Sie sind auf Seite 1von 8

CLINICAL FOCUS

Primary Psychiatry. 2006;13(10):48-55

Glutamate in the Neurobiology and


Treatment of Dementias
Kelly M. Cosman, BS, and Anton P. Porsteinsson, MD

ABSTRACT
FOCUS POINTS
How might the neurotransmitter glutamate be involved in the
• Glutamate is one of the chief neurotransmitters in the brain
pathology of dementia? Glutamate is an essential component in and is necessary for the formation and storage of memories.
physiologic processes, playing a critical role in cognition and the • The role of glutamate in the pathogenesis of dementia and
the use of glutamate antagonists in dementia treatment
formation of memories. Under pathologic conditions, however, have been increasingly prevalent areas of research.
glutamate has been associated with excitotoxicity (neuronal dam- • In pathologic conditions, excessive stimulation by gluta-
mate is associated with excitotoxicity (neuronal injury)
age) and apoptosis (cell death). This excitotoxic effect may be an
and apoptosis (cell death), which may play a role in the
underlying culprit in the pathology of dementia. Numerous research pathogenesis of dementia.
studies have been conducted to evaluate the drug memantine, an • Antagonism of glutamate seems to preserve cognitive func-
tion and global measures in varying degrees of severity of
N-methyl-D-aspartate-receptor antagonist of glutamate, and its Alzheimer’s disease and mild-to-moderate vascular dementia.
effects on patients with dementia. Results have lent support for • The N-methyl-D-aspartate receptor site has been identi-
the use of memantine in the treatment of dementias caused by fied as an ideal target for glutamate antagonism.
• The glutamate antagonist memantine has been shown to be
Alzheimer’s disease and vascular insult. both efficacious and safe in numerous clinical trials.

INTRODUCTION Alzheimer’s disease is by far the best known and most


Dementia is a devastating disorder that possesses the prevalent variant of neurodegenerative dementia, accounting
potential to deprive a person of his or her identity, caus- for approximately 70% of cases.7 It is estimated that approxi-
ing dramatic changes in personality, memory, and cognitive mately 4.5 million United States residents are afflicted with
abilities.1 The prevalence of dementia in people ≥65 years of Alzheimer’s disease.8 The precise etiology of Alzheimer’s disease
age in North America has been estimated to be between 6% is not completely understood. Studies indicate that it is a com-
and 10%, which is approximately double when milder cases plex, heterogenous disorder involving numerous pathophysi-
are included.2 Numerous studies demonstrate that both the ologic processes. A cascade of events typically produces the
prevalence and incidence of dementia increases dramatically classic neuropathologic features of neuritic plaques, neurofi-
with age,2-5 specifically in people >85 years of age.6 brillary tangles, and synaptic degeneration; however, genetic

Ms. Cosman is a research assistant in the Program in Neurobehavioral Therapeutics at the University of Rochester School of Medicine and Dentistry in Rochester, New York. Dr. Porsteinsson is director of
the Program in Neurobehavioral Therapeutics and associate professor in the Department of Psychiatry at the University of Rochester School of Medicine and Dentistry.
Disclosure: Ms. Cosman reports no affiliation with or financial interest in any organization that may pose a conflict of interest. Dr. Porsteinsson is a consultant to Abbott, AstraZeneca, Bristol-Myers Squibb, Eisai,
Forest, Janssen, Novartis, Organon, and Pfizer; is on the speaker’s bureaus of Abbott, AstraZeneca, Bristol-Myers Squibb, Eisai, Janssen, Novartis, Organon, and Pfizer; and receives grant support from Abbott,
AstraZeneca, Bristol-Myers Squibb, Eisai, Elan, Eli Lilly, Forest, Janssen, Merck, Mitsubishi, Myriad Neurosciences, Neurochem, Novartis, Ono Pharma, Pfizer, and sanofi-aventis.
Please direct all correspondence to: Anton P. Porsteinsson, MD, University of Rochester School of Medicine, Monroe Community Hospital, 435 East Henrietta Rd, Rochester, NY 14620; Tel: 585-760-6560; Fax: 585-760-
6572; E-mail: Anton_Porsteinsson@urmc.rochester.edu.

Primary Psychiatry © MBL Communications 48 October 2006


Glutamate in the Neurobiology and Treatment of Dementias

predisposition and epidemiologic factors contribute to the risk onic acid, and kainate receptors, each with complex substruc-
of developing Alzheimer’s disease as well as to the nature of tures. The receptors and respective substructures provide a
its progression.7,9,10 Cholinesterase inhibitors (ChEIs) are the variety of potential sources from which to modify glutamater-
only medications currently Food and Drug Administration gic neurotransmission.12,18,19
approved in the US for the treatment of mild-to-moderate Modulation of glutamate activity at the NMDA recep-
Alzheimer’s disease. ChEIs confer modest cognitive, global, tor site has been of particular interest, as there has been an
and functional benefits but do not appear to prevent cell increasing amount of evidence suggesting that it is a logical
death in Alzheimer’s disease and thus cannot alter the disease’s target for cognitive enhancement and possible neuropro-
progression.11 However, in addition to cholinergic pathways, tection associated with Alzheimer’s disease and vascular
mechanisms of neuronal degeneration and other neurotrans- dementia. These receptors are present at high concentra-
mitter pathways may also be involved in the pathogenesis of tions throughout the brain, especially in the neocortex and
Alzheimer’s disease, including the glutamatergic system.12 hippocampus. Normal activation of the NMDA receptor is
Although not as widely known in society as Alzheimer’s a required step in memory encoding and storage, a process
disease, vascular dementia is another leading form of demen- which occurs via a mechanism of synaptic plasticity called
tia, accounting for approximately 10% to 20% of dementia long-term potentiation (LTP).19 However, under pathologic
cases.13 The etiology of dementia related to vascular insult is conditions the receptor may become excessively stimulated
diverse and cannot be depicted as one universal mechanism.14 by glutamate. In this instance, the excessive activation at
The general course of events includes the blockage of blood the receptor site leads to an overload of calcium in the cell,
vessels, resulting in damage and death of tissues in the sur- which impairs neuronal homeostasis. Continued impairment
rounding areas.15 As neurons die at the site of the blockage, gradually causes excitotoxicity (neuronal damage) and may
they release excess glutamate and overstimulate neighboring eventually lead to cellular apoptosis (programmed mecha-
neurons, potentially expanding the area of damage.16 nisms of cell death).20 Such excitotoxicity has been impli-
In addition to its physical effects, there are tremendous eco- cated in neurodegenerative disorders such as Alzheimer’s
nomic and social consequences of dementia. For example, in disease, Parkinson’s disease, and Huntington’s disease, as well
1994, the lifetime cost for care for a patient with Alzheimer’s as cerebral ischemia.12,16,18
disease averages approximately $175,000 in the US.17 As Particular to Alzheimer’s disease, the protein β-amyloid
dementia worsens, patients become increasingly impaired (Aβ), whose level is typically heightened as part of the disease
cognitively and functionally, creating high stress levels for the pathology, is thought to enhance the toxicity of glutamate.
patient and caregivers. Advances in medical treatment and tech- Several preclinical studies have shown that the presence of Aβ
nology are needed to reduce these effects. Research is ongoing peptide fragment 25–35 adversely affects calcium homeostasis
to investigate the role of glutamate in such dementias and to in the presence of glutamate, consequently exacerbating the
examine the therapeutic use of glutamate antagonists. neurotoxic effects of glutamate.21,22 Exposure to glutamate
alone was found to significantly reduce the survival rate of
both mature cerebellar granule cells and human cortical neu-
EXCITOTOXICITY IN DEMENTIAS rons. Furthermore, addition of Aβ25–35 significantly exac-
Glutamate is the principal excitatory neurotransmitter in erbated the neurotoxicity of glutamate; however, this effect
the brain. It plays a central role in synaptic transmission in was not demonstrated when the Aβ peptide was a scrambled
corticocortical association fibers and the majority of hippo- sequence of amino acids from the Aβ25–35 peptide.21,22
campal pathways. Glutamate stimulates a number of post- In addition to the effects that Aβ is thought to have on gluta-
synaptic membrane receptors, of which there are two main mate toxicity, glutamate and excessive activation of the NMDA
classes. The metabotropic class of receptors mediates slower receptor are believed to enhance the production of pathologic
and longer-lasting cellular effects, using G-protein second forms of Aβ and another Alzheimer’s disease-related protein, tau.
messenger systems. Conversely, ionotropic receptors control Excessive stimulation by glutamate reduces the energy levels of
rapid synaptic activity via the regulation of the flow of cations affected neurons, causing neurotoxicity and cell death. Cellular
(serum sodium [Na+], potassium [K+], and serum calcium energy reduction has been associated with the elevation of the
[Ca++]) into the cell through ligand-gated ion channels. There β-site amyloid precursor protein-cleaving enzyme, β-secretase,
are three types of ionotropic receptors: N-methyl-D-aspartate which is essential for the rate-limiting step in the formation
(NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazole propi- of Aβ.23 Thus, reduction of energy levels caused by glutamate

Primary Psychiatry © MBL Communications 49 October 2006


K.M. Cosman, A.P. Porsteinsson

neurotoxicity may indirectly increase the production of Aβ. MEMANTINE


Studies have also shown that glutamate, as a neurotransmitter,
strengthens tau immunoreactivity, indicating an increase in the
production of phosphorylated tau.24 Additionally, both acute Mode of Action
and chronic toxic activation by NMDA increases tau immuno- Memantine (1–amino-3, 5-dimethyladamantane hydrochlo-
reactivity.25 Hence, it seems that a vicious cycle emerges, where ride) is a moderate-affinity, non-competitive, NMDA receptor
each pathologic condition tends to exacerbate the other.12,18,26 antagonist that exerts voltage-dependent effects with rapid
In the case of vascular dementia, hypoxic-ischaemia has blocking/unblocking kinetics. Memantine, acting at the NMDA
been associated with increasing extracellular levels of gluta- receptor site, allows normal glutamatergic neurotransmission
mate. The lack of oxygen and glucose resulting from isch- under physiologic circumstances while inhibiting excitotoxicity
aemia results in depleted cellular energy levels, which can under conditions of chronic glutamatergic stimulation.28-30
activate glutamatergic mechanisms.26 Excessive activation Memantine occupies the same NMDA receptor channel
may lead to overstimulation of the glutamate receptor, pro- as Mg2+ but is less voltage dependent and does not leave the
moting excitotoxicity and apotosis. channel so easily with minor depolarization, thus prevent-
The glutamatergic system has been identified as a logical ing the sustained influx of Ca2+ during pathologic release of
target for both cognitive enhancement and interruption of the glutamate. During physiologic glutamate release, memantine
pathophysiologic conditions associated with Alzheimer’s disease leaves the channel and allows normal influx of Ca2+. A signal
and vascular dementia.27,28 Stimulation of NMDA receptors is produced which can be recognized and processed due to
by glutamate may improve memory and cognition; however, reduced intraneuronal noise.28-30
excessive stimulation is associated with neuronal injury and Memantine reduces acute excitotoxic damage in vitro and in
toxicity. In normal circumstances, magnesium (Mg)2+ occupies vivo following administration of glutamate agonists. In models
the NMDA receptor channel and blocks Ca2+ entry into the of global and focal ischemia, it confers protection against Aβ-
neuron. During physiologic learning and memory processes, induced neurotoxicity and improved cognitive performance in
high concentrations of synaptic glutamate are transiently rats.28-30 This particular mechanism yields support for the use
released; due to its strong voltage dependency, Mg2+ leaves the of memantine in the treatment of vascular dementia.
NMDA receptor and allows normal Ca2+ influx. However, In addition to its action at the NMDA receptor cite,
under pathologic conditions such as with Alzheimer’s disease memantine also blocks serotonin (5-HT)3 receptors at physi-
and other neurologic diseases, neurons and glia cells continue ologic concentrations. Antagonism at 5-HT3 has been pos-
a sustained release of glutamate, prolonging the depolariza- tulated to facilitate LTP, have antipsychotic and antinausea
tion that displaces Mg2+ from the NMDA receptor channel. effects, and decrease gastric hypermotility.29,30
Consequently, continuous influx of Ca2+ is allowed into the
neuron, potentially elevating the intraneuronal Ca2+ pool to a Pharmacokinetics
level that will prevent the transmission of a physiologic signal. Following oral administration, memantine is completely
Neuronal degeneration may also occur if the Ca2+ concentra- absorbed with a Tmax of 4–6 hours and an oral bioavailability
tion becomes too high or is elevated for too long a duration. of 100%. Food does not affect its bioavailability. Memantine
Preclinical data support the NMDA receptor as a prom- shows linear pharmacokinetics over the therapeutic dose range.
ising target for neuroprotective agents as the high Ca2+ It is extensively distributed in tissue and readily crosses the
permeability of this receptor likely underlies its neurotoxic blood-brain barrier. Memantine is approximately 45% protein
potential. It is possible to counteract glutamate excitotoxicity bound. The terminal half-life of memantine is 60–80 hours. It
by blocking the NMDA receptor with a compound that has a undergoes little metabolism and is excreted largely unchanged
higher affinity to the receptor than Mg2+. However, if NMDA in the urine. There are no therapeutically active metabolites.
receptor activity is completely blocked, normal physiologic Memantine clearance is reduced with increasing degrees of
activity of the NMDA receptor (such as learning activity renal impairment. The cytochrome P450 system is minimally
and LTP) is blocked along with the pathologic activity. High involved in the metabolism of memantine. These data indicate
affinity compounds such as MK801 or phencyclidine impair that no pharmacokinetic interactions with drugs metabolized
learning in animal models and can produce psychotomimetic by these enzymes are to be expected. Medications or conditions
effects. Approaches with moderate-affinity NMDA receptor that raise urine pH may decrease the urinary elimination of
antagonists have been more fruitful.12,18,26,27 memantine, resulting in increased plasma levels.31

Primary Psychiatry © MBL Communications 50 October 2006


Glutamate in the Neurobiology and Treatment of Dementias

Data from preclinical trials, pharmacokinetic studies, pivot- Assessment Scale–Cognitive Subscale and the Clinician
al trials, and postmarketing studies suggest that the combined Interview Based Impression of Change-Plus Caregiver Input
use of memantine and a ChEI is safe and well tolerated.32-34 at certain data points.40 The second unpublished trial evalu-
The following sections provide a review of results from several ated memantine’s use in combination with a ChEI in the
multicenter, randomized, double-blind, placebo-controlled, treatment of mild-to-moderate Alzheimer’s disease. Neither
parallel-arm studies which evaluated the use of memantine in measure used for the efficacy analysis elicited statistically
the treatment of dementias. significant differences using the initially planned analyses.
However, both measures displayed numerical advantages
in favor of the use of memantine. Furthermore, additional
MEMANTINE IN ADVANCED analyses revealed statistically significant findings in support of
ALZHEIMER’S DISEASE memantine when differences due to ChEI type and duration
of treatment were considered (A.P. Porsteinsson, unpublished
Studies of memantine in the treatment of moderate-to-severe data, July 2006). A meta-analysis that included all of these tri-
Alzheimer’s disease have provided the strongest results favoring als shows a statistically significant but very small advantage to
its use (Table 1).35-38 One study that evaluated memantine mono- memantine over placebo on cognition (intent-to-treat analy-
therapy for the treatment of moderate-to-severe Alzheimer’s sis), which was barely detectable clinically, but no effect on
disease found statistically significant differences in measures of behavior, activities of daily living, or observed cases analysis of
cognition, daily functioning, and global assessment, with the cognition. Overall, the results that were reported from all of
largest effect seen in the measure of cognition.36 The double- these studies found memantine treatment to be safe and well
blind period of this study as well as an open-label extension study tolerated in the treatment of early Alzheimer’s disease (A.P.
with the same group of participants showed that memantine Porsteinsson, unpublished data, July 2006).39,40
was both safe and effective in this group over a 1-year period.
Another study was conducted to assess the efficacy and safety of
memantine when used in combination with a ChEI for the treat- MEMANTINE IN MIXED AND
ment of moderate-to-severe Alzheimer’s disease. Analyses from
this study provided strong and statistically significant results
VASCULAR DEMENTIAS
favoring dual therapy with memantine and ChEI. Furthermore, Several studies have also considered the use of memantine in
treatment with dual therapy resulted in improved cognitive the treatment of vascular dementia (Table 3).41-44 One study that
performance relative to baseline, whereas treatment with ChEI included participants with vascular dementia (51%) as well as
alone was associated with continued cognitive decline. This moderate-to-severe Alzheimer’s disease (49%) found statistically
study also supported the safety of memantine therapy in patients significant differences between treatment groups in both a mea-
with advanced Alzheimer’s disease.38 sure of global assessment and a measure of functional and behav-
ioral disturbances; however, the effect of the second measure was
relatively small.42 Mean scores were not provided for the results
MEMANTINE IN EARLY ALZHEIMER’S from the global assessment scale; thus, effect sizes could not be
calculated. However, the efficacy results were statistically superior
DISEASE for memantine over placebo. Two other studies were conducted
Studies of the use of memantine in the treatment of to evaluate the use of memantine exclusively in vascular demen-
mild-to-moderate Alzheimer’s disease also show support for tia.43,44 Both studies used measures of global assessment and
memantine, although results were not as strong as those seen cognition as their primary efficacy outcomes. The measures of
in studies of moderate-to-severe Alzheimer’s disease (Table global assessment did not show statistically significant differences
2; A.P. Porsteinsson, unpublished data, July 2006).39,40 One between treatment groups. However, the measures of cognition
study of memantine monotherapy demonstrated significant did provide significant differences with small-to-moderate effect
differences in cognitive and global measures, but not in sizes. It was noted that the normal decline in function that is
measures of daily function.39 Two unpublished trials did not associated with vascular dementia tends to be slower than that
find benefit of memantine over placebo at endpoint (A.P. seen in Alzheimer’s disease, which may make it more difficult to
Porsteinsson, unpublished data, July 2006).40 During the show a strong treatment effect with this study group. The results
course of the first study,40 the placebo group did not decline as from all three studies demonstrated that memantine use was safe
anticipated and instead improved on the Alzheimer’s Disease and well tolerated in the treatment groups.42-44

Primary Psychiatry © MBL Communications 51 October 2006


K.M. Cosman, A.P. Porsteinsson

TABLE 1
MEMANTINE IN ADVANCED ALZHEIMER’S DISEASE36-38
Target
Inclusion Concomitant Study Memantine Efficacy Outcomes: Conclusions/
Study Name Criteria Medications Duration Dose Measure (Effect Size)* Safety Outcomes Comments
MRZ960536 Diagnosis of Anticonvulsants, 28 weeks 10 mg BID Primary: No clinically impor- Memantine was shown
probable AD antiparkinso- tant differences in to be safe, well toler-
CIBIC-plus (0.27 LOCF/0.27
nians, hypnot- AEs between the ated, and efficacious
OC)
ics, anxiolytics, treatment groups in the treatment of
MMSE 3–15 neuroleptics, or ADCS-ADLsev (0.32 moderate-to-severe
GDS stage 5 cholinomimetic LOCF/0.53 OC) AD
or 6 agents, and any
FAST ≥stage investigational
medications were Secondary:
6a
excluded SIB (0.49 LOCF/0.48 OC)
MMSE (0.26 LOCF/0.11 OC)
FAST (0.31 LOCF/0.31 OC)
GDS (0.21 LOCF/0.21 OC)
NPI (0.21 LOCF/0.18 OC)
MRZ9605 Completion Same as above 24 weeks 10 mg BID The measures used in No clinically impor- Memantine was shown
open-label of MRZ9605 MRZ9605 were continued for tant differences in to be well tolerated
extension37 placebo-con- the open-label extension AEs between those over a period of 52
trolled study who switched to and weeks
those who remained
After switching to meman- on memantine
tine, patients from the
placebo group experienced
significant benefits in all
measures when their per-
formance was compared to
their rate of decline in the
double-blind phase (P<.05)
MEM-MD- Diagnosis of Concomitant 24 weeks 10 mg BID Primary: Incidence of treat- Treatment with
0238 probable AD medications were ment-emergent AEs, memantine in addition
SIB (5.01 LOCF/4.74 OC)
permitted if dos- EKG abnormalities, to donepezil resulted
ing was stable ADCS-ADL19 (2.78 and potentially in improvements in
MMSE 5–14 LOCF/3.03 OC) clinically significant measures of cogni-
laboratory findings tion, function, clinical
Ongoing treat- or vital sign mea- global status, and
ment with Secondary: surements was simi- behavior relative to
donepezil for 6 CIBIC-plus (3.36 LOCF/3.11 lar between groups baseline
months prior to OC)
enrollment (last 3
months at stable NPI (3.87 LOCF/3.33 OC) Early discontinua- Memantine therapy
dose of 5–10 mg) BGP (4.01 LOCF/4.18 OC) tion was less in the was shown to be
was required memantine group safe when combined
(7%) than in the with donepezil for the
placebo group (12%) treatment of moder-
ate-to-severe AD

*Effect size=(meanmem–meanplac)/(SDpooled), where SDpooled=√[(nmem–1)(sdmem)2+(nplac–1)(sdplac)2)/(nmem+nplac)] and ≥.2=small effect, ≥.5=medium effect, ≥.8=large effect.
AD=Alzheimer’s disease; MMSE=Mini-Mental State Examination; GDS=Global Deterioration Scale; FAST=Functional Assessment Staging instrument; CIBIC-plus=Clinician’s Interview-
Based Impression of Change Plus Caregiver Input; LOCF=last observation carried forward; OC=observed cases; ADCS-ADLsev=Alzheimer’s Disease Cooperative Study Activities of Daily
Living Inventory modified for more severe dementia; SIB=Severe Impairment Battery; NPI=Neuropsychiatric Inventory; AEs=adverse events; ADCS-ADL19=19-item Alzheimer’s Disease
Cooperative Study Activities of Daily Living Inventory; BGP=Behavioral Rating Scale for Geriatric Patients.
Cosman KM, Porsteinsson AP. Primary Psychiatry. Vol 13, No 10. 2006.

Primary Psychiatry © MBL Communications 52 October 2006


Glutamate in the Neurobiology and Treatment of Dementias

TABLE 2
MEMANTINE IN EARLY ALZHEIMER’S DISEASE39,40*
Target
Inclusion Concomitant Study Memantine Efficacy Outcomes: Conclusions/
Study Name Criteria Medications Duration Dose Measure (Effect Size)* Safety Outcomes Comments
MEM-MD- Diagnosis of Treatment with 24 weeks 10 mg BID Primary: The only significantly This study sup-
1039 probable AD a ChEI within 30 different AE was ports the safety and
ADAS-cog (0.15 LOCF/0.13
days of screening somnolence (7% efficacy, with small
OC)
and during the memantine vs. 1% effects on global mea-
MMSE 10–22 study period was CIBIC-plus (0.32 LOCF/0.02 placebo, P=.002) sures and behavior,
MADRS <22 not permitted OC) of memantine in the
treatment of mild-to-
No clinically mean- moderate AD
Any previous Secondary: ingful differences
treatment with NPI (0.21 LOCF/0.13 OC) between groups’
memantine was changes in lab test
ADCS-ADL23 (0.01 LOCF/0.0
not permitted values, vitals signs,
OC)
or EKG results
Study Diagnosis of Treatment with 24 weeks 10 mg BID Primary: Safety analysis It was noted that
9967940 probable AD a ChEI within 30 results have not during the course of
CIBIC-plus (0.04 LOCF)
days of screening been reported the study, the placebo
and during the ADAS-cog (0.09 LOCF) group did not decline
MMSE 11–23 study period was as anticipated, and
not permitted Discontinuation due instead improved on
Secondary: to AEs was higher the ADAS-cog and
ADCS-ADL (0.07 LOCF) in the memantine CGIC-plus at certain
group (9%) than the data points
NPI (0.19 LOCF)
placebo group (4%)
Extension of study is
currently ongoing to
evaluate memantine
use over 2.5–3-year
period
MEM-MD-12* Diagnosis of Ongoing treat- 24 weeks 20 mg/day Primary: No statistically Additional analyses
probable AD ment with ChEI given once important differ- revealed statistically
ADAS-cog (0.11 LOCF)
for at least 6 at night ences in AEs and no significant findings in
months prior to CIBIC-plus (0.04 LOCF/0.04 clinically significant support of memantine
MMSE 10–22 enrollment (last OC) differences between when considering dif-
MADRS <22 3 months at the groups’ labora- ferences due to ChEI
stable dose) was tory findings, vital type and duration
Secondary:
required sign measurement, of use
ADCS-ADL23, NPI, MMSE, and and EKG results
RUD: no statistically sig-
nificant differences between
groups
* A.P. Porsteinsson, unpublished data, July 2006.
AD=Alzheimer’s disease; MMSE=Mini-Mental State Examination; MADRS=Montgomery-Asberg Depression Rating Scale; ChEI=cholinesterase inhibitor; ADAS-cog=Alzheimer’s
Disease Assessment Scale-Cognitive Subscale; LOCF=last observation carried forward; OC=observed cases; CIBIC-plus=Clinician’s Interview-Based Impression of Change Plus
Caregiver Input; NPI=Neuropsychiatric Inventory; ADCS-ADL23=23-item Alzheimer’s Disease Cooperative Study Activities of Daily Living Inventory; RUD=Resource Utilization in
Dementia scale.
Cosman KM, Porsteinsson AP. Primary Psychiatry. Vol 13, No 10. 2006.

Primary Psychiatry © MBL Communications 53 October 2006


K.M. Cosman, A.P. Porsteinsson

TABLE 3
MEMANTINE IN MIXED AND VASCULAR DEMENTIA41-44
Target
Inclusion Concomitant Study Memantine Efficacy Outcomes: Safety Conclusions/
Study Name Criteria Medications Duration Dose Measure (effect size)* Outcomes Comments
M-Best DSM-III-R MAOIs, neuro- 12 weeks 10 mg/day Primary: No significant No valid measure of
Study41,42 diagnosis of leptics, tricyclic differences cognitive function was
CGI-C: mean scores were not reported,
dementia antidepres- in safety used for this study
however stratified Wilcoxon test showed
sants, hypnot- measures
significant treatment differences favor-
ics, nootropics, were observed
ing memantine at week 12 (memantine
GDS stages or cerebral between treat- Memantine was found
response=73%, placebo response=45%,
5–7 circulation ment groups to be safe and effica-
P<.001)
stimulating cious on global mea-
MMSE <10
agents were sures in the treatment
not permitted of severe AD and VaD
BGP care dependence sub-score (0.17 ITT)
MMM 30043 Symptomatic Anticonvulsant, 28 weeks 20 mg/day Primary: Frequency Memantine was safe
mild-moder- anti-Parkinson, of AEs was and well tolerated in
CIBIC-plus (0.20 OC)
ate VaD centrally acting similar the treatment of mild-
antihyperten- ADAS-cog (0.48 OC) between treat- to-moderate VaD
sive, hypnotic, ment groups
MMSE 12–20 anxiolytic, (memantine
antipsychotic, Secondary: 76%, placebo Small effects were
and cogni- MMSE (0.33 OC) 74%) seen in most mea-
No diagnosis tion-enhancing sures, with the great-
of AD agents were CGI-C physician (0.24 OC) est effect occurring
not permitted CGI-C caregiver (0.23 OC) No clinically with measures of
significant cognitive function
NOSGER total score (0.04 OC) effects of
GBS total score (0.24 OC) memantine
were found in It was noted that
biochemical natural decline is
markers, vital slower in VaD than in
sign measure- AD, which may reduce
ments, or EKG the emergence of a
results treatment effect

MMM 50044 Diagnosis of Investigational 28 weeks 10 mg/day Primary: Emergence of Memantine was found
probable VaD drugs, psy- AEs was simi- to be well tolerated
ADAS-cog (0.24 LOCF)
chotropic lar between in the treatment of
medications, CGI-C: results were not statistically sig- groups (77% mildto-moderate VaD
HIS ≥4 drugs with psy- nificant and means were not reported memantine,
MMSE 10–22 chiatric side- 75% placebo)
effects and oral A small treatment
anticoagulants Secondary: effect was found only
were not per- NOSGER (0.10 TPP) with the ADAS-cog
mitted
GBS total score (0.06 TPP)
MMSE: minimal changes from baseline,
and no statistically significant differ-
ences between groups at endpoint
DSM-III-R=Diagnostic and Statistical Manual of Mental Disorders, Third Edition-Revised; GDS=Global Deterioration Scale; MMSE=Mini-Mental State Examination; MAOI=monoamine
oxidase inhibitor; CGI-C=Clinical Global Impression of Change; BGP=Behavioral Rating Scale for Geriatric Patients; ITT=intent-to-treat analysis; AD=Alzheimer’s disease;
VaD=vascular dementia; CIBIC-plus=Clinician’s Interview-Based Impression of Change Plus Caregiver Input; OC=observed cases; ADAS-cog=Alzheimer’s Disease Assessment
Scale-Cognitive Subscale; NOSGER=Nurse’s Observational Scale for Geriatric Patients; GBS=Gottsfries-Brane-Steen scale; AE=adverse event; EKG=electrocardiogram;
HIS=Hachinski ischaemic score; LOCF=last observation carried forward; TPP=treated-per-patient.
Cosman KM, Porsteinsson AP. Primary Psychiatry. Vol 13, No 10. 2006.

Primary Psychiatry © MBL Communications 54 October 2006


Glutamate in the Neurobiology and Treatment of Dementias

CONCLUSION 11. Schneider LS, Tariot PN. Cognitive enhancers and treatments for Alzheimer’s disease. In: Tasman A, Kay
J, Lieberman JA, eds. Psychiatry. 2nd ed. London, UK: John Wiley and Sons; 2003:2096-2108.
12. Procter A. Abnormalities in non-cholinergic neurotransmitter systems in Alzheimer’s disease. In: O’Brien
Dementia is a critical public health concern. It produces a J, Ames D, Burns A, eds. Dementia. 2nd ed. New York, NY: Oxford University Press; 2000:433-442.
13. Kase CS. Epidemiology of multi-infarct dementia. Alzheimer Dis Assoc Disord. 1991;5(2):71-76.
heavy burden on patients, caregivers, and general society with 14. Rockwood K. Vascular cognitive impairment and vascular dementia. J Neurol Sci. 2002;203-204:23-27.
its emotional and psychological effects as well as associated 15. Chapman DP, Williams SM, Strine TW, Anda RF, Moore MJ. Dementia and its implications for public
health. Prev Chronic Dis. 2006;3(2):1-13. Available at: www.cdc.gov/pcd/issues/2006/apr/05_0167.htm.
costs for health care. Available treatments are useful as dem- Accessed August 22, 2006.
onstrated by cognitive, functional, and behavioral outcome 16. Sonkusare SK, Kaul CL, Ramarao P. Dementia of Alzheimer’s disease and other neurodegenerative disor-
ders–memantine, a new hope. Pharmacol Res. 2005;51(1):1-17.
measures; however, currently available therapies provide only 17. Alzheimer’s Association. Statistics about Alzheimer’s Disease. Available at: http://www.alz.org/AboutAD/
statistics.asp. Accessed August 22, 2006.
modest improvements and cannot offer a cure for the disease.
18. Greenamyre JT, Maragos WF, Albin RL, Penney JB, Young AB. Glutamate transmission and toxicity in
In addition to enhancement of the cholinergic system, Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 1988;12(4):421-430.
19. Shimizu E, Tang YP, Rampon C, Tsien JZ. NMDA receptor-dependent synaptic reinforcement is a crucial
it is beneficial to consider alternative means for modifying process for memory consolidation. Science. 2000;290(5494):1170-1174.
processes associated with the pathology of dementia. The 20. Rothman SM, Thurston JH, Hauhart RE. Delayed neurotoxicity of excitatory amino acids in vitro.
Neuroscience. 1987;22(2):471-480.
investigation of glutamatergic pathways suggests a role in 21. Scorziello A, Meucci O, Florio T, et al. beta 25-35 alters calcium homeostasis and induces neurotoxicity in
cerebellar granule cells. J Neurochem. 1996;66(5)1995-2003.
the pathogenesis of dementia and has shown to be promis-
22. Mattson MP, Cheng B, Davis D, Bryant K, Lieberburg I, Rydel RE. beta-Amyloid peptides destabilize
ing as an additional means for treating dementia. Currently, calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity. J Neurosci.
1992;12(2):376-389.
memantine, a moderate-affinity antagonist of glutamate, has 23. Velliquette RA, O’Connor T, Vassar R. Energy inhibition elevates beta-secretase levels and activity and is
Food and Drug Administration approval for use in the treat- potentially amyloidogenic in APP transgenic mice: possible early events in Alzheimer’s disease pathogen-
esis. J Neurosci. 2005;25(47):10874-10883. Erratum in: J Neurosci. 2006;26(7):2140-2142.
ment of moderate-to-severe Alzheimer’s disease. The mecha- 24. Sindou P, Lesort M, Couratier P, Yardin C, Esclaire F, Hugon J. Glutamate increases tau phosphorylation in
primary neuronal cultures from fetal rat cerebral cortex. Brain Res. 1994;646(1):124-128.
nism of action may include enhanced neurotransmission 25. Couratier P, Lesort M, Sindou P, Esclaire F, Yardin C, Hugon J. Modifications of neuronal phosphorylated
in multiple systems in addition to possible antiexcitotoxic tau immunoreactivity induced by NMDA toxicity. Mol Chem Neuropathol. 1996;27(3):259-273.
26. Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev
effects. Memantine is used both as monotherapy and in com- Drug Discov. 2006;5(2):160-170.
bination with a ChEI. Further studies have suggested that it 27. Winblad B, Möbius HJ, Stoffler A. Glutamate receptors as a target for Alzheimer’s disease--are clinical
results supporting the hope? J Neural Transm Suppl. 2002;(62):217-225.
may have a small clinical benefit in the treatment of other 28. Parsons CG, Danysz W, Quack G. Memantine is a clinically well tolerated N-methyl-D-aspartate (NMDA)
receptor antagonist--a review of preclinical data. Neuropharmacology. 1999;38(6):735-767.
variations of dementia, such as mild-to-moderate Alzheimer’s 29. Danysz W, Parsons CG, Kornhuber J, Schmidt WJ, Quack G. Aminoadamantanes as NMDA receptor antago-
disease and vascular dementia. However, this result is not nists and antiparkinsonian agents--preclinical studies. Neurosci Biobehav Rev. 1997;21(4):455-468.
30. Danysz W, Parsons CG, Mobius HJ, Stoffler A, Quack G. Neuroprotective and symptomalogical action of
conclusive. Memantine has only a minimal risk for side memantine relevant for Alzheimer’s disease – a unified glutamatergic hypothesis on the mechanism of
action. Neurotox Res. 2000;2(2-3):85-97.
effects. These results suggest that modulation of NMDA 31. Namenda [package insert]. New York, NY: Forest Pharmaceuticals, Inc.; 2003.
receptors to reduce glutamate dysregulation and glutamate- 32. Periclou A, Ventura D, Sherman T, et al. A pharmacokinetic study of the NMDA receptor antagonist meman-
tine and donepezil in healthy young subjects [abstract]. J Am Geriatr Soc. 2003;51(suppl 4):S225.
induced excitotoxicity alleviates the symptoms of dementia. 33. Wenk GL, Quack G, Moebius HJ, Danysz W. No interaction of memantine with acetylcholinesterase inhibi-
Future investigations will be necessary to determine if the use tors approved for clinical use. Life Sci. 2000;66(12):1079-1083.
34. Hartmann S, Mobius HJ. Tolerability of memantine in combination with cholinesterase inhibitors in
of memantine is not only complimentary to the use of cholin- dementia therapy. Int Clin Psychopharmacol. 2003;18(2):81-85.
esterase inhibitors, but possibly even synergistic. PP 35. Thalheimer W, Cook S. How to calculate effect sizes from published research articles: a simplified meth-
odology. Work-Learning Research, Inc. Available at: http://work-learning.com/effect_sizes.htm. Accessed
August 22, 2006.
36. Reisberg B, Doody R, Stoffler A, et al. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med.
2003;348(14):1333-1341.
REFERENCES 37. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. A 24-week open-label extension study of
memantine in moderate to sever Alzheimer disease. Arch Neurol. 2006;63(1):49-54.
1. Glanze WD, Anderson KN, Anderson LE, eds. The Mosby Medical Encyclopedia. Rev ed. New York, NY: Plume; 1992.
38. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine treatment in patients with moderate to severe
2. Hendrie HC. Epidemiology of dementia and Alzheimer’s disease. Am J Geriatr Psychiatry. 1998;6(2 suppl 1):S3-S18. Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.
3. Bachman DL, Wolf PA, Linn R, et al. Prevalence of dementia and probable senile dementia of the Alzheimer 39. Peskind ER, Potkin SG, Pomara N, et al. Memantine treatment in mild to moderate Alzheimer disease: a
type in the Framingham study. Neurology. 1992;42(1):115-119. 24-week randomized controlled trial. Am J Geriatr Psychiatry. 2006;14(8):704-715.
4. Jorm AF, Jolley D. The incidence of dementia: a meta-analysis. Neurology. 1998;51(3):728-733. 40. McShane R, Areosa Sastre A, Minakaran N. Memantine for dementia. Cochrane Database Syst Rev.
5. Kukull WA, Higdon R, Bowen JD, et al. Dementia and Alzheimer disease incidence: a prospective cohort 2006;(2):CD003154.
study. Arch Neurol. 2002;59(11):1737-1746. 41. Diagnostic and Statistical Manual of Mental Disorders. 3rd ed rev. Washington, DC: American Psychiatric
6. Ebly EM, Parhad IM, Hogan DB, Fung TS. Prevalence and types of dementia in the very old: results from Association; 1987.
the Canadian Study of Health and Aging. Neurology. 1994;44(9):1593-1600. 42. Winblad B, Poritis N. Memantine in severe dementia: results of the 9M-BEST Study (Benefit and efficacy in
7. Cummings JL, Cole G. Alzheimer disease. JAMA. 2002;287(18):2335-2338. severely demented patients during treatment with memantine). Int J Geriatr Psychiatry. 1999;14(2):135-146.
8. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer’s disease in the US population: preva- 43. Orgogozo JM, Rigaud AS, Stoffler A, Mobius HJ, Forette F. Efficacy and safety of memantine in patients
lence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122. with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke.
9. Butler SM, Ashford JW, Snowdon DA. Age, education, and changes in the Mini-Mental State Exam scores 2002;33(7):1834-1839.
of older women: findings from the Nun Study. J Am Geriatr Soc. 1996;44(6):675-681. 44. Wilcock G, Mobius HJ, Stoffler A; MMM 500 group. A double-blind, placebo-controlled multicentre
10. Riley KP, Snowdon DA, Markesbery WR. Alzheimer’s neurofibrillatory pathology and the spectrum of cogni- study of memantine in mild to moderate vascular dementia (MMM500). Int Clin Psychopharmacol.
tive function: findings from the Nun Study. Ann Neurol. 2002;51(5):567-577. 2002:17(6):297-305.

Primary Psychiatry © MBL Communications 55 October 2006

Das könnte Ihnen auch gefallen