled analysis and Alzheimer's Disease. Neurology. 1984;34:939-44.
editorial assistance for this manuscript was provided by ESP Ltd (Sandhurst, UK) with 25. Atri A, Shaughnessy LW, Locascio JJ, Growdon JH. Long-term course and
funding from H. Lundbeck A/S. effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc

References
Disord. 2008;22:209-21.
26. Robinson DM, Keating GM. Memantine: a review of its use in Alzheimer's disease.
Drugs. 2006;66:1515-34.
1. Cummings JL. Alzheimer's disease. N Engl J Med. 2004;351:56-67. 27. Wimo A, Winblad B, Stoffler A, Wirth Y, Mobius HJ. Resource utilisation and cost
2. Galasko D, Schmitt F, Thomas R, Jin S, Bennett D. Detailed assessment of activities analysis of memantine in patients with moderate to severe Alzheimer's disease.
of daily living in moderate to severe Alzheimer's disease. J Int Neuropsychol Soc. Pharmacoeconomics. 2003;21:327-40.
2005;11:446-53. 28. Rive B, Vercelletto M, Damier FD, Cochran J, Francois C. Memantine enhances
3. Lobo A, Launer LJ, Fratiglioni L, Andersen K, Di Carlo A, Breteler MM, et al. autonomy in moderate to severe Alzheimer's disease. Int J Geriatr Psychiatry.
Prevalence of dementia and major subtypes in Europe: A collaborative study of 2004;19:458-64.
population-based cohorts. Neurologic Diseases in the Elderly Research Group. 29. Lopez OL, Becker JT, Wahed AS, Saxton J, Sweet RA, Wolk DA, et al. Long-term
Neurology. 2000;54(Suppl 5):S4-9. effects of the concomitant use of memantine with cholinesterase inhibition in
4. Smith GE, O'Brien PC, Ivnik RJ, Kokmen E, Tangalos EG. Prospective analysis of Alzheimer disease. J Neurol Neurosurg Psychiatry. 2009;80:600-7.
risk factors for nursing home placement of dementia patients. Neurology.
2001;57:1467-73.

774

You might also like