You are on page 1of 25

Neuropharmacology 53 (2007) 699e723

www.elsevier.com/locate/neuropharm

Review

Memantine: a NMDA receptor antagonist that improves memory by


restoration of homeostasis in the glutamatergic system - too little
activation is bad, too much is even worse
Chris G. Parsons, Albrecht Stöffler, Wojciech Danysz*
Merz Pharmaceuticals, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
Received 30 January 2007; received in revised form 19 June 2007; accepted 17 July 2007

Abstract

The neurotransmitter glutamate activates several classes of metabotropic receptor and three major types of ionotropic receptor e a-amino-3-
hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainate and N-methyl-D-aspartate (NMDA). The involvement of glutamate mediated
neurotoxicity in the pathogenesis of Alzheimer’s disease (AD) is finding increasing scientific acceptance. Central to this hypothesis is the as-
sumption that glutamate receptors, in particular of the NMDA type, are overactivated in a tonic rather than a phasic manner. Such continuous,
mild, chronic activation ultimately leads to neuronal damage/death. Additionally, impairment of synaptic plasticity (learning) may result not only
from neuronal damage per se but may also be a direct consequence of this continuous, non-contingent NMDA receptor activation. Complete
NMDA receptor blockade has also been shown to impair neuronal plasticity, thus, both hypo- and hyperactivity of the glutamatergic system
leads to dysfunction.
Memantine received marketing authorization from the EMEA (European Medicines Agency) for the treatment of moderate to severe AD in
Europe and was subsequently also approved by the FDA (Food and Drug Administration) for use in the same indication in the USA. Memantine
is a moderate affinity, uncompetitive NMDA receptor antagonist with strong voltage-dependency and fast kinetics. This review summarizes
existing hypotheses on the mechanism of action (MOA) of memantine in an attempt to understand how the accepted interaction with NMDA
receptors could allow memantine to provide both neuroprotection and reverse deficits in learning/memory by the same MOA.
Ó 2007 Elsevier Ltd. All rights reserved.

Keywords: Memantine; NMDA; Alzheimer’s disease; LTP; Learning; Memory; Symptomatic

1. Introduction namely, the glutamatergic N-methyl-D-aspartate (NMDA) re-


ceptor (Figs. 1 and 2). It is well recognised that blockade of
The ability to maintain homeostasis, which assures an opti- NMDA receptors leads to impairment of neuronal plasticity
mal physiological balance, is a basic feature of all living organ- (learning) (Collingridge and Bliss, 1995) while their overacti-
isms and is normally achieved by the presence of opposing vation leads to cell death due to calcium overload (Choi,
dynamic mechanisms, e.g. antagonistic muscles or the sym- 1992). There is however, clear evidence, that dysfunctional
pathetic and parasympathetic vegetative systems. Such an opti- overactivation of NMDA receptors may also lead to disturbances
mal balance is particularly important for very sensitive organs of plasticity, even before the development of overt excitotoxicity.
such as the central nervous system (CNS). In the present Acceptance and understanding of this principle e as indicated in
review, the authors describe one very vivid example of such the title e is crucial for understanding the cognition-improving
an obligatory balance between activation and inhibition, effects of the NMDA receptor antagonist memantine in
Alzheimer’s disease (AD).
* Corresponding author. Tel.: þ49 69 150 35 64; fax: þ49 69 596 21 50. Memantine was approved several years ago for the treat-
E-mail address: wojciech.danysz@merz.de (W. Danysz). ment of mild-severe to severe AD (2002 EU, 2003 USA)

0028-3908/$ - see front matter Ó 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuropharm.2007.07.013
700 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Channel blockers Agonists Coagonists


Glutamate Glycine
M Memantine NMDA D-serine
Na+/ Ca2+
+ Mg 2+

Antagonists
SH Modulators
D-AP5
Polyamines
5,7-DCKA
Ifenprodil Zn2+
NR2B NR1
M
Histamine

+ SH Redox
N N
Pregnenolone

NR2A NR1

NR1 NR2B
K+

Fig. 1. Schematic of the topology and pharmacological recognition sites of NMDA receptors. Two major subunit families designated NR1, NR2 as well as a minor
modulatory subunit designated NR3 have been cloned. Functional receptors in the mammalian CNS are, almost certainly, only formed by tetrameric assemblies of
two NR1 and two NR2 subunits which express the glycine and glutamate recognition sites, respectively (Hirai et al., 1996; Laube et al., 1997). NR3 (NRL or Chi-1)
is expressed predominantly in the developing CNS and does not seem to form functional homomeric glutamate-activated channels but co-expression of NR3 with
NR1 and NR2 subunits decreases response magnitude (Sucher et al., 1995; Kinsley et al., 1999). There are four known subtypes of NR2 (A, B e illustrated, C and
D) and eight splice variants of NR1. Binding of agonists such as glutamate and NMDA and co-agonists such as glycine and D-serine to both recognition sites on the
receptor subunit assembly are required for receptor activation. Competitive antagonists such as D-AP5 and glycine antagonists such as 5,7-DCKA (5,7-dichloro-
kynurenic acid) bind to recognition sites, which may be distinct to the agonist recognition sites but isosterically tightly coupled in such a way as to give a com-
petitive interaction. All NMDA receptors are permeant to Ca2þ, Naþ and Kþ. The open NMDA channel is blocked by Mg2þ and uncompetitive NMDA receptor
antagonists such as memantine and (þ)MK-801 in a voltage-dependent manner although the potency, speed and voltage-dependence of this effect depends on
antagonist affinity and subunit composition. There are similarities in the channel domain of NMDA and non-NMDA receptors and mutations at the Q/R site
responsible for determining Ca2þ permeability of AMPA/kainate receptors also influence the potency of channel blockers of the NMDA receptor at the asparagine
N-site. Polyamines such as spermine and spermidine are positive modulators binding to NR2B subunits but also block the channel at higher concentrations. Ifen-
prodil is the prototypic ‘‘selective‘‘ antagonist for NR2B containing receptors. Zn2þ is a potent, voltage-independent antagonist at NR2A containing receptors. In
addition, most NMDA receptors are influenced by Zn2þ ions in a voltage-independent manner and by redox, histamine and steroids.

and since this time is on the market in Europe, USA and many problems in clinical trials, usually with a frequency of about
countries in the world. twice that for placebo (for an overview see package inserts
Treatment with AchEIs (Acetylcholinesterase Inhibitors) of marketed AchEIs or Thompson et al. (2004); Takeda
was found to be efficacious in numerous randomized con- et al. (2006); Birks (2006)). Combination of memantine and
trolled trials (RCTs) and meta analyses but there was consen- AchEIs has been reported to produce enhanced therapeutic ef-
sus that the magnitude of effects is limited and, depending on fects over AchEIs monotherapy (Hartmann and Mobius, 2003;
the response criteria used, that a considerable portion of pa- Tariot et al., 2004; Gauthier et al., 2005; Dantoine et al., 2006;
tients do not respond (Lanctot et al., 2003; Burns et al., van Dyck et al., 2006; Cummings et al., 2006).
2006; Birks, 2006). If improvement of at least two points in Memantine is an NMDA receptor channel blocker and this
the Mini-Mental State Examination (MMSE) is required for MOA for an anti-dementia drug is, at first sight, not easy to
the definition of response, less than 20% of patients were clas- comprehend bearing in mind the accepted crucial permissive
sified as responders after 9 months of treatment (Raschetti role of NMDA receptors in synaptic plasticity (Morris et al.,
et al., 2005). With the advent of memantine a new therapeutic 1986; Herron et al., 1986; Danysz et al., 1995; Collingridge
treatment was offered. It is likely, that because of the different and Bliss, 1995). This apparent paradox can be explained by
mechanism of action (MOA) of memantine such patients bringing analogy to magnesium, the endogenous NMDA re-
would respond to this therapy, although, this has not been sys- ceptor antagonist, which is vital for normal neuronal function-
tematically studied. ing and for neuronal plasticity (Nowak et al., 1984; Coan
Memantine’s mechanism of action, which differs from the et al., 1989; Parsons et al., 1999b; Danysz and Parsons,
major alternative therapies in AD which are all AchEIs, is 2003). The present review is aimed at elaborating this apparent
also a clear rationale for combination therapy. Additionally, paradox and providing a plausible hypothesis based on our
the most frequent side-effects of AchEIs (nausea, vomiting, current knowledge of memantine’s pharmacology and pharma-
and diarrhoea) are not shared by memantine and are linked cokinetics. A number of previous reviews on memantine have
to the actions of AchEIs on peripheral cholinergic synapses. been published tackling different preclinical and/or clinical
They may present in more than 10% of patients and may aspects (Danysz et al., 1997, 2000; Parsons et al., 1999b;
also lead to treatment discontinuation due to tolerability Molinuevo, 2003; Ferris, 2003; Danysz and Parsons, 2003;
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 701

NR1111 NR2
NH2 NH2
S1 S2 S1 S2
N1

M M M M M M
1 3 4 1 3 4

N N
M M
2 2

P
HOOC
C2
C1 COOH
Glycine P
Glutamate

N asparagine 598 N asparagine 595/596

S1 - glycine binding site S1 - glutamate binding site

S2 - glycine binding site S2 - glutamate binding site


P Phosphorylation by PKA (serine 897)
P Phosphorylation by CaMKII (serine 1303)
and by PKC (serine 890,896)

Splices

Fig. 2. Schematic of the topology and pharmacological recognition of individual NMDA receptor subunits. NR1 and NR2 subunits have four membrane-inserted
domains. M1, M3 and M4 are all true transmembrane domains (TMD) whereas M2 makes a hairpin bend and forms the narrowest channel domain containing an
asparagine residue (N) at the Q/R/N site which determines channel permeation/block by cations. This residue is found at position 598 on NR1 subunits and position
595 or 596 on NR2 subunits and is believed to experience 80% of the transmembrane electric field. The agonist recognition site or ligand binding domain (LBD) is
formed by a ‘‘Venus fly trap’’ juxtaposition of an S1 site on the extracellular domain attached to M1 and an S2 site on the extracellular loop between M3 and M4
for both NR1 and NR2 subunits. Alternative splicing generates eight isoforms for the NR1 subfamily. The variants arise from splicing at three exons: one encodes
a 21-amino acid insert in the N-terminal domain (N1, exon 5), and two encode adjacent sequences of 37 and 38 amino acids in the C-terminal domain (C1, exon 21
and C2, exon 22). Rat NR1 variants are sometimes denoted by the presence or absence of these three alternatively spliced exons (N, C1 and C2). NR1111 has all
three exons, NR1000 has none, and NR1100 has only the N-terminal exon. The most common forbrain splice variant in the adult rat CNS is NR1011. Additionally,
receptor function/trafficking, etc., can be influenced by phosphorylation by PKA, PKC and CaMKII as well as by glycosylation.

Rogawski and Wenk, 2003; Lipton, 2006; Wenk et al., 2006; Cognitive performance was measured by the cognitive sub-
Bullock, 2006). For aspects not covered by the present review, scale of the Alzheimer’s Disease Assessment Scale (ADAS-
e.g. details of studies related to neuroprotection or aspects out- cog; Rosen et al., 1984) in the studies in mild-to-moderate
side of the indication dementia, readers are encouraged to use AD as well as in the vascular dementia studies. In the moder-
the comprehensive reviews. ate-to-severe AD studies the Severe Impairment Battery (SIB;
Table 1 chronologically lists milestones in the development Saxton et al., 1993; Schmitt et al., 1996) was used, which was
of our knowledge related to memantine focusing on, or sup- specifically developed to measure cognition in more advanced
portive for, symptomatological actions in AD which is the cur- stages of AD. Two vascular dementia studies (Orgogozo et al.,
rent therapeutic label. 2002; Wilcock et al., 2002) showed a statistically significant
Since the present review discusses mechanisms of meman- better cognitive performance vs. placebo after 6 months of
tine-induced positive (symptomatological) effects on cognition treatment (while the effects on the clinical global ratings of
in dementia, evidence that such an effect has indeed been change were not significant).
proven in placebo controlled clinical trials is crucial. The ef- In AD, six, 6-month Phase III trials were conducted in the
fects of memantine on cognition in AD and vascular dementia USA and Europe ((Reisberg et al., 2003; Tariot et al., 2004;
have been investigated in more than 3000 patients. A number Bakchine et al., 2005; Peskind et al., 2006); for two trials,
of randomized controlled clinical trials in the indications of MD-01 and MD-12, see Forest Labs, Clinical Trials Registry).
AD and vascular dementia were performed in the last decade All studies showed a strong tendency for improvement by
that assessed cognition using standard performance-based in- memantine treatment over placebo, while three demonstrated
struments. Apart from a cognitive benefit, these clinical trials clear statistical significance, and the beneficial effects on cog-
also showed positive effects of memantine upon activities of nition were also obvious in various meta analyses (Doody
daily living and behavioural disturbances of the AD patients et al., 2005; McShane et al., 2006). For two of the AD trials
as compared to placebo (Doody et al., 2004; Gauthier et al., (Reisberg et al., 2003; Tariot et al., 2004), the cognitive effects
2005; Cummings et al., 2006). are reviewed in more in detail in separate papers (Schmitt
702 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Table 1
Development of knowledge and concepts on memantine (focus on cognitive effect in dementia)
Year Finding References
1963 First description of memantine as part of a synthetic pathway. Gerzon et al. (1963)
1989 Inhibition of [3H]MK-801 binding by memantine in human postmortem cortical tissue. Kornhuber et al. (1989)
Description of NMDA antagonistic activity of memantine in patch clamp experiments. Bormann (1989)
1991 Description of positive effects of memantine on cognition in demented patients. Ditzler (1991)
1993 Importance of voltage-dependency of memantine for clinical tolerability postulated and Parsons et al. (1993)
similarity to the endogenous NMDA antagonist Mg2þ emphasized. Signal-to-noise concept
as an explanation for symptomatic improvements in AD discussed for the first time.
1993 Review emphasizing importance of affinity for tolerability of memantine and related agents. Rogawski (1993)
1994 Demonstration of behavioural differences between memantine and high affinity NMDA Danysz et al. (1994)
channel blockers.
1994 Demonstration of NMDA antagonism in an in vivo preparation. Herrero et al. (1994)
1994 Study showing free brain memantine levels close to affinity at NMDA receptors. Spanagel et al. (1994)
1995 Data on concentration of memantine in plasma and CSF of patients. Kornhuber and Quack (1995)
1995 Comparison of a series of NMDA channel blockers emphasizing the importance of voltage-dependency Parsons et al. (1995)
and kinetics.
1996 Study showing that memantine may improve neuronal plasticity and learning in old animals. Barnes et al. (1996)
1996 Demonstration that memantine is relatively less potent in blocking neuronal plasticity (LTP) than Frankiewicz et al. (1996)
NMDA receptors as compared to the high affinity NMDA antagonist MK-801.
1996 Demonstration of learning enhancement in rats with learning deficit produced by entorhinal cortex lesion. Zajaczkowski et al. (1996b)
1997 Review describing non-published and published data on the in vitro activities of memantine at numerous Danysz et al. (1997)
receptors and relating pharmacokinetics of memantine to pharmacodynamics.
1997 Importance of partial trapping for tolerability of memantine proposed. Blanpied et al. (1997)
1997 Experimental basis for the concept that overactivation of NMDA receptors impairs neuronal plasticity Zajaczkowski et al. (1997)
(LTP) and learning which are restored by memantine, as suggested through a decrease in synaptic noise.
1998 Efficacy shown in mixed vascular and Alzheimer dementia patients. Winblad and Poritis (1998)
1999 Noise reduction hypothesis of memantine symptomatological action substantiated by experiments showing Frankiewicz and Parsons (1999)
restoration of synaptic plasticity (LTP) under low-magnesium concentrations.
1999 Quantitative analysis of relation between memantine plasma, CSF, ECF and brain homogenates concentration Hesselink et al. (1999a)
using in vivo recovery.
1999 Review describing pharmacology of memantine and hypothesis on the mechanism of action. Parsons et al. (1999b)
2000 Description of the signal-to-noise hypothesis explaining both neuroprotective and symptomatological effects Danysz et al. (2000)
of memantine.
2001 Support for voltage-dependent NMDA channel blockade by memantine in vivo. Jones et al. (2001)
2003 Review on proposed MOA of memantine. Danysz and Parsons (2003)
2003 Study showing cognitive improvement vs. placebo in AD patients treated with memantine. Reisberg et al. (2003)
2003 Review on proposed MOA of memantine. Rogawski and Wenk (2003)
2004 Memantine shown to improve spatial learning in transgenic (PS1/Aß) mice. Minkeviciene et al. (2004)
2004 Memantine showed no change of AchE inhibition by rivastigmine and donepezil in vivo. Enz and Gentsch (2004),
Periclou et al. (2004)
2006 Memantine improves water radial maze learning in rats. Zoladz et al. (2006)

et al., 2002, 2006). It is an open issue whether some of the colleagues an acute dose of 30 mg was used. Note that both
effects on cognition may be attributed to a disease-modifying in LTP experiments and in in vivo studies in experimental an-
effect of memantine (slowing of decline). But in Reisberg and imals memantine showed a bell-shaped doseeresponse curve,
Tariot trials (Reisberg et al., 2003; Tariot et al., 2004) there showing even impairment at the higher doses (Frankiewicz
was a separation in SIB scores from placebo already after and Parsons, 1999; Zajaczkowski et al., 1997). Thus, it is
12 and 8 weeks, respectively, and this is indicative of an early likely that given the ‘‘lack of pathological conditions’’, the
symptomatic benefit on cognition for memantine treatment vs. higher dose used, and the lack of up-titration, the effects
placebo. seen reflect the right end of the bell-shaped doseeresponse
Rammsayer and colleagues (Rammsayer, 2001) reported curve with memantine.
that memantine impaired object recognition but not face rec- In summary, these clinical data provide clear evidence that
ognition performance in healthy volunteers. This, at first memantine is able to improve cognition in demented patients
glance seems, to contradict the hypothesis discussed below. relative to placebo.
However, two factors should be taken into account. First, the
study was performed in healthy volunteers, not AD patients 2. PDePK relationship e possible memantine targets
with presumed glutamatergic hyperactivity. In turn, since there
was no hyperfunction to correct, this is a plausible reason for 2.1. NMDA receptors
the lack of positive effects. Second, in contrast to typical clin-
ical use where the daily dose of 20 mg/day is achieved follow- Memantine inhibits NMDA receptors at near resting mem-
ing a 4-week titration, in the study by Rammsayer and brane potentials with an approximate affinity of 1 mM (Parsons
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 703

et al., 1999b). The correspondence between pharmacody- et al., 1994; Misztal et al., 1996; Danysz et al., 1997;
namics (PD, in vitro targets) and pharmacokinetics (PK, extra- Chen et al., 1998).’’ The cited papers Wenk et al. (1994)
cellular levels in the brain) is a prerequisite to claim any and Misztal et al. (1996) all used infusion of 20 mg/kg/
therapeutic MOA. In other words, active doses both in clinical d which leads to maximal plasma level which are at least
studies and in animal models should lead to levels at the locus four times lower than the peak concentration achieved af-
of action (here, the central nervous system, CNS), which are ter a single i.p. administration of the same dose. Readers
close to the concentrations active at the claimed target (here, are referred to our review (Danysz et al., 1997) which ex-
NMDA receptors). This is especially the case for ionotropic plicitly states that, if acute administration is used, then
receptors where factors such as receptor reserve seem to be 5 mg/kg i.p. is the maximal therapeutically relevant dose
less pronounced than for other targets such as G-protein cou- for 30e60 min time point. A similar oversight was made
pled receptors (GPCRs). by Chen et al. (1998).
Microdialysis experiments, with in vivo recovery correc-
tions, are the best method to access free extracellular levels Thus, a maximal acute dose of 5 mg/kg i.p. in rats can
in the brain. Following administration of behaviourally active probably be considered to be of therapeutic relevance for its
doses in rats, e.g. infusion of 20 mg/kg/d using Alzet osmotic use in AD, taking the peak concentration (c.a. 30 min) as
pumps (for review see e.g. (Danysz et al., 1997; Parsons et al., the reference point (Zoladz et al., 2006), although age, strain,
1999b)), free extracellular levels in the brain are similar to free gender, and health status of animals should also be considered
plasma levels (Hesselink et al., 1999a) and these are close since they can change pharmacokinetics considerably. For ex-
to plasma concentrations seen in patients, i.e. around 0.5e ample older rats and female rats generally show higher free
1 mM (Quack et al., 1995; Kornhuber and Quack, 1995). In peak concentrations whereas young adult male rats were
the case of acute injection the picture is much more compli- used for most preclinical pharmacodynamic studies to date
cated since the plasma/brain ratio also depends on the time (see below).
of assessment (Hesselink et al., 1999c). Nevertheless, acute The estimate of the therapeutically relevant acute dose in
administration of memantine at 10 and 20 mg/kg i.p. in rats young adult male rats is based on the following rationale:
leads to respective peak free brain concentrations of 1.2 and (a) the difference in drug sensitivity between man and rat is
2.6 mM assessed by microdialysis in the striatum (Quack mainly due to pharmacokinetic rather than pharmacodynamic
et al., 1995). A similar study using in vivo recovery revealed aspects; (b) it is very likely that the serum/extracellular brain
somewhat higher peak levels, i.e. 4 mM levels after 20 mg/kg ratio is similar for man and rats and the CSF/total serum ratio
(Hesselink et al., 1999c). is c.a. 0.5 for both rat and man (Kornhuber and Quack, 1995;
There are two important aspects related to memantine phar- Hesselink et al., 1999a).
macokinetics that have, too often, led to confusion: Bearing in mind these considerations, 5 mg/kg i.p. in the rat
is the acute dose giving rise to serum concentrations at 20e
1. The fact that brain levels assessed in homogenates are very 30 min which correspond to the upper limit of those seen in
different from concentrations in the extracellular fluid the serum of patients and healthy volunteers following treat-
should be appreciated, i.e. the difference is over 30-fold ment with well-tolerated doses of memantine. More relevant
in favour of homogenates (Hesselink et al., 1999a). This for the clinical use of memantine is repetitive administration.
is due to intracellular, and in particular lysosomal accumu- However, considering the much shorter half-life in rat (3e
lation (Honegger et al., 1993) and does not reflect free 5 h) than in humans (up to 100 h) repetitive treatment in rats
concentrations available at CNS plasma membrane recep- would result in substantial fluctuations in brain concentrations.
tors in vivo (Wesemann et al., 1980; Wesemann and This would be entirely different from clinical practice where
Ekenna, 1982; Kornhuber and Quack, 1995; Danysz stable steady-state levels are present in chronically treated pa-
et al., 1997). This has sometimes been overlooked by tients. It is known that treatment regimes with fluctuating
some researchers (Chen et al., 1998) who stated that ‘‘a levels lead to very different pharmacodynamic changes than
20 mg/kg i.p. memantine administration to the rodent re- those with stable concentrations (e.g. Hesselink et al.,
sults in a brain concentration of 1e10 mM within 30e 1999b). One of the ways to overcome this problem is the
60 min’’. These authors cited the reference Wesemann use of Alzet osmotic minipumps for s.c. infusion. Such treat-
et al. (1982) that presented concentrations in brain homog- ments using memantine (20 mg/kg/d) lead to plasma levels of
enates and then determined values of over 100 mM after an c.a. 1 mM (Misztal et al., 1996; Zajaczkowski et al., 1996b;
i.p. dose of 20 mg/kg (not 1e10 mM) (Fig. 2 of the cited Nakamura et al., 2006) (see Table 2).
paper, 1 h time point), but this does not reflect extracellu- Free cerebrospinal fluid (CSF) levels of memantine after
lar levels, which are over 30-fold lower. therapeutic doses in man are within the range of affinity for
2. It is not always recognised that infusion of 20 mg/kg/d is NMDA receptors in vitro (Kornhuber and Quack, 1995). Phar-
not comparable with acute i.p. injection of the same dose. macokinetic studies together with extensive screening for
For example, Creeley et al. (2006) wrote ‘‘we chose many targets imply that, apart from NMDA receptors, four
20 mg/kg as the initial dose because this is the dose that other candidates could potentially play a role in memantine’s
others have reported is effective in protecting the adult MOA, i.e. alpha7, alpha4/beta2, alpha9/10 and 5-HT3 recep-
rat brain against excitotoxic neurodegeneration (Wenk tors. Before the MOA hypotheses related to NMDA receptor
704 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Table 2
Compilation of studies showing neuroprotective activity of memantine, assessed plasma level and projected brain levels at the neuroprotective dose
Type of insult Parameter assessed Effective dose Plasma concentrations Projected brain/ECF References
(mM) at active dose concentrations (mM)c
Aggregated Aß and Histology (cresyl 10 and 20 mg/kg/da 0.7 and 1.4b 0.6 and 1.1 Nakamura et al. (2006)
ibotenate injection in violet), and Morris
the hippocampus in maze learning
rats
APP23 mice Morris maze learning 14.4 mg/kg/d infusion ed e Van Dam and De Deyn
(2006)
Aggregated Aß Bcl2 immunostaining, 15 mg/kg/d infusion 1.0 0.8 Miguel-Hidalgo et al.
injection in the DNA fragmentation, (2005)
hippocampus in rats caspase 8 activity and
active avoidance
learning
Aggregated Aß DNMTS e operant 20 mg/kg/d infusion 1.2e 1.0 Yamada et al. (2005)
injection in the chamber, histology CA1
hippocampus in rats
NMDA or 3-NP Cortical ChAT activity 20 mg/kg/d infusion 0.9 0.7 Wenk et al. (1996)
infusion in the NBM
in rats
LPS infusion into the ChAT activity in the 20 mg/kg/d infusion 1.2e 1.0 Willard et al. (2000)
NBM in rats frontal cortex
Aß into frontoparietal ChAT staining, 35 mg/kg/d in drinking 1.6f,g 1.3 Nyakas et al. (2006)
cortex in rats microglia activation, water
object recognition test,
passive avoidance,
Morris water maze
APP/PS1/Tau triple Learning in Morris 30 mg/kg/d po for 3 ? ? Luhrs et al. (2006)
transgenic mice at 12 maze and object months in drinking
month recognition water
Negative effects on learning are not seen at plasma levels providing neuroprotection indicating that, in contrast to a claim by Creeley et al. (2006) e it is possible
to achieve neuroprotective effects through NMDA receptor blockade without negative effects on learning. Please note that memantine affinity at NMDA receptors
is approximately 1 mM.
a
Note that in this study the same dosing regime did not impair Morris maze learning when administered to naive rats.
b
Not assessed in this study but taken from Misztal et al. (1996) using the same gender, strain of rats of similar age.
c
Assumed to be 80% of free plasma levels (Hesselink et al., 1999a).
d
In a previous study it has been shown that in the same tg model memantine at 10 mg/kg had no negative effect on cognitive performance (Van Dam et al., 2005).
e
Not assessed in this paper but, based on rats’ gender and weight, predicted to be around the indicated value, in this study memantine treatment after the insult
had no effect on performance of control or Aß treated rats rats in the DNMTP task.
f
No effect of memantine was observed in mice at doses providing neuroprotection against Aß in the object recognition test, passive avoidance and Morris water
maze.
g
This value refers to plasma levels obtained in a pilot study using 40 mg/kg/d. In turn, the dose of 35 mg/kg/d was selected for the main experiments. Expected
plasma level would be then c.a. 1.2 mM.

blockade are discussed in detail, it seems fair that the other et al., 2004, 2005; Elgoyhen, unpublished data) and 5 mM
potential targets are first addressed. These possibilities are dis- observed in heterologously expressed human receptors in
cussed and largely dismissed below. Xenopus oocytes (Maskell et al., 2003), the latter finding mak-
ing it also seem to be less likely to be of real human therapeutic
2.2. Alpha7 and alpha4/beta2 nicotinic receptors relevance in AD, although here it should be stressed that poten-
cies can be influenced by the heterologous expression system
The possibility that therapeutically relevant doses of mem- used. The effect of memantine on all tested ‘‘neuronal’’ nico-
antine could affect alpha7 and alpha4/beta2 nicotinic receptors tinic receptors was only weakly voltage-dependent, as is the
has recently attracted considerable attention from focused case for 5-HT3 receptors (see below) e which often share a
groups (Buisson and Bertrand, 1998; Maskell et al., 2003; common pharmacology with neuronal nicotinic receptors e
Aracava et al., 2004, 2005). and implies a site of action which is not deep inside the channel.
Memantine has been reported to inhibit human alpha4/ Memantine produces discriminative effects indicative of
beta2 responses with an IC50 ¼ 6.6 mM (Buisson and Bertrand, NMDA antagonism that are shared neither by 5-HT3 blockade
1998), however, this level is probably too high to be of real nor by alpha7 antagonism indicating that NMDA antagonism
therapeutic significance. The reported potency of memantine predominates in rats at relevant doses. In a drug discrimination
at alpha7 receptors varies considerably from 0.33 to 1.68 mM paradigm, memantine exhibited generalization to other recep-
seen in native/heterologously expressed rat receptors (Aracava tor NMDA antagonists (Grant et al., 1996; Zajaczkowski et al.,
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 705

1996a) but produced only modest attenuation (33% decrease) impairment’’. Such a generalized statement that blockade of
of a nicotine discriminative cue at 10 mg/kg (this acute dose is NMDA receptors must result in very serious CNS-mediated
over the therapeutic range) as compared to a 70% decrease side-effects is not substantiated by experimental evidence,
with the non-selective neuronal nicotinic antagonist mecamyl- even at the most basic level of our understanding of NMDA
amine at 1 mg/kg (Zakharova et al., 2005). receptor physiology and pharmacology. The authors ignored
Moreover, acute administration of therapeutically relevant the fact that magnesium is an NMDA receptor antagonist
dose of 5 mg/kg memantine had no effect on dopamine levels and it is present in the brain at concentrations fully blocking
in the striatum, and higher doses produced an increase (Spana- NMDA receptors at resting membrane potentials (Nowak
gel et al., 1994), that would be inconsistent with blockade of et al., 1984). Magnesium is neuroprotective since its removal
alpha7 receptors which produces a decrease (Alessandri-Haber produces neurodegeneration (Novelli et al., 1988) and at the
et al., 2006). This indicates, at least, that the NMDA receptor same concentrations magnesium does not produce side-effects,
blocking activity predominates. and in particular, it does not impair learning. In complete con-
trast, removal of magnesium actually impairs neuronal plastic-
2.3. Alpha9/alpha10 nicotinic receptors ity (Coan and Collingridge, 1987).
Moreover, kainate-induced excitotoxicity e which was used
Although memantine blocks alpha9/alpha10 nicotinic re- by Creeley et al. (2006) as a model to support this point e is
ceptors with similar affinity to NMDA receptors (Oliver an acute insult not compatible with AD and chronic neurode-
et al., 2001), these receptors show a very discrete, non-CNS, generation in general. In turn, in their studies, the neuroprotec-
distribution, i.e. in cochlear hair cells, and this action is there- tive potency of memantine was predictably low in contrast to
fore highly unlikely to be responsible for therapeutic effects of previous studies (see Table 2 and Wenk et al. (2006)). Addi-
memantine on symptoms in AD (Elgoyhen et al., 2001; Katz tionally, higher plasma levels of memantine than in most
et al., 2004). previous studies may be expected in this study due to the use
of female rats and older animals (6e8 month; see Table 2
2.4. 5-HT3 receptors and discussion above). This is because memantine metabolism
and/or elimination is slower in female rats and also decreases
Memantine blocks murine and human 5-HT3 receptors with with age. The former difference has been shown after infusion
similar or somewhat lower affinity to NMDA receptors in vi- or acute injection of memantine, where serum levels were up to
tro, but in a non-use, non-voltage-dependent manner (Rammes two times higher in female rats (Zajaczkowski et al., 2000) e it
et al., 2001). However, at doses of up to 10 mg/kg, memantine should however, be noted that differences do not apply to
does not block vomiting evoked by cisplatin in ferrets (Naylor, humans. Regarding the age effects, there are no published
internal Merz report), a reaction sensitive to blockade by 5- data showing such comparison, however, several unpublished
HT3 antagonists such as ondansetron. However, as a word of studies have been performed. For example memantine infusion
caution, it should be kept in mind that the pharmacokinetics at 24 mg/kg/d for 1 week using Alzet 2ML2 pumps implanted
of memantine in ferrets is not known. under the back skin produced 4.35  1.64 mM plasma levels in
In conclusion, effects on other neurotransmitter systems 24 month SD male rats while concentrations of only 1.28 
may, to some extent, be involved in the good therapeutic pro- 0.24 mM (Mean  SD, N ¼ 5) were observed in 3 month old
file of memantine, but the specific mode of NMDA receptor animals. Memantine concentration was assessed using gas
antagonism is, in our view, the main MOA that is responsible chromatography.
for both the pharmacodynamic effects and the tolerability of In turn, possibly the acute dose of 2.5 mg/kg used by Cree-
memantine seen in AD. ley et al. (2006) may correspond to higher dose in studies per-
formed in young adult male rats when plasma concentration is
2.5. Can neuroprotective effects of memantine be considered. This could explain impairment of learning at ap-
expected at tolerable doses that block NMDA receptors? parently moderate doses.
A compilation of studies showing neuroprotective effects of
Although a detailed discussion of neuroprotective potential memantine in relation to plasma levels is shown in Table 2.
of memantine is beyond the scope of the present review, one Note that free brain levels are approximately 80% (brain mi-
aspect is discussed here as it is pertinent to our hypothesis un- crodialysis) of total plasma levels (Hesselink et al., 1999a).
derlying the clinically proven symptomatic effects of meman- As can be seen from this table, memantine was neuroprotec-
tine on cognition. This hypothesis assumes that NMDA tive in various insult models (relevant for AD) at doses that
receptor blockade may, under certain conditions, both provide did not produce impairment of learning in control animals (in-
neuroprotection and improve neuronal plasticity via the same dicated studies in the Table 2) and led to projected concentra-
MOA. In a recent paper, impairment of learning by memantine tions in the brain close to the affinity at NMDA receptors.
was observed at 2.5 mg/kg and much higher doses were
needed to attenuate kainate-induced toxicity (Creeley et al., 3. NMDA receptors and neuronal plasticity
2006). Based on this observation, Creeley et al. (2006) stated
that ‘‘it is impossible to block NMDA receptors sufficiently to Although, as discussed in detail below, it is widely accepted
provide neuroprotection without side-effects such as memory that phasic, contingent synaptic NMDA receptor activation is
706 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

necessary for learning-related synaptic plasticity, the glutama- and 5 mg/kg (acute doses indicated above to be close to the
tergic system requires a finely balanced homeostatic control. therapeutic range), but not at 10 mg/kg. The bell-shaped
Under certain conditions, overactivation of these receptors dose-dependency of this effect of memantine was highly rem-
may, in fact, lead to an impairment and their inhibition may iniscent to that seen with LTP in hippocampal slices (Frankie-
actually lead to enhancement of synaptic plasticity/learning. wicz and Parsons, 1999). Similarly, in mice maintained on
Below a compilation of arguments supporting this thesis is a low-magnesium diet and showing other signs of deficiency
provided with emphasis on data with memantine. It shows of this physiologically very important cation, a deficit of con-
that the assumption that any blockade of NMDA receptors un- ditioned context freezing was observed that could not be at-
equivocally leads to impairments of neuronal plasticity/learn- tributed to unspecific effects on activity, exploration, or pain
ing (and NMDA receptor activation to the opposite effects) is sensitivity (Bardgett et al., 2005). Unfortunately, the potential
a rather drastic over simplification. reversal of this effect by an NMDA receptor antagonist was
not tested in this study.
3.1. NMDA receptor activation can impair, and blockade In a lever-pressing instrumental task, NMDA (20e50 mg/
can improve, neuronal plasticity and learning kg) decreased accuracy in both the repeated acquisition and
performance components of a lever-pressing paradigm (Cohn
3.1.1. Long-term potentiation (LTP) and Coryslechta, 1994). The disruption of repetitive acquisi-
Since some forms of LTP are NMDA receptor-dependent tion accuracy was related to initial perseverative errors fol-
(see below), complete inhibition of these receptors also in- lowed by errors of skipping forward and backwards in the
hibits the induction of LTP, but factors such as antagonist three-member response sequence. Again, NMDA receptor an-
type and their concentrations play a crucial role. For example, tagonists were unfortunately not tested in this study.
in moderately aged rats, memantine actually prolonged the NMDA receptor antagonists can, under certain conditions,
duration of LTP in vivo (Barnes et al., 1996). Similarly, the paradoxically improve learning. Most studies supporting
competitive NMDA receptor antagonist CPP (3-((þ)-2- such statement have been discussed in a previous review (Da-
carboxypiperazin-4-yl)-propyl-1-phosphonic acid) displayed nysz et al., 1995) but selected studies, in particular those using
biphasic effects on LTP in CA1 in vivo e a low dose of memantine, are discussed below. In a recent study using a low
1 mg/kg caused a borderline significance potentiation of LTP level of training in the water radial maze followed by a test
whereas a higher dose of 10 mg/kg reduced LTP (Abraham trial 24 h later, memantine (at 5 and 7.5 mg/kg but not
and Mason, 1988). In contrast, MK-801((þ) 5 methyl 10,11 3.75 mg/kg) improved the performance when given after the
dihydro 5H dibenzo[a,d]cyclohepten 5,10 imine, 0.1 to training (Zoladz et al., 2006). It was suggested by the authors
1 mg/kg) showed only a dose-dependent inhibition of LTP that the poor performance in this model is related to the effect
(Abraham and Mason, 1988). of stress on memory storage and involves overactivity of the
Removal or reduction of magnesium in hippocampal slices glutamatergic system with resulting overactivation of NMDA
causes deficits in LTP (Coan and Collingridge, 1987) and this receptors. Another study confirmed this hypothesis of the im-
could be reversed by memantine with a bell-shaped concentra- pact of stress and also showed that the glutamate transport in-
tioneresponse curve with near complete reversal of this deficit hibitor (DL-TBOA) produced a similar impairment of learning
by therapeutically relevant concentrations (Frankiewicz and and LTP which may depend on NMDA receptor activation
Parsons, 1999). Similarly, in hippocampal slices, low concen- (Howland et al., 2006). The data also suggest that both effects
trations of NMDA also caused a reduction of LTP in the CA1 may be related to an enhancement of LTD (long-term depres-
region (Izumi et al., 1992; Zajaczkowski et al., 1997). This sion). In fact, learning impairment (Morris water maze) and
deficit was also reversed by memantine at a concentration of LTD enhancement were prevented by the NR2B antagonist
1 mM (Zajaczkowski et al., 1997). Finally, in knockout mice Ro25-6981. Also in rats with thiamine deficiency, a decrease
lacking the glutamate transporter GLT-1, LTP was impaired of glutamate uptake in the prefrontal cortex was observed
in the hippocampal CA1 region in vitro. When tetanic stimu- and was accompanied by a deficit in Morris water maze
lation was applied in the presence of a very low concentration learning (Carvalho et al., 2006) giving another example of dis-
of the competitive NMDA receptor antagonist AP5 (0.5 mM), ruption of learning likely resulting from non-contingent over-
the impairment was reversed (Katagiri et al., 2001). activity in glutamatergic systems.
In rats with learning deficits following an excitotoxic lesion
3.1.2. Learning of the entorhinal cortex, memantine delivered by osmotic
As early as in 1989 Jones et al. demonstrated that systemic pumps (20 mg/kg/d) after the lesion, improved reference but
administration of NMDA, given before acquisition, dose- not working memory learning in the radial maze (Zajaczkow-
dependently (3e50 mg/kg) impaired passive avoidance learn- ski et al., 1996b). This effect was unlikely due to neuroprotec-
ing retention in rats tested 24 h later. In another study, NMDA tive activity per se, but could reflect suppression of ongoing,
also impaired passive avoidance learning starting at 25 mg/kg residual noise at the remaining, perturbed glutamatergic
when given before acquisition but not directly after or before synapses.
the retention test (Zajaczkowski et al., 1997). This effect was In F344 9e12 month old rats, memantine produced a strong
not due to state-dependency and was not related to toxic trend for improvement in the Morris water maze expressed as
actions. This impairment was reversed by memantine at 2.5 significantly more time spent in the target quadrant during the
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 707

probe trial (Barnes et al., 1996). Interestingly, it has been re- before stress, but improved learning when administered after
ported that in 24-month-old F344 rats, a high magnesium the stress application at 10 mg/kg (Barber et al., 2002).
diet improved T-maze reversal learning and improved high fre- The studies discussed above indicate that NMDA receptor
quency potentiation in CA1 hippocampal region (Landfield antagonists in general may potentially produce a beneficial ef-
and Morgan, 1984). This again implies background NMDA re- fect in AD. However, as discussed above and below, the good
ceptor overactivation as a cause for this observed symptomatic tolerability and specific features of NMDA receptor channel
deficit, but the use of magnesium for therapy is questionable blockade by memantine seem to render memantine particu-
(see below). larly suitable for this indication. It should however, be men-
In another study, the effects of memantine treatment at tioned that although clear superiority of memantine over the
2 mg/kg given for 23 days and starting 1 day after i.c.v. cho- high affinity uncompetitive antagonist MK-801 has been
linergic toxin injection of AF64A (aziridinium ion of ethyl- shown in LTP experiments (Frankiewicz and Parsons, 1999),
choline) were assessed in the Morris maze test. On test days in vivo studies provided mixed results (Zajaczkowski et al.,
11 and/or 14 memantine tended to decrease the latency and 1997; Lukoyanov and Paula-Barbosa, 2001; Si et al., 2004).
significantly improved the ‘‘straightness index’’ (Bachurin
et al., 2001). It is not clear to what extent this effect can be at- 3.2. The NMDA receptor as a co-incidence detector in
tributed to the functional improvement of memory or to neuro- Hebbian learning with magnesium block governing the
protective/restorative activity since it extended over 10 days filter function
beyond the treatment period (Bachurin et al., 2001).
In the Morris water maze, rats withdrawn from chronic al- Ionotropic glutamate receptors are ligand gated ionic chan-
cohol treatment also showed a learning deficit that was atten- nels permeable to the monovalent cations Naþ and Kþ and,
uated by memantine but not by MK-801 (Lukoyanov and depending on the subtype, also to the divalent cation Ca2þ.
Paula-Barbosa, 2001), but in this study the rationale for the AMPA (a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
dose-selection design is difficult to follow, i.e. an excessively acid) receptors show very fast activation and inactivation ki-
high bolus dose was followed by a maintenance dose that was netics, are largely postsynaptic, mostly impermeable to Ca2þ
clearly too low. (at least those containing the edited GluR2 subunit) and partic-
There are only few studies in transgenic mice models of ipate in most forms of fast synaptic transmission. In contrast,
AD addressing the issue of symptomatological improvement NMDA receptors are normally only synaptically activated un-
of cognition. In 4-month-old APP23 mice (APP ¼ Amyloid der certain physiological conditions, i.e. during the induction
Precursor Protein) with established cognitive deficits, meman- of synaptic plasticity. The NMDA receptor has three cardinal
tine (10 mg/kg daily) given for 1 week improved performance features that permit its function in synaptic plasticity: high
in the Morris water maze at the end of the training period (Van permeability to Ca2þ ions; voltage-dependent block by
Dam et al., 2005). Similarly, in APP/PS1 (PS1 ¼ preseniline 1) Mg2þ ions and relatively slow gating kinetics.
double transgenic mice, memantine (30 mg/kg p.o. daily for An example of such plastic changes is LTP which can be in-
2e3 weeks) improved acquisition without affecting swimming duced both in vitro and in vivo and is believed to model basic
speed (Minkeviciene et al., 2004). Most recently, LaFerla and mechanisms of memory formation in mammals (Collingridge
colleagues (Luhrs et al., 2006) showed that in triple transgenic and Bliss, 1995). There are both presynaptic and postsynaptic
mice (APP/PS1/Tau), treatment with memantine in drinking forms of LTP. Postsynaptic NMDA receptor-dependent LTP,
water (30 mg/kg for 3 months) improved performance both which is most relevant for the proposed MOA of memantine
in the Morris maze (acquisition and probe trial at 1 and in AD, can be described by the following, very simplified,
24 h) and object recognition. Interestingly, the same treatment sequence of events (Fig. 3) although it should be stressed that
resulted in a decrease in plaque load but this is outside of the many mechanisms are involved in this process.
scope of the present review. Glutamate binds to both NMDA and AMPA postsynaptic
Interestingly, negative effects of NMDA receptor over- receptors, however, only the latter are transiently activated
stimulation on learning can be also seen in other species. during fast normal synaptic transmission since positively
For example, even in the honeybee Apis melifera the glutamate charged Mg2þ ions block the NMDA receptor channel and
transporter inhibitor, L-trans-2,4-PDC (L-trans-2,4-pyrrolidine the activation kinetics of NMDA receptors are also much
dicarboxylate), impaired long-term (24 h), but not short-term slower than those of AMPA receptors (Fig. 3A).
(1 h), memory (Si et al., 2004). This effect was reversed by When a prolonged, high frequency signal (or convergence
memantine (but it is difficult to refer to the ‘‘doses’’ used) of several signals) arrives at a glutamatergic synapse, this
but not by MK-801 which produced impairment not only fol- leads to a stronger/more prolonged glutamate release and
lowing L-trans-2,4-PDC but also on its own. a more pronounced influx of Naþ ions into the postsynaptic
Similarly, in 1-day-old chicks, stress lead to learning impair- neuron via AMPA receptors (Fig. 3B).
ment which was suggested by the authors to be due to enhanced Additionally, slow activation of presynaptic GABAB
glutamatergic activity. In this model, in non-stressed chicks, receptors on inhibitory interneurones causes dis-inhibition of
memantine impaired passive avoidance memory at 5 and GABAergic synapses. Both processes, in turn, lead to a strong,
15 mg/kg but not 10 mg/kg. However, under isolation-induced relatively long lasting (hundreds of milliseconds) decrease
stress conditions memantine did not alter memory when given in membrane potential (partial depolarization). This
708 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

A Normal Synaptic Transmission B Tetanic Stimulation AMPA


Glia Glia
NMDA
Na+ Na+
NMDA
Glia Glia
+ + Subtype
- -
GABAA
Cl- Cl-
GABAB
Glia Glia
Secondary
Messagers

C D Mg2+
Tetanic Stimulation Enhanced Synaptic Transmission
Glia Glia
Glutamate
[mM] 1ms
Na+ Na+ GABA
Glia Glia
+ + Glutamate
Ca2+ uptake
Ca 2+ - -
Na+
Cl- Cl-
Ca2+
Glia Glia
Cl-

Fig. 3. Schematic for the processes involved in the induction of NMDA receptor-dependent LTP. Sequence A to D. See text for details.

depolarization removes blockade of the NMDA receptor chan- synaptic plasticity and memory formation (Coan and Colling-
nel by Mg2þ since the relative charge of the neuronal mem- ridge, 1987). Memantine completely restored normal synaptic
brane is now much less negative for a sufficient time to plasticity functionality following reduction in Mg2þ concen-
allow this divalent cation to leave the channel during the tration (Frankiewicz and Parsons, 1999) and this effect was
slower activation of NMDA receptors. At this stage, Ca2þ seen at a therapeutically relevant concentration of 1 mM .
ions can freely enter the cell via the NMDA receptor channel
and trigger a cascade of second messenger processes that are
involved in the fixation of increased synaptic strength (neuro- 4. Link between glutamate system disturbances and AD
nal memory formation) (Fig. 3C).
This postsynaptic change is manifested as an enhancement 4.1. Why normal function of magnesium could be
of AMPA receptor-mediated responses which increases subse- disturbed in AD?
quent normal synaptic signals (Fig. 3D).
One should emphasize the crucial physiological role of en- In AD several factors are likely to be involved in the aeti-
dogenous Mg2þ ions in this process which function as a switch ology (causal events) and certainly many factors contribute to
that keep NMDA receptors blocked under ‘normal’ conditions the final pathomechanisms. The latter are more relevant for the
but allow Ca2þ ion flux when the pattern of activation has fea- current discussion and are systemically, but only briefly listed
tures characteristic for those required for learning processes, below. For more extensive discussion of these factors (which is
i.e. temporal and spatial convergence (co-operativity). not the main aim of this review) the readers should refer to one
In dementia, one of the consequences of neuronal energy of the recent reviews focusing on this aspect (Greenamyre,
deficits (Greenamyre, 1991) (see also below) could be a de- 1991; Albin and Greenamyre, 1992; Delatorre, 1994; Beal,
crease in Mg2þ blockade of NMDA receptors (which is 1995, 2000). The possible contribution of energy deficit as
strongly voltage-dependent, i.e. decreases with the degree of a primary deficit in the pathomechanism of AD, although sug-
depolarization) and in turn enhanced chronic, low-level activa- gested by some findings (see Hoyer, 2000; Blass et al., 2000),
tion of this receptor type (for review see, e.g. Parsons et al. is very controversial. Nevertheless, it can be predicted that at
(1999b)). In this regard, it is important to note that memantine least a secondary deficit may occur as a reaction to Aß oligo-
bears only a single positive charge and is therefore thought to mers, oxidative stress, etc.
act as a somewhat more effective surrogate for the divalent In AD energy demand may increase because of:
Mg2þ cations which show an even stronger functional
voltage-dependence (Parsons et al., 1999b). 1. Partial depolarization (Blanchard et al., 2002);
As detailed above, in vitro studies using rat hippocampal 2. Impairments of Ca2þ homeostasis (Mattson et al., 1993);
slices provide evidence that reduction in Mg2þ concentration 3. Increased neuronal activity due to increase in glutamate
induces severe functional perturbations, including complete levels (Hoyer and Nitsch, 1989; Harris et al., 1996;
impairment of LTP, the neuronal model of mammalian Noda et al., 1999);
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 709

4. Increased NMDA receptor sensitivity to glutamate (Wu anticipated. Severe hypermagnesemia may result in loss of
et al., 1995); tendon reflexes and muscle weakness, bradycardia and cardiac
5. Increased oxidative stress (Mecocci et al., 1994; de la arrest, hypotension (due to peripheral vasodilatation), flushing
Torre and Stefano, 2000; Emerit et al., 2004); of the skin, parasympathetic blockade, fixed and dilated pu-
6. Decreased activity of Naþ/Kþ ATPase (Hattori et al., pils, CNS effects such as drowsiness, confusion, respiratory
1998; Kairane et al., 2002; Dickey et al., 2005; Vignini depression and coma (Fung et al., 1995). This is of specific im-
et al., 2006); and portance as impaired renal function, which is quite frequent in
7. Presence of Aß or Tau (Copani et al., 1991; Bowen et al., the elderly, as well as gastrointestinal disorders may, in fact,
1992; Mattson et al., 1993; Keller et al., 1997; Brewer and markedly increase magnesium concentration (Clark and
Wallimann, 2000). Brown, 1992).
Specifically, AD could even be regarded as a clear contra-
In AD energy supply may decrease because of: indication for therapeutic treatment with magnesium. For ex-
ample, it was shown that magnesium triggers pathological
1. Mitochondrial dysfunctions (Blanchard et al., 1993; Me- deposition of Tau-paired helical filaments and the formation
cocci et al., 1994; Hutchin and Cortopassi, 1995; Hoyer, of neurofibrillary tangles (Yang and Ksiezak-Reding, 1999)
1996; Kwong et al., 2006); and may even cause inhibition of acetylcholine release
2. Changes in blood flow (it is still a matter of debate (Fung et al., 1995) and muscarinic responses (Ladner and
whether this is a primary or secondary effect) (Delatorre, Lee, 1999), both of which would worsen cholinergic deficits.
1994; Franceschi et al., 1995; Crawford, 1996; de la Torre These arguments indicate that, while it may be useful to
and Stefano, 2000); and avoid magnesium deficits by careful oral substitution, meman-
3. Decrease in glucose metabolism/supply (Hoyer et al., tine therapy in AD cannot be simply replaced by high doses of
1991; Mielke et al., 1992; Goto et al., 1993; Simpson magnesium.
et al., 1994; Kennedy et al., 1995; Hoyer, 1996; Parpura-
Gill et al., 1997). 4.2. Amyloid beta and NMDA receptors

It should be kept in mind that these are, to a great extent, Amyloid beta (Aß) is the principal constituent of senile
overlapping processes and many are coupled by positive feed- plaques in AD and is believed to impair neuronal function and
back mechanisms, as is the energy balance (Henneberry, cognition, even before the appearance of overt toxicity (Lesne
1989). Increased energy deficit will result in increased energy et al., 2006). It seems at present to be accepted that soluble Aß
demand. oligomers rather than their deposits, i.e. plaques are responsible
If we believe that in AD there is an energy deficit, and there for impairment of synaptic function and neurodegeneration in
are many reports suggesting that this might be the case AD (Golde, 2006). Moreover, such oligomers may be central
(Greenamyre, 1991), one could expect weaker NMDA recep- to AD pathology as they also increase phosphorylation of Tau
tor blockade by Mg2þ. There are several papers connecting which could, then be a secondary effect (De Felice et al., 2007b).
disturbances of magnesium homeostasis to AD (see for review Most of the studies cited below used an undefined species
Durlach, 1990; Glick, 1990; Ozturk and Cillier, 2006) suggest- of Aß, however, it has been recently suggested that oligomers,
ing decrease in magnesium levels. Additionally, it has even in particularly dodecamers (>50 kDa) may be the most reac-
been reported that in AD patients there is a lower brain level tive species in this regard (Walsh and Selkoe, 2004; Klein
of Mg2þ in autopsy brains (Andrasi et al., 2000). Independent et al., 2004; Barghorn et al., 2005).
of whether or not there are changes in Mg2þ concentrations in There are numerous arguments that Aß may increase gluta-
AD, the function of this ion can be disturbed, i.e. weakened by matergic activity, in particular through NMDA receptors (see
the above mentioned energy deficits resulting in partial re- Fig. 4 and reviews by Greenamyre, 1991; Danysz et al.,
moval of Mg2þ block, by even moderate depolarization. 2000; Danysz and Parsons, 2003; Walsh and Selkoe, 2004;
Bearing in mind the facts discussed above, one could argue Klein et al., 2004).
that magnesium could potentially be used to normalize NMDA Inflammatory process may be involved in this enhanced
receptors dysfunction in AD instead of an NMDA receptor an- activity of NMDA receptors through the cascade increase in
tagonists such as memantine (Ozturk and Cillier, 2006). There Aß-activation of microglia, increase in TNFalpha / microglia
are, however, numerous reasons why such a therapeutic ap- activation, oxidative stress, decrease in glutamate uptake/
proach would not work. Firstly, even chronic oral administra- increase in glutamate release, increase in NMDA receptor
tion of magnesium does not produce elevated concentrations sensitivity, increased NMDA receptor activation, which ulti-
either in plasma, red blood cells or in brain, as evidenced mately results in neuronal death (Wenk et al., 2006). Addition-
in vivo using 31P-NMR spectroscopy (Wary et al., 1999). In ally, Lesne et al. (2005) have shown that sub-toxic activation of
fact, in order to obtain significant increases in brain magne- NMDA receptors increases the proportion of Kunitz pro-
sium concentration, very high, potentially toxic parenteral tease inhibitor domain containing APP which favours ß-secre-
doses would be required (Hallak, 1998; see below). tase over a-secretase processing thus enhancing Aß production,
Additionally, multiple side-effects/problems related to i.e. a vicious circle. This vicious cascade could be blocked by
treatment with high parenteral magnesium doses could be memantine at low, 1 mM, concentrations (Floden et al., 2005).
710 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

1 diffusible Aß1e42 oligomers (ADDLs) for 45 min prevented


APP LTP in hippocampal slices well before any overt signs of
cell degeneration (Lambert et al., 1998; Wang et al., 2002).
- More recent publications confirm this finding (Walsh et al.,
+ Ca2+ 2005; Townsend et al., 2006; Puzzo and Arancio, 2006). Ad-
3 ditionally, chronic exposure of hippocampal cultures to solu-
ble Aß oligomers produced abnormal spine morphology and
2 4
ß-Amyloid a decrease in their density. Subsequent consequences of this
Glu such as synaptic deterioration including loss of the spine cyto-
5 -
T + skeletal protein debrin were completely prevented by meman-
+
tine at a concentration of 5 mM (Lacor et al., 2007). Similarly,
Glu
memantine (5 or 10 mM) prevented oxidative stress and
Glia calcium influx produced by Aß oligomers in hippocampal
Fig. 4. Schematic showing how Aß may increase glutamatergic activity and/or neuronal cultures (De Felice et al., 2007a). Interestingly, Aß
NMDA receptor sensitivity, while soluble APP may have opposite effects. Un- co-immunoprecipitate with extracellular domain of NR1 sub-
der normal conditions, APP has functionally antagonistic effects on NMDA re- unit suggesting direct interaction with NMDA receptors.
ceptors (1) and enhances glutamate transport activity removing glutamate from Moreover, smaller fragments of the Aß peptide have similar
the synaptic cleft (2). When APP is falsely processed to ß-amyloid, this has the
opposite effects on NMDA receptors (3) and glutamate uptake (4) thereby in-
effects. Aß25e35 was also found to impair both posttetanic po-
creasing glutamate concentrations in the synaptic cleft (5) and may even drive tentiation (PTP) and LTP in the hippocampal CA1 in vitro and,
glutamate transporters in to reversed uptake mode. Numbers refer to selected, in agreement with previous data (Wang et al., 2004b), these ef-
example references: (1) Furukawa and Mattson (1998); (2) Masliah et al. fects were proposed to involve activation of the JNK signalling
(1998); (3) Mattson et al. (1993), Wu et al. (1995); (4) Harris et al. (1996); pathway (Costello and Herron, 2004). Similarly, 0.1 mM of the
and (5) Noda et al. (1999).
short Aß fragment Aß31e35 suppressed the induction of LTP of
population spikes (PS) in CA1 of rat hippocampal slices in a
Detailed discussion of this aspect would require a separate similar manner to the longer fragment Aß25e35, whereas neither
review, thus, only selected in vitro papers in relation to LTP/ treatment changed the amplitude of the baseline population
synaptic plasticity are discussed below e for other reviews spikes. These fragments had no effect on NMDA receptor-
please see Rowan et al. (2003, 2004, 2005). mediated multiple PSs when recorded in Mg2þ-free medium,
In rat hippocampal slices, a non-toxic concentration of which could suggest that these Aß fragments suppress the
Aß1e40 (200 nM) caused rapid inhibition of LTP in CA1 whilst induction of LTP through an NMDA receptor-independent
having no long-term effect on normal synaptic transmission or pathway (Ye and Qiao, 1999). A similar conclusion was drawn
LTD (Raymond et al., 2003). In this study, Aß1e40 also produced by Nomura et al. (2005).
a small, but significant, inhibition (approx. 25%) of NMDA re- Similar effects were also reported for Schaffer-collateral to
ceptor-mediated synaptic potentials (Raymond et al., 2003). CA1 LTP in vivo (Freir et al., 2001). LTP was markedly
The authors claim that NMDA receptors were unlikely to be in- reduced by i.c.v. Aß25e35 (10 nmol) and completely blocked
volved in this effect because MK-801 (0.5 mM) had no effect on by Aß25e35 (100 nmol). The effects of this Aß fragment on
LTP e but this conclusion may be questioned due to the short LTP were probably mediated via a postsynaptic mechanism
incubation (20 min) used e see Frankiewicz et al. (1996). In because they did not affect paired pulse facilitation (Freir
the same concentration range effective for inhibiting LTP, et al., 2001). The negative effects of i.c.v. Aß1e42 oligomers
Aß1e42 also reduced the amplitude of NMDA receptor-medi- on hippocampal LTP in vivo were completely prevented by
ated synaptic currents in dentate granule cells in vitro via a post- co-administration of monoclonal Aß antibodies (Kim et al.,
synaptic mechanism (Chen et al., 2002). Moreover, Aß1e42 2001; Walsh et al., 2002). Aß1e42 was also reported to facili-
strongly inhibited the induction of NMDA receptor-dependent tate the induction of LTD and depotentiation of LTP in the
LTP in the dentate gyrus of hippocampal slices, but not CA1 area the rat hippocampus in vivo in an NMDA recep-
NMDA receptor-independent LTP or long-term depression tor-dependent manner (Kim et al., 2001). Effects of different
(LTD) (Wang et al., 2004a). Furthermore, in one study, pre- classes/doses of NMDA receptor antagonists in such models
treatment of rat hippocampal slices with Aß1e42 alone only have, unfortunately, not yet been addressed.
moderately inhibited LTP but co-treatment with a sub-threshold The direct involvement of NMDA receptors in mediating
concentration of glutamate (30 mM) impaired LTP more these effects of Aß is still a matter of debate. Application of
strongly, implicating an interplay between Aß and the glutama- Aß1e40 by extracellular perfusion (200 nM) or intracellularly
tergic system (Nakagami and Oda, 2002). There is also evidence via the recording pipette (100 nM) resulted in a gradual en-
that the Aß1e42-mediated inhibition of LTP induction involves hancement of NMDA receptor-mediated synaptic currents in
stimulation of the kinases JNK, Cdk5, and p38 MAPK, the granule cells in the rat dentate gyrus in vitro with no effect
cytokine tumour necrosis factor (TNFa) and metabotropic on AMPA receptor-mediated transmission, resting membrane
glutamate receptors (mGluR5) (Wang et al., 2004b, 2005). potential or input resistance (Wu et al., 1995). Similarly, con-
The most toxic species of Aß seems to be the oligomeric ditioned media from aAPPs/p3- and ßAPPs/Aß-stimulated
form. Thus, incubation of rat hippocampal slices with small cultured human monocyte-derived macrophages directly
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 711

induced inward currents via NR1a/NR2B receptors expressed similar to that seen in GLT-1 knockout mice (Katagiri et al.,
in X. oocytes that were blocked by the NMDA receptor antag- 2001). The decisive experiment would be to test the ability
onist AP5 (2-amino-5-phosphonovalerate, 50 mM) but not by of memantine to reverse such Aß-induced deficits in LTP
the AMPA receptor antagonist CNQX (6-cyano-2,3-dihy- and we are presently performing such studies.
droxy-7-nitroquinoxaline, 20 mM) (Xiong et al., 2004). In con-
trast, others have reported that although Aß25e35 induced 5. Hypothesis of memantine MOA in AD
increases in intracellular Ca2þ in cultured hippocampal neuro-
nes that was enhanced by Mg2þ removal and blocked by Several hypotheses have been proposed to explain the
NMDA receptor antagonists, this was due to enhanced gluta- MOA of memantine in AD. Most of them concentrate on ex-
matergic synaptic network activity but not due to direct effects planations as to why memantine has a better therapeutic toler-
on NMDA receptors (Brorson et al., 1995). However, more ability compared to other NMDA receptor channel blockers
recently, Ye et al. (2004) showed using primary neocortical such as (þ)MK-801 and phencyclidine rather than on why
cultures that Aß assemblies in a prefibrilar form produced neu- memantine provides a clinical improvement in cognition in
ronal activation (patch clamp) that was attenuated by NMDA this disease. These theories are discussed below and presented
receptor antagonist AP5. in a graphic form in several figures.
Although it is still not clear whether NMDA receptors are
directly or indirectly activated/modulated by Aß, enhanced 5.1. Low affinity and kinetics
NMDA receptor sensitivity might, at first sight, seem contra-
intuitive for the observed deficits in the LTP. However, we Most hypotheses are based on the widely documented and
propose similar mechanisms as discussed above for the im- accepted fact that memantine and other well-tolerated open
pairment of learning and synaptic plasticity under conditions channel blockers show much faster open channel blocking/un-
of chronic, non-phasic activation of NMDA receptors as blocking kinetics than compounds burdened with psychotropic
seen following reduction of Mg2þ concentration or application side-effects such as (þ)MK-801 or phencyclidine (Rogawski
of non-toxic concentrations of NMDA receptor agonists (see et al., 1991; Chen et al., 1992; Rogawski, 1993; Parsons
above). In line with this assumption, Aß25e35 also was et al., 1993; Black et al., 1996; Parsons et al., 1999b) (see
reported to inhibit L-glutamate uptake in rat hippocampal as- Fig. 5). Unblocking kinetics are directly related to affinity,
trocyte cultures (Harris et al., 1996) e this could also lead i.e. less potent compounds have faster unblocking kinetics
to a tonic activation of NMDA receptors and deficits in LTP whereas the on-rate of blockade does not seem to depend on

Low affinity antagonists assure fast association and dissociation


Memantine Glutamate

MK-801 Glutamate

Fig. 5. Schematic illustrating faster blocking kinetics of lower affinity channel blockers in the continuous presence of agonist and without changes in membrane
potential. As lower affinity compounds are required at higher concentrations than higher affinity compounds to provide a similar level of equilibrium block for the
whole receptor population then, by the law of mass action, they block each receptor channel with faster onset kinetics. Similar fast unblocking kinetics of lower
affinity compounds is an inherent feature, directly related to affinity. In the top sequence running left to right, the ephemeral/fast flicking block/unblock in the
continuous presence of memantine is illustrated for one receptor. In the lower sequence, lower concentrations of the more potent compound MK-801 take
much longer to gain access to each receptor channel, but once blocked, also take much longer to unblock. After achieving equilibrium blockade, in the continuous
presence of antagonist, the proportion of receptors in the blocked state at any time is, by definition, the same e here 2 of 6, i.e. 33%.
712 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

affinity, at least when expressed as Kon (Parsons et al., 1995). is important in determining their potential therapeutic safety,
However, the onset kinetics are also faster for lower affinity at least in epilepsy for which memantine has not yet been
compounds when the rate of blockade is expressed as ton at proven to be clinically useful and which, unfortunately, seems
IC50 concentrations (Parsons et al., 1995). The latter is the unlikely on the basis of both preclinical and clinical data (Ro-
therapeutically more relevant parameter and this is a fact gawski et al., 1991; Chen et al., 1992; Rogawski, 1993). How-
that should be apparent, at least to a biophysicist, due to the ever, this hypothesis alone cannot explain the clinically proven
law of mass action, i.e. the higher the concentration of com- better tolerability of memantine in AD because, even if recep-
pound needed to block a receptor, the faster the rate of this tors were only blocked following pathological activation, they
block, at least if Kon is not changed. would then remain mostly blocked in the real therapeutic sit-
As to the published data, memantine blocks NMDA chan- uation, i.e. the continuous presence of memantine in AD, and
nels activated by high concentrations of agonist at 70 mV therefore would still be unavailable for subsequent physiolog-
with a Kon of 2e4  105 M1 s1 and Koff of around 0.2 s1 ical activation.
(Parsons et al., 1993, 1995, 1996, 1998; Bresink et al., 1996; The partial untrapping hypothesis (Blanpied et al., 1997;
Chen and Lipton, 1997; Blanpied et al., 1997; Sobolevsky Johnson and Kotermanski, 2006) partially addresses this issue,
and Koshelev, 1998; Sobolevsky et al., 1998). More detailed at least for potential neuroprotective effects of memantine in,
analysis reveals that memantine blocks and unblocks open e.g. AD but cannot alone be used to explain the clinically
NMDA receptor channels with double or even triple exponen- proven symptomatic effects of cognition e see below.
tial kinetics. The amplitude and speed of the fast component of
block increase with memantine concentration. In contrast, the
speed of fast unblock remains constant but the amplitude de- 5.2. Voltage-dependency and rapid unblocking kinetics
creases with memantine concentration (Frankiewicz et al.,
1996; Bresink et al., 1996; Blanpied et al., 1997; Sobolevsky The voltage-dependency of memantine rendered by its pos-
et al., 1998; Sobolevsky and Koshelev, 1998). These data indi- itive charge on the amino group at physiological pH and its
cate that memantine binds to at least two sites within the binding at a deep channel site of NMDA channel is reflected
NMDA receptor channel. in estimated d values of 0.71e0.83, i.e. memantine seems to
There are divergent opinions as to why such blocking char- experience 70e80% of the transmembrane field (Parsons
acteristics are advantageous. Both Lipton’s and Rogawski’s et al., 1993, 1995, 1996, 1998; Bresink et al., 1996; Chen
groups assumed that the ability of low affinity open channel and Lipton, 1997; Blanpied et al., 1997; Sobolevsky and Kosh-
blockers to gain rapid access to the NMDA receptor channel elev, 1998; Sobolevsky et al., 1998).

High Agonist Concentration Low Agonist Concentration

Equilibrium block is the same

Fig. 6. Schematic illustrating agonist concentration-dependence of channel blocking kinetics for single channel. At high concentrations of agonist, more receptor
channels are open and available for blockade at any one time e left, all three of the three receptors; right, one of the three receptors. The sequence of activation
followed by block is the same for both, i.e. from top to bottom as indicated by the blue arrows. Although the time taken to achieve equilibrium block is different,
the level of this block at the end is the same e in this case three of the three receptors in each case.
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 713

In addition, when offset kinetics in the continuous presence Memantine Low NMDA
of memantine following depolarization are fit by double expo-
nentials, the predominant effect of depolarization is to increase
dramatically the weight of the faster recovery time-constant of
memantine (Frankiewicz et al., 1996; Bresink et al., 1996; Par- Memantine Moderate NMDA
sons et al., 1998) which provides further support for there be-
ing at least two channel blocking sites for memantine e see
Section 5.1.
Although this d value is very similar to that of Mg2þ, the
functional consequences are different because memantine
bears only one positive charge whereas Mg2þ is a divalent
cation and therefore reacts with even stronger functional
Memantine High NMDA
dependence on changes in membrane potential. As such, in
comparison to Mg2þ, memantine shows somewhat less pro-
nounced functional voltage-dependency, but is still a strongly
voltage-dependent channel blocker when compared to high af-
finity uncompetitive NMDA receptor antagonists.
The kinetics of (þ)MK-801 and phencyclidine are too slow
to allow them to leave the channel upon transient depolariza-
tion plus concomitant NMDA receptor activation and this is
reflected in apparently weaker functional voltage-dependency
reported in almost all studies published to date.
In other words, under physiologically relevant conditions, Memantine Normalized NMDA
the two parameters kinetics and voltage-dependency are
Non-steady state
directly related to antagonist affinity, with lower affinity com- unblock different
pounds showing faster kinetics and apparently stronger func-
tional voltage-dependency (Parsons et al., 1995, 1999a) than
high affinity compounds. These aspects provide the basis for
signal-to-noise hypothesis (Parsons et al., 1999a; Danysz
et al., 2000; Danysz and Parsons, 2003) which is discussed
Non-steady state
in detail below. block different

5.3. Agonist concentration-dependency (use dependency) Fig. 7. Schematic illustrating agonist concentration-dependence of channel
blocking kinetics for whole cell patch clamp currents in concentration clamp
experiments. At high concentrations of agonist, more receptor channels are
NMDA receptor agonist concentration-dependency of
open and available for blockade at agonist equilibrium and this is reflected
block by memantine has been discussed intensively by some as larger whole cell currents e red low, green moderate and black high con-
groups in the literature. The hypothesis is that the higher the centrations of agonist (e.g. NMDA). As a direct consequence, concentration
NMDA receptor agonist concentration, the greater the block clamp application/removal of antagonist blocks/unblocks these whole cell cur-
by memantine. This has been claimed to have profound ther- rents with faster kinetics for higher concentrations of agonist. Although this is
reflected in different levels of non-steady state block/unblock by the arrows in
apeutic relevance and is discussed as one of the main reasons
the normalized superimposed traces at the bottom, the degree of block at an-
why memantine is different from other NMDA receptor chan- tagonist equilibrium is the same. When assessing the degree of blockade by
nel blockers (Lipton and Chen, 2004; Lipton, 2004, 2005, lower, therapeutically relevant concentrations of memantine then, by the law
2006). The fundament of this hypothesis to explain the better of mass action, the onset kinetics of blockade would also become much slower.
therapeutic utility of memantine is in disaccord with the mM Finally, it should be clear from the agonist onset kinetics in the normalized fig-
ure, that lower concentrations of agonist take longer to reach steady state re-
concentrations of glutamate known to be released during phys-
ceptor activation. If low concentrations of agonist have not reached equilibrium
iological activation of NMDA receptors (Clements et al., levels of activation at the time of antagonist addition then a continued, unobserved
1992) (see later sections). background increase of ‘‘control’’ levels of low concentration agonist activation
In retrospect, any apparent agonist concentration dependent might apparently counteract any observed block by uncompetitive antagonist. In
blocking effects of memantine reported in early experiments other words, there are three very large technical hurdles that require that such ex-
periments have to be performed with extreme care and that a very long time must
seem to have been observed purely due to technical limita-
be allowed for the both agonist and antagonist to reach true equilibrium activation/
tions. When one works with low concentrations of agonist, blockade under such conditions. One needs very good, cultured, neuronal cells
the open probability of NMDA receptor channels is reduced without significant run down to be able to address this issue.
and an open channel blocker therefore needs more time to
gain access to the channel (Figs. 6 and 7). In other words, Parsons et al. (1993). It should also be noted that kinetics are
an equilibrium block had almost certainly not been achieved much slower at room temperature used in most in vitro exper-
under the experimental conditions used with low agonist con- iments than in vivo, i.e. 37  C (Davies et al., 1988). One needs
centrations at room temperature in both Chen et al. (1992) and very good, primary, neuronal cells without significant run
714 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

down to be able to address this issue. On another note Chen Other important differences between ketamine and meman-
et al. (1992) used retinal ganglion cells, which are not really tine are more likely to be responsible for the differences
so relevant for the CNS, and never described or subsequently observed in the clinical tolerability of these two fast, volt-
discussed the possible technical problems discussed above, age-dependent open channel NMDA receptor antagonists e
nor the additional potential problems of receptor run down. namely PK and route of administration. In strong contrast to
Our recent experiments (Gilling et al., 2007) attempt to take memantine, ketamine has an extremely short half-life in
these problems into account, and indicate, that there is NO true both rodents and man and was normally administered i.v. in
agonist concentration-dependency of equilibrium NMDA re- most clinical studies addressing, e.g. effects on chronic pain.
ceptor channel blockade by memantine (Fig. 7). Any further dis- It is therefore probably, that excessive peak plasma/CSF levels
cussion on the possible relevance of such an effect seems to be were needed in such studies and that this was the cause for
beyond the scope of this review, because this previously pub- poor tolerability.
lished and very much discussed ‘‘observation’’ was most likely
an experimental artefact (Gilling et al., 2007).
5.5. NMDA receptor subtype selectivity
5.4. Partial untrapping
Memantine blocked NR2A, NR2B, NR2C and NR2D re-
Partial untrapping has been claimed to explain the better ceptors expressed in X. oocytes with IC50s of 0.89, 0.40,
therapeutic utility of memantine as a proportion of channels e 0.32 and 0.28 mM, respectively (Parsons et al., 1999a).
around 15e20% e would always unblock upon cessation of NR2B selective antagonists also seem to have a better thera-
agonist activation and would thereby be available for subse- peutic profile in many animal models (Parsons et al., 1998).
quent physiological activation (Blanpied et al., 1997) There are several hypotheses why this could be the case that
(Fig. 8). The phenomenon also occurs in the continuous pres- are outside the scope of the present review but the most prev-
ence of memantine, which is the case in the therapeutic use of alent are related to the selective expression of NR2B in fore-
memantine (Parsons et al., 1999b). The newest data from brain regions (Monyer et al., 1994); a use- and agonist
Johnson’s group are consistent with a model in which there concentration state-dependent block by NR2B selective antag-
are two binding sites for memantine (see above), a deep site onists (Kew et al., 1998) and their extrasynaptic localisation
inside the external gate that allows full trapping, and a shallow (Li et al., 1998). Memantine and other well-tolerated NMDA
site outside the gate that does not (Kotermanski and Johnson, receptor antagonists such as dextromethorphan and dextrorphan
2004). Ketamine only binds to the deep site and this might ac- were considerably more potent against [14C]acetylcholine than
count for the fact that ketamine and (þ)MK-801 do not share [3H]spermidine release from rat striatal slices in vitro whereas
this property with memantine (Lanthorn et al., 2000). How- (þ)MK-801 and phencyclidine were equipotent e for review
ever, attractive this hypothesis might be for the tolerability see Nankai et al. (1998). This similarity was also proposed to
of memantine, such a MOA alone cannot explain the symp- be due to NR2B selective effects of memantine, an assumption
tomatic therapeutic effects of memantine on learning and syn- partially supported by the finding that memantine was three
aptic plasticity observed in animal models and proven in the times more potent against NMDA-induced Ca2þ influx in hu-
clinical situation. man NR1a/NR2B receptors than in human NR1a/NR2A

MK-801 Glutamate Complete trapping

Memantine Glutamate Partial untrapping

Fig. 8. Very simplified schematic illustrating partial untrapping by memantine. Top, MK-801 takes longer to block open NMDA receptor channels due to slower
onset kinetics at the lower concentrations needed. Once inside the channel, block is maintained for a long time and agonist can dissociate and trap MK-801 in the
closed, inactive channel. Bottom, memantine gains quicker access to the open NMDA receptor channel but a proportion (around 20%) of the receptor channels
release memantine before closure and release of agonist. See Fig. 5 for a more temporal schematic of differences between memantine and MK-801.
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 715

receptors permanently expressed in L(tk-) cells (Grimwood Mg2þ is so pronounced that it also leaves the NMDA channel
et al., 1996). upon moderate depolarization under pathological conditions.
However, the relevance of this moderate two- to three-fold Although uncompetitive antagonists also block the NMDA re-
greater potency at NR2B over NR2A receptors is unclear and ceptor channel, high affinity compounds such as (þ)MK-801
is not supported by other studies. Thus, memantine was equi- have much slower unblocking kinetics than Mg2þ and appar-
potent against glutamate-induced currents in NR2A and NR2B ently far less pronounced functional voltage-dependency (see
receptors expressed in HEK-293 cells (IC50 0.93 and 0.82 mM, above) and are therefore unable to leave the channel within
respectively, at 70 mV) but again somewhat more potent the time course of a normal NMDA receptor-mediated excit-
against NR2D receptors (IC50 ¼ 0.47 mM) (Bresink et al., atory postsynaptic potential. As a result, (þ)MK-801 blocks
1996). Memantine was also previously reported to be almost both the pathological and physiological activation of NMDA
equipotent against NR2A and NR2B receptors expressed in receptors.
X. oocytes (IC50 of 0.29 and 0.23 mM, respectively) although We were the first to suggest that the combination of fast off-
the absolute potencies reported in this study (Avenet et al., set kinetics and relatively strong functional voltage-depen-
1997) where considerably higher than those reported by sev- dency allow memantine to rapidly leave the NMDA channel
eral different groups. Even if memantine were to show upon transient physiological activation by mM concentrations
a greater potency at NR2B receptors than NR2A, there is no of synaptic glutamate but block the sustained activation by mM
published reason that would support the notion that meman- concentrations of glutamate under moderate pathological
tine shares the same reported mechanisms for state-dependent conditions (Parsons et al., 1993, 1995, 1996) (Fig. 9). This
effects as classical NR2B antagonists (Kew et al., 1998).
Binding studies indicate that the better therapeutic profile Pathology Physiology
Rest
of memantine and other drugs known to be well tolerated in
humans may be related to relatively higher affinity in the cer- Mg2+ Ca2+ Ca2+
ebellum than in forebrain regions (Bresink et al., 1995; Porter
and Greenamyre, 1995). This increased potency in the
cerebellum could be related to the enhanced expression of
NR2C receptors in this region (Monyer et al., 1994) and
memantine has indeed been reported to be three-fold more po-
tent against NR2C receptors than NR2A receptors (Parsons - 70 mV - 50 mV - 20 mV
et al., 1999a). In this regard, a clinical study showing that
memantine can block classical eyeblink conditioning at doses Ca2+
Memantine
having no effect on verbal or visuospatial memory is of par-
ticular interest (Schugens et al., 1997) as classical eyeblink
conditioning is critically dependent upon intact cerebral cir-
cuitry. It is not clear why memantine should block synaptic
plasticity mediated via NR2C but not NR2A/B receptors but
it is likely that the three-fold greater potency at these recep-
- 70 mV - 50 mV - 20 mV
tors is associated with slower blocking kinetics and therefore
less ability to leave these receptor channels upon transient
MK-801
physiological activation. However, it is again unclear how
such a moderate preference of memantine for NR2C/2D re-
ceptors can explain its improved therapeutic profile in AD,
in particular, improvement of cognition. In fact, NR2C block-
ade might rather be expected to cause more pronounced cer-
ebellar motor deficits.
- 70 mV - 50 mV - 20 mV

5.6. Signal-to-noise hypothesis Fig. 9. Schematic illustrating the hypothesis explaining how the fast unblock-
ing kinetics of memantine allow this voltage-dependent compound to differen-
Physiologically, NMDA receptors are transiently activated tiate between the physiological and pathological activation of NMDA
by mM concentrations of glutamate in the synapse (Clements receptors. Under resting therapeutic conditions, i.e. in their continuing pres-
ence at 70 mV (left), Mg2þ (top), memantine (middle) and MK-801 (bottom)
et al., 1992) following strong depolarization of the postsynap- all occupy the NMDA receptor channel. Both Mg2þ and memantine are able to
tic membrane which rapidly relieves their voltage-dependent leave the NMDA receptor channel upon strong synaptic depolarization
blockade by Mg2þ (Nowak et al., 1984) whereas during path- (20 mV, right) due to their pronounced voltage-dependency and rapid un-
ological activation, NMDA receptors are activated by lower blocking kinetics whereas the slow blocker MK-801 remains trapped. How-
concentrations of glutamate but for much longer periods of ever, memantine e in contrast to Mg2þ e does not leave the channel so
easily upon moderate prolonged depolarization during chronic excitotoxic in-
time (Benveniste et al., 1984; Andine et al., 1991; Globus sults (50 mV, centre). Transient strong and prolonged moderate Ca2þ influx
et al., 1991a,b; Mitani et al., 1992; Buisson et al., 1992). How- illustrated by the full and dashed red arrows, respectively. Modified after Korn-
ever, the functional voltage-dependency of the divalent cation huber and Weller (1997) and Parsons et al. (1999b).
716 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

1 2

noise noise

NMDA receptor NMDA receptor


magnesium magnesium
glutamate glutamate
-70mV -70mV

3 4
signal
noise noise

NMDA receptor NMDA receptor


magnesium magnesium
glutamate glutamate

-20mV -70mV

Fig. 10. Under normal conditions, synaptic plasticity/learning depends on the detection of a relevant (sufficiently strong) signal over background activity (here
referring to transient strong vs. prolonged moderate intracellular Ca2þ levels), i.e. sufficient signal-to-noise ratio. Intracellular Ca2þ levels at any one time-point
represented by different sizes of the Ca2þ containing circles. Electrodes/potentiometers illustrate the postsynaptic membrane potential: 70 mV, resting; 50 mV,
mild prolonged pathology; 20 mV, transient strong synaptic activation. Jagged red arrow indicates arrival of a presynaptic signal. Mg2þ and glutamate are illus-
trated by blue circles and red triangles, respectively.

hypothesis is further supported by the fact that, although the expressed in X. oocytes following voltage-steps is very rapid
predominant component of offset kinetics upon rapid removal and well within the time course of an NMDA receptor-mediated
of memantine at room temperature in vitro at near resting EPSP (Parsons et al., submitted for publication). This finding
membrane potentials is still too slow to allow synaptic activa- adds further support to our hypothesis that rapid, voltage-depen-
tion e i.e. around 5 s, the relief of blockade in the continuous dent relief of blockade following synaptic activation of NMDA
presence of memantine upon depolarization is much faster due receptors underlies the clinical utility of memantine.
to an increase in the weight of the faster recovery time- These kinetics are likely to be even faster in vivo due to
constant (see above). The unblocking rate of memantine from higher temperatures (Davies et al., 1988). Furthermore, the
cultured hippocampal neurones and NR1a/NR2A receptors rate of recovery from memantine blockade is dependent on

Aß, Tau, Energy deficit Aß, Tau, Energy deficit

1 2 noise 3 noise

noise

NMDA receptor NMDA receptor NMDA receptor


magnesium magnesium magnesium
glutamate glutamate glutamate
-70 mV -50 mV -50mV

Aß , Tau, Energy deficit Aß, Tau, Energy deficit

4 signal noise 5

NMDA receptor NMDA receptor


magnesium magnesium
glutamate glutamate
-20mV 0 mV

Fig. 11. The signal-to-noise ratio hypothesis assumes that in AD, due to a tonically overactive glutamatergic system, Mg2þ is not effective enough to play its
‘filtering’ function. In turn, synaptic noise rises, impairing detection of the relevant signal such as in learning. One bound glutamate to NMDA receptor represents
continuous, low-level NMDA receptor activation (even though two molecules in fact have to be bound to activate the receptor). Potentiometer at 0 mM indicates
cell death. Aß, Tau and energy deficit are shown as factors favouring, either directly or indirectly, conditions for overactivation of NMDA receptors. Other aspects
are as in Fig. 10.
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 717

Aß, Tau, Energy deficit Aß, Tau, Energy deficit

1 noise 2

M noise

NMDA receptor NMDA receptor


magnesium magnesium
glutamate glutamate
M Memantine -50 mV -70 mV

Aß, Tau, Energy deficit Aß, Tau, Energy deficit

3 4
signal
M
M noise noise

NMDA receptor NMDA receptor


magnesium magnesium
glutamate glutamate
-70 mV -20 mV

Fig. 12. Schematic explaining the MOA of memantine in AD based on the signal-to-noise hypothesis. Memantine is able to serve as a filter blocking ‘synaptic
noise’ and thereby allowing detection of the relevant signal, i.e. synaptic plasticity is restored. Memantine illustrated as a large, ‘‘M&M like’’ blue circle; for other
aspects, see legends to Figs. 10 and 11.

the open probability of NMDA channels (Chen and Lipton, Under resting conditions, and in their continuing presence,
1997), i.e. would be faster in the presence of higher, synaptic both Mg2þ and memantine occupy the NMDA receptor
concentrations of glutamate (Clements et al., 1992). channel. Likewise, both are able to leave the NMDA receptor
In summary, we feel that the biophysical properties of channel upon strong synaptic depolarization due to their pro-
memantine would allow this compound to clearly differentiate nounced voltage-dependency and rapid unblocking kinetics.
between chronic low-level pathological activation of NMDA However, memantine e in contrasts to Mg2þ e does not leave
receptors in AD (‘‘noise’’) and physiological synaptic the channel so easily upon moderately prolonged depolariza-
NMDA receptor activation (‘‘signal’’) (Danysz et al., 2000; tion during chronic excitotoxic insults (Parsons et al., 1993,
Danysz and Parsons, 2003). It is our belief that memantine, 1995). This property of memantine could indeed provide
at therapeutically relevant concentrations, is able to suppress both neuroprotection and symptomatic improvement by the
the pathological background ‘‘noise’’ but preserve or even same MOA, i.e. fast, moderate affinity, voltage-dependent
enhance the desired physiological synaptic NMDA receptor- NMDA receptor channel blockade.
mediated plasticity ‘‘signal’’. In our opinion, memantine pro-
vides both neuroprotection and symptomatic improvement by
the same MOA, i.e. moderate-affinity NMDA receptor channel Acknowledgements
blockade (Figs. 10, 11 and 12).
The authors would like to thank Andrzej Dekundy for the
6. Conclusions compilation of information of magnesium effects in man;
Claudia Jatzke, Kate Gilling (Merz Pharmaceuticals), Jan
Egebjerg, and Julie Lotharius (Lundbeck) for valuable com-
As discussed above, various hypotheses attempt to explain
ments to the manuscript.
why memantine, in contrast to some other NMDA receptor an-
tagonists, may provide both the clinically proven symptomatic
improvement in AD plus the potential for neuroprotective ac-
References
tivity with few side-effects. However, in our opinion, only the
signal-to-noise hypothesis e which considers both voltage- Abraham, W.C., Mason, S.E., 1988. Effects of NMDA receptor/channel antag-
dependence and fast offset kinetics in the continuous presence onists CPP and MK-801 on hippocampal field potentials and long-term
of memantine e can adequately explain both the rapid symp- potentiation in anasthetized rats. Brain Res. 462, 40e46.
tomatological, cognitive effects of memantine seen already Albin, R.L., Greenamyre, J.T., 1992. Alternative excitotoxic hypotheses.
after a few weeks of treatment in clinical trials e and the pre- Neurology 42, 733e738.
Alessandri-Haber, N., Dina, O.A., Joseph, E.K., Reichling, D., Levine, J.D.,
dicted long-term neuroprotective effects of this compound. 2006. A transient receptor potential vanilloid 4-dependent mechanism of
To sum up our working hypothesis, the effects of meman- hyperalgesia is engaged by concerted action of inflammatory mediators.
tine can be likened to a slightly more potent Mg2þ block. J. Neurosci. 26, 3864e3874.
718 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Andine, P., Sandberg, M., Bagenholm, R., Lehmann, A., Hagberg, H., 1991. Bormann, J., 1989. Memantine is a potent blocker of N-methyl-D-aspartate
Intra- and extracellular changes of amino acids in the cerebral cortex of (NMDA) receptor channels. Eur. J. Pharmacol. 166, 591e592.
the neonatal rat during hypoxic-ischemia. Dev. Brain Res. 64, 115e120. Bowen, D.M., Francis, P.T., Sims, N.R., 1992. Beta-amyloid increased
Andrasi, E., Igaz, S., Molnar, Z., Mako, S., 2000. Disturbances of magnesium neuronal susceptibility to injury by glucose deprivation. Neuroreport 3,
concentrations in various brain areas in Alzheimer’s disease. Magnes. Res. 733.
13, 189e196. Bresink, I., Benke, T.A., Collett, V.J., Seal, A.J., Parsons, C.G., Henley, J.M.,
Aracava, Y., Pereira, E.F., Maelicke, A., Albuquerque, E.X., 2004. Meman- Collingridge, G.L., 1996. Effects of memantine on recombinant rat NMDA
tine at clinically relevant concentrations blocks more potently a7 nicotinic receptors expressed in HEK-293 cells. Br. J. Pharmacol. 119, 195e204.
than NMDA receptors: implications for treatment of Alzheimer’s disease, Bresink, I., Danysz, W., Parsons, C.G., Mutschler, E., 1995. Different binding
P1e416. affinities of NMDA receptor channel blockers in various brain regions e
Aracava, Y., Pereira, E.F., Maelicke, A., Albuquerque, E.X., 2005. Memantine indication of NMDA receptor heterogeneity. Neuropharmacology 34,
blocks alpha7* nicotinic acetylcholine receptors more potently than N- 533e540.
methyl-D-aspartate receptors in rat hippocampal neurons. J. Pharmacol. Brewer, G.J., Wallimann, T.W., 2000. Protective effect of the energy precursor
Exp. Ther. 312, 1195e1205. creatine against toxicity of glutamate and beta-amyloid in rat hippocampal
Avenet, P., Leonardon, J., Besnard, F., Graham, D., Depoortere, H., neurons. J. Neurochem. 74, 1968e1978.
Scatton, B., 1997. Antagonist properties of eliprodil and other NMDA re- Brorson, J.R., Bindokas, V.P., Iwama, T., Marcuccilli, C.J., Chisholm, J.C.,
ceptor antagonists at rat NR1a/NR2A and NR1a/NR2B receptors Miller, R.J., 1995. The ca2þ influx induced by beta-amyloid peptide
expressed in Xenopus oocytes. Neurosci. Lett. 223, 133e136. 25e35 in cultured hippocampal neurons results from network excitation.
Bachurin, S., Tkachenko, S., Baskin, I., Lermontova, N., Mukhina, T., J. Neurobiol. 26, 325e338.
Petrova, L., Ustinov, A., Proshin, A., Grigoriev, V., Lukoyanov, N., Buisson, A., Callebert, J., Mathieu, E., Plotkine, M., Boulu, R.G., 1992. Stria-
Palyulin, V., Zefirov, N., 2001. Neuroprotective and cognition-enhancing tal protection induced by lesioning the substantia nigra of rats subjected to
properties of MK-801 flexible analogs. Structureeactivity relationships. focal ischemia. J. Neurochem. 59, 1153e1157.
Ann. N. Y. Acad. Sci. 939, 219e236. Buisson, B., Bertrand, D., 1998. Open-channel blockers at the human alpha 4
Bakchine, S., Pascual-Gangnant L., Loft, H., 2005. Results of a randomised, beta 2 neuronal nicotinic acetylcholine receptor. Mol. Pharmacol. 53,
placebo-controlled 6-month study of memantine in the treatment of mild 555e563.
to moderate Alzheimer’s disease in Europe. ADIS title: memantine: ther- Bullock, R., 2006. Efficacy and safety of memantine in moderate-to-severe
apeutic use, Alzheimer’s disease in patients with mild to moderate illness Alzheimer disease: the evidence to date. Alzheimer Dis. Assoc. Disord.
2087. 20, 23e29.
Barber, T.A., McGettigan, B.F., Meyers, R.A., 2002. The effects of memantine Burns, A., O’Brien, J., Auriacombe, S., Ballard, C., Broich, K., Bullock, R.,
on memory formation for passive avoidance learning in day e old chicks. Feldman, H., Ford, G., Knapp, M., McCaddon, A., Iliffe, S., Jacova, C.,
Soc. Neurosci. Abst. 28, 684.6. Jones, R., Lennon, S., McKeith, I., Orgogozo, J.M., Purandare, N.,
Bardgett, M.E., Schultheis, P.J., McGill, D.L., Richmond, R.E., Wagge, J.R., Richardson, M., Ritchie, C., Thomas, A., Warner, J., Wilcock, G.,
2005. Magnesium deficiency impairs fear conditioning in mice. Brain Wilkinson, D., 2006. Clinical practice with anti-dementia drugs: a consen-
Res. 1038, 100e106. sus statement from British Association for Psychopharmacology. J. Psy-
Barghorn, S., Nimmrich, V., Striebinger, A., Krantz, C., Keller, P., Janson, B., chopharmacol. 20, 732e755.
Bahr, M., Schmidt, M., Bitner, R.S., Harlan, J., Barlow, E., Ebert, U., Carvalho, F.M., Pereira, S.R., Pires, R.G., Ferraz, V.P., Romano-Silva, M.A.,
Hillen, H., 2005. Globular amyloid beta-peptide oligomer - a homogenous Oliveira-Silva, I.F., Ribeiro, A.M., 2006. Thiamine deficiency decreases
and stable neuropathological protein in Alzheimer’s disease. J. Neuro- glutamate uptake in the prefrontal cortex and impairs spatial memory per-
chem. 95, 834e847. formance in a water maze test. Pharmacol. Biochem. Behav. 83, 481e489.
Barnes, C.A., Danysz, W., Parsons, C.G., 1996. Effects of the uncompetitive Chen, H.S., Lipton, S.A., 1997. Mechanism of memantine block of NMDA-ac-
NMDA receptor antagonist memantine on hippocampal long-term potenti- tivated channels in rat retinal ganglion cells: uncompetitive antagonism.
ation, short-term exploratory modulation and spatial memory in awake, J. Physiol. (Lond.) 499 (pt 1), 27e46.
freely moving rats. Eur. J. Neurosci. 8, 565e571. Chen, H.S.V., Pellegrini, J.W., Aggarwal, S.K., Lei, S.Z., Warach, S.,
Beal, M.F., 1995. Aging, energy, and oxidative stress in neurodegenerative Jensen, F.E., Lipton, S.A., 1992. Open-channel block of N-methyl-D-aspar-
diseases. Ann. Neurol 38, 357e366. tate (NMDA) responses by memantine e therapeutic advantage against
Beal, M.F., 2000. Energetics in the pathogenesis of neurodegenerative NMDA receptor-mediated neurotoxicity. J. Neurosci. 12, 4427e4436.
diseases. Trends Neurosci. 23, 298e304. Chen, H.S.V., Wang, Y.F., Rayudu, P.V., Edgecomb, P., Neill, J.C.,
Benveniste, H., Drejer, J., Schusboe, A., Diemer, N.H., 1984. Elevation of the Segal, M.M., Lipton, S.A., Jensen, F.E., 1998. Neuroprotective concentra-
extracellular concentrations of glutamate and aspartate in rat hippocampus tions of the N-methyl-D-aspartate open-channel blocker memantine are
during transient cerebral ischemia monitored by intracerebral microdialy- effective without cytoplasmic vacuolation following post-ischemic admin-
sis. J. Neurochem. 43, 1369e1374. istration and do not block maze learning or long-term potentiation.
Birks, J., 2006. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Neuroscience 86, 1121e1132.
Database Syst. Rev. CD005593. Chen, Q.S., Wei, W.Z., Shimahara, T., Xie, C.W., 2002. Alzheimer amyloid
Black, M., Lanthorn, T., Small, D., Mealing, G., Lam, V., Morley, P., 1996. beta-peptide inhibits the late phase of long-term potentiation through
Study of potency, kinetics of block and toxicity of NMDA receptor antag- calcineurin-dependent mechanisms in the hippocampal dentate gyrus.
onists using fura-2. Eur. J. Pharmacol. 317, 377e381. Neurobiol. Learn. Mem. 77, 354e371.
Blanchard, B.J., Park, T., Fripp, W.J., Lerman, L.S., Ingram, V.M., 1993. A mi- Choi, D.W., 1992. Excitotoxic cell death. J. Neurobiol. 23, 1261e1276.
tochondrial DNA deletion in normally aging and in Alzheimer brain tissue. Clark, B.A., Brown, R.S., 1992. Unsuspected morbid hypermagnesemia in
Neuroreport 4, 799e802. elderly patients. Am. J. Nephrol. 12, 336e343.
Blanchard, B.J., Thomas, V.L., Ingram, V.M., 2002. Mechanism of membrane Clements, J.D., Lester, R.A.J., Tong, G., Jahr, C.E., Westbrook, G.L., 1992.
depolarization caused by the Alzheimer Abeta1e42 peptide. Biochem. The time course of glutamate in the synaptic cleft. Science 258, 1498e
Biophys. Res. Commun. 293, 1197e1203. 1501.
Blanpied, T.A., Boeckman, F.A., Aizenman, E., Johnson, J.W., 1997. Trapping Coan, E.J., Collingridge, G.L., 1987. Characterization of an N-methyl-D-aspar-
channel block of NMDA-activated responses by amantadine and meman- tate receptor component of synaptic transmission in rat hippocampal slice.
tine. J. Neurophysiol. 77, 309e323. Neuroscience 22, 1e8.
Blass, J.P., Sheu, R.K., Gibson, G.E., 2000. Inherent abnormalities in energy Coan, E.J., Irving, A.J., Collingridge, G.L., 1989. Low-frequence activation of
metabolism in Alzheimer disease. Interaction with cerebrovascular com- the NMDA receptor system can prevent the induction of LTP. Neurosci.
promise. Ann. N. Y. Acad. Sci. 903, 204e221. Lett. 105, 205e210.
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 719

Cohn, J., Coryslechta, D.A., 1994. Lead exposure potentiates the effects of van Dyck, C.H., Schmitt, F.A., Olin, J.T., 2006. A responder analysis of mem-
NMDA on repeated learning. Neurotoxicol. Teratol. 16, 455e465. antine treatment in patients with Alzheimer disease maintained on donepe-
Collingridge, G.L., Bliss, T.V.P., 1995. Memories of NMDA receptors and zil. Am. J. Geriatr. Psychiatry 14, 428e437.
LTP. Trends Neurosci. 18, 54e56. Elgoyhen, A.B., Vetter, D.E., Katz, E., Rothlin, C.V., Heinemann, S.F.,
Copani, A., Koh, J.Y., Cotman, C.W., 1991. B-amyloid increases neuronal sus- Boulter, J., 2001. Alpha 10: a determinant of nicotinic cholinergic receptor
ceptibility to injury by glucose deprivation. Neuroreport 2, 763e765. function in mammalian vestibular and cochlear mechanosensory hair cells.
Costello, D.A., Herron, C.E., 2004. The role of c-Jun N-terminal kinase in the Proc. Natl. Acad. Sci. U.S.A. 98, 3501e3506.
A beta-mediated impairment of LTP and regulation of synaptic transmis- Emerit, J., Edeas, M., Bricaire, F., 2004. Neurodegenerative diseases and
sion in the hippocampus. Neuropharmacology 46, 655e662. oxidative stress. Biomed. Pharmacother. 58, 39e46.
Crawford, J.G., 1996. Alzheimer’s disease risk factors as related to cerebral Enz, A., Gentsch, C., 2004. Co-administration of memantine has no effect on
blood flow. Med. Hypotheses 46, 367e377. the in vitro or ex vivo determined acetylcholinesterase inhibition of riva-
Creeley, C., Wozniak, D.F., Labruyere, J., Taylor, G.T., Olney, J.W., 2006. stigmine in the rat brain. Neuropharmacology 47, 408e413.
Low doses of memantine disrupt memory in adult rats. J. Neurosci. 26, Ferris, S.H., 2003. Evaluation of memantine for the treatment of Alzheimer’s
3923e3932. disease. Expert Opin. Pharmacother. 4, 2305e2313.
Cummings, J.L., Schneider, E., Tariot, P.N., Graham, S.M., 2006. Behavioral Floden, A.M., Li, S., Combs, C.K., 2005. Beta-amyloid-stimulated microglia
effects of memantine in Alzheimer disease patients receiving donepezil induce neuron death via synergistic stimulation of tumor necrosis factor al-
treatment. Neurology 67, 57e63. pha and NMDA receptors. J. Neurosci. 25, 2566e2575.
Dantoine, T., Auriacombe, S., Sarazin, M., Becker, H., Pere, J.J., Bourdeix, I., Franceschi, M., Alberoni, M., Bressi, S., Canal, N., Comi, G., Fazio, F.,
2006. Rivastigmine monotherapy and combination therapy with meman- Grassi, F., Perani, D., Volonte, M.A., 1995. Correlations between cogni-
tine in patients with moderately severe Alzheimer’s disease who failed tive impairment, middle cerebral artery flow velocity and cortical glu-
to benefit from previous cholinesterase inhibitor treatment. Int. J. Clin. cose metabolism in the early phase of Alzheimer’s disease. Dementia
Pract. 60, 110e118. 6, 32e38.
Danysz, W., Essmann, U., Bresink, I., Wilke, R., 1994. Glutamate antagonists Frankiewicz, T., Parsons, C.G., 1999. Memantine restores long term potentia-
have different effects on spontaneous locomotor activity in rats. Pharma- tion impaired by tonic N-methyl-D-aspartate (NMDA) receptor activation
col. Biochem. Behav. 48, 111e118. following reduction of Mg2þ in hippocampal slices. Neuropharmacology
Danysz, W., Parsons, C.G., 2003. The NMDA receptor antagonist memantine 38, 1253e1259.
as a symptomatological and neuroprotective treatment for Alzheimer’s dis- Frankiewicz, T., Potier, B., Bashir, Z.I., Collingridge, G.L., Parsons, C.G.,
ease preclinical evidence. Int. J. Geriatr. Psychiatry 18, S23eS32. 1996. Effects of memantine and MK-801 on NMDA-induced currents in
Danysz, W., Parsons, C.G., Kornhuber, J., Schmidt, W.J., Quack, G., 1997. cultured neurones and on synaptic transmission and LTP in area CA1 of
Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian rat hippocampal slices. Br. J. Pharmacol. 117, 689e697.
agents e preclinical studies. Neurosci. Biobehav. Rev. 21, 455e468. Freir, D.B., Holscher, C., Herron, C.E., 2001. Blockade of long-term potenti-
Danysz, W., Parsons, C.G., Möbius, H.J., Stöffler, A., Quack, G., 2000. Neuro- ation by beta-amyloid peptides in the CA1 region of the rat hippocampus
protective and symptomatological action of memantine relevant for in vivo. J. Neurophysiol. 85, 708e713.
Alzheimer’s disease e an unified glutamatergic hypothesis on the mecha- Fung, M.C., Weintraub, M., Bowen, D.L., 1995. Hypermagnesemia. Elderly
nism of action. Neurotox. Res. 2, 85e97. over-the-counter drug users at risk. Arch. Fam. Med. 4, 718e723.
Danysz, W., Zajaczkowski, W., Parsons, C.G., 1995. Modulation of learning Furukawa, K., Mattson, M.P., 1998. Secreted amyloid precursor protein alpha
processes by ionotropic glutamate receptor ligands. Behav. Pharmacol. 6, selectively suppresses N-methyl-D-aspartate currents in hippocampal neu-
455e474. rons: involvement of cyclic GMP. Neuroscience 83, 429e438.
Davies, S.N., Martin, D., Millar, J.D., Aram, J.A., Church, J., Lodge, D., 1988. Gauthier, S., Wirth, Y., Mobius, H.J., 2005. Effects of memantine on behaviou-
Differences in results from in vivo and in vitro studies on the use-depen- ral symptoms in Alzheimer’s disease patients: an analysis of the Neuropsy-
dency of N-methyl-aspartate antagonism by MK-801 and other phencycli- chiatric Inventory (NPI) data of two randomised, controlled studies. Int. J.
dine receptor ligands. Eur. J. Pharmacol. 145, 141e152. Geriatr. Psychiatry 20, 459e464.
De Felice, F.G., Velasco, P.T., Lambert, M.P., Viola, K.L., Fernandez, S.J., Gerzon, K., Krumkalns, E.V., Brindle, R.L., Marshall, F.J., Root, M.A., 1963.
Ferreira, S.T., Klein, W.L., 2007a. A beta oligomers induce neuronal oxi- The adamantyl group in medicinal agents. I. Hypoglycemic N-arylsul-
dative stress through an NMDA receptor-dependent mechanism that is fonyl-N0 -adamantylureas. J. Med. Chem. 6, 760e763.
blocked by the Alzheimer’s drug memantine. J. Biol. Chem. 282 (15), Gilling, K.E., Jatzke, C., Parsons, C.G., 2007. Agonist concentration-depen-
11590e11601. dency of blocking kinetics but not equilibrium block of N-methyl-D-aspartate
De Felice, F.G., Wu, D., Lambert, M.P., Fernandez, S.J., Velasco, P.T., Lacor, receptors by memantine. Neuropharmacology 53, 415e420.
P.N., Bigio, E.H., Jerecic, J., Acton, P.J., Shughrue, P.J., Chen-Dodson, Glick, J.L., 1990. Dementias: the role of magnesium deficiency and an hypoth-
E., Kinney, G.G., Klein, W.L., 2007b. Alzheimer’s disease-type neuronal esis concerning the pathogenesis of Alzheimer’s disease. Med. Hypotheses
Tau hyperphosphorylation induced by Abeta oligomers. Neurobiol. 31, 211e225.
Aging. Globus, M.Y.T., Busto, R., Martinez, E., Valdes, I., Dietrich, W.D.,
Delatorre, J.C., 1994. Impaired brain microcirculation may trigger Alzheim- Ginsberg, M.D., 1991a. Comparative effect of transient global ischemia
er’s disease. Neurosci. Biobehav. Rev. 18, 397e401. on extracellular levels of glutamate, glycine, and gamma-aminobutyric
Dickey, C.A., Gordon, M.N., Wilcock, D.M., Herber, D.L., Freeman, M.J., acid in vulnerable and nonvulnerable brain regions in the rat. J. Neuro-
Morgan, D., 2005. Dysregulation of Naþ/Kþ ATPase by amyloid in chem. 57, 470e478.
APPþPS1 transgenic mice. BMC Neurosci. 6, 7. Globus, M.Y.T., Ginsberg, M.D., Busto, R., 1991b. Excitotoxic index e a bio-
Ditzler, K., 1991. Efficacy and tolerability of memantine in patients with de- chemical marker of selective vulnerability. Neurosci. Lett. 127, 39e42.
mentia syndrome. A double-blind, placebo controlled trial. Arzneimittel- Golde, T.E., 2006. Disease modifying therapy for AD? J. Neurochem. 99,
forschung 41, 773e780. 689e707.
Doody, R., Wirth, Y., Schmitt, F., Mobius, H.J., 2004. Specific functional ef- Goto, I., Taniwaki, T., Hosokawa, S., Otsuka, M., Ichiya, Y., Ichimiya, A.,
fects of memantine treatment in patients with moderate to severe Alzheim- 1993. Positron emission tomographic (PET) studies in dementia. J. Neurol.
er’s disease. Dement. Geriatr. Cogn. Disord. 18, 227e232. Sci. 114, 1e6.
Doody, R.S., Tariot, P.N., Pfeiffer, E., Olin, J.T., Graham, S.M., Bell, J.M., Grant, K.A., Colombo, G., Grant, J., Rogawski, M.A., 1996. Dizocilpine-like
2005. Meta-analysis of 6-month memantine clinical trials in Alzheimer’s discriminative stimulus effects of low-affinity uncompetitive NMDA an-
disease. Ann. Neurol. 58 (suppl. 9), 49e50. tagonists. Neuropharmacology 35, 1709e1719.
Durlach, J., 1990. Magnesium depletion and pathogenesis of Alzheimer’s Greenamyre, J.T., 1991. Neuronal bioenergetic defects, excitotoxicity and
disease. Magnes. Res. 3, 217e218. Alzheimer’s disease e use it and lose it. Neurobiol. Aging 12, 334e336.
720 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Grimwood, S., Gilbert, E., Ragan, C.I., Hutson, P.H., 1996. Modulation of Johnson, J.W., Kotermanski, S.E., 2006. Mechanism of action of memantine.
45Ca2þ influx into cells stably expressing recombinant human NMDA re- Curr. Opin. Pharmacol. 6, 61e67.
ceptors by ligands acting at distinct recognition sites. J. Neurochem. 66, Jones, K.W., Schaeffer, C.L., DeNoble, V.J., 1989. Systemically administered
2589e2595. N-methyl-D-aspartate interferes with acquisition of passive avoidance re-
Hallak, M., 1998. Effect of parenteral magnesium sulfate administration on ex- sponse in rats. Pharmacol. Biochem. Behav. 34, 181e186.
citatory amino acid receptors in the rat brain. Magnes. Res. 11, 117e131. Jones, M.W., McClean, M., Parsons, C.G., Headley, P.M., 2001. The in vivo
Harris, M.E., Wang, Y.N., Pedigo, N.W., Hensley, K., Butterfield, D.A., significance of the varied channel blocking properties of uncompetitive
Carney, J.M., 1996. Amyloid beta peptide (25e35) inhibits Naþ-depen- NMDA receptor antagonists. Neuropharmacology 41, 50e61.
dent glutamate uptake in rat hippocampal astrocyte cultures. J. Neuro- Kairane, C., Roots, K., Uusma, T., Bogdanovic, N., Karelson, E., Koks, S.,
chem. 67, 277e286. Zilmer, M., 2002. Regulation of the frontocortical sodium pump by Naþ
Hartmann, S., Mobius, H.J., 2003. Tolerability of memantine in combination in Alzheimer’s disease: difference from the age-matched control but sim-
with cholinesterase inhibitors in dementia therapy. Int. Clin. Psychophar- ilarity to the rat model. FEBS Lett. 531, 241e244.
macol. 18, 81e85. Katagiri, H., Tanaka, K., Manabe, T., 2001. Requirement of appropriate gluta-
Hattori, N., Kitagawa, K., Higashida, T., Yagyu, K., Shimohama, S., mate concentrations in the synaptic cleft for hippocampal LTP induction.
Wataya, T., Perry, G., Smith, M.A., Inagaki, C., 1998. CI-ATPase and Eur. J. Neurosci. 14, 547e553.
Naþ/K(þ)-ATPase activities in Alzheimer’s disease brains. Neurosci. Katz, E., Elgoyhen, A.B., Gomez-Casati, M.E., Knipper, M., Vetter, D.E.,
Lett. 254, 141e144. Fuchs, P.A., Glowatzki, E., 2004. Developmental regulation of nicotinic
Henneberry, R.C., 1989. The role of neuronal energy in the neurotoxicity of synapses on cochlear inner hair cells. J. Neurosci. 24, 7814e7820.
excitatory amino acids. Neurobiol. Aging 10, 611e613. Keller, J.N., Pang, Z., Geddes, J.W., Begley, J.G., Germeyer, A., Waeg, G.,
Herrero, J.F., Headley, P.M., Parsons, C.G., 1994. Memantine selectively Mattson, M.P., 1997. Impairment of glucose and glutamate transport and
depresses NMDA receptor-mediated responses of rat spinal neurones induction of mitochondrial oxidative stress and dysfunction in synapto-
in vivo. Neurosci. Lett. 165, 37e40. somes by amyloid beta-peptide: Role of the lipid peroxidation product
Herron, C.E., Lester, R.A.J., Coan, E.J., Collingridge, G.L., 1986. Frequency- 4-hydroxynonenal. J. Neurochem. 69, 273e284.
dependent involvement of NMDA receptors in the hippocampus: a novel Kennedy, A.M., Frackowiak, R.S.J., Newman, S.K., Bloomfield, P.M.,
synaptic mechanism. Nature 322, 265e269. Seaward, J., Roques, P., Lewington, G., Cunningham, V.J., Rossor, M.N.,
Hesselink, M.B., DeBoer, B.G., Breimer, D.D., Danysz, W., 1999a. Brain pen- 1995. Deficits in cerebral glucose metabolism demonstrated by positron
etration and in vivo recovery of NMDA receptor antagonists amantadine emission tomography in individuals at risk of familial Alzheimer’s disease.
and memantine: a quantitative microdialysis study. Pharm. Res. 16, Neurosci. Lett. 186, 17e20.
637e642. Kew, J.N.C., Trube, G., Kemp, J.A., 1998. State-dependent NMDA receptor
Hesselink, M.B., Smolders, H., DeBoer, A.G., Breimer, D.D., Danysz, W., antagonism by Ro 8-4304, a novel NR2B selective, non-competitive, volt-
1999b. Modifications of the behavioral profile of non-competitive age-independent antagonist. Br. J. Pharmacol. 123, 463e472.
NMDA receptor antagonists, memantine, amantadine and (þ)MK-801 af- Kim, J.H., Anwyl, R., Suh, Y.H., Djamgoz, M.B., Rowan, M.J., 2001. Use-de-
ter chronic administration. Behav. Pharmacol. 10, 85e98. pendent effects of amyloidogenic fragments of (beta)-amyloid precursor
Hesselink, M.B., Smolders, H., Eilbacher, B., DeBoer, A.G., Breimer, D.D., protein on synaptic plasticity in rat hippocampus in vivo. J. Neurosci.
Danysz, W., 1999c. The role of probenecid-sensitive organic acid transport 21, 1327e1333.
in the pharmacokinetics of N-methyl-D-aspartate receptor antagonists Kinsley, C.H., Madonia, L., Gifford, G.W., Tureski, K., Griffin, G.R.,
acting at the glycine(B)-site: microdialysis and maximum electroshock Lowry, C., Williams, J., Collins, J., McLearie, H., Lambert, K.G., 1999.
seizures studies. J. Pharmacol. Exp. Ther. 290, 543e550. Motherhood improves learning and memory. Nature 402, 137e138.
Hirai, H., Kirsch, J., Laube, B., Betz, H., Kuhse, J., 1996. The glycine binding Klein, W.L., Stine Jr., W.B., Teplow, D.B., 2004. Small assemblies of unmod-
site of the N-methyl-D-aspartate receptor subunit NR1: identification of ified amyloid beta-protein are the proximate neurotoxin in Alzheimer’s
novel determinants of co-agonist potentiation in the extracellular m3e disease. Neurobiol. Aging 25, 569e580.
m4 loop region. Proc. Natl. Acad. Sci. U.S.A. 93, 6031e6036. Kornhuber, J., Mack-Burkhardt, F., Kornhuber, M.E., Riederer, P., 1989.
Honegger, U.E., Quack, G., Wiesmann, U.N., 1993. Evidence for lysosomotrop- [3H]MK-801 binding sites in post-mortem human frontal cortex. Eur. J.
ism of memantine in cultured human cells e cellular kinetics and effects of Pharmacol. 162, 483e490.
memantine on phospholipid content and composition, membrane fluidity Kornhuber, J., Quack, G., 1995. Cerebrospinal fluid and serum concentrations
and beta-adrenergic transmission. Pharmacol. Toxicol. 73, 202e208. of the N-methyl-D-aspartate (NMDA) receptor antagonist memantine in
Howland, J.G., Wong, T., Fox, C.J., Ge, Y., Russell, K.I., Brebner, K., man. Neurosci. Lett. 195, 137e139.
Phillips, A.G., Weinberg, J., Christie, B.R., Wang, Y., 2006. Stress-induced Kornhuber, J., Weller, M., 1997. Psychotogenicity and N-methyl-D-aspartate
spatial memory disruption depends on long-term depression in the hippo- receptor antagonism: implications for neuroprotective pharmacotherapy.
campus. Soc. Neurosci. Abst. 32, 266.16. Biol. Psychiatry 41, 135e144.
Hoyer, S., 1996. Oxidative metabolism deficiencies in brains of patients with Kotermanski, S.E., Johnson, J.W., 2004. Partial trapping of memantine by
Alzheimer’s disease. Acta Neurol. Scand. 93, 18e24. NMDA receptors. Soc. Neurosci. Abst. 30, 731.7.
Hoyer, S., 2000. Brain glucose and energy metabolism abnormalities in Kwong, J.Q., Beal, M.F., Manfredi, G., 2006. The role of mitochondria in
sporadic Alzheimer disease. Causes and consequences: an update. Exp. inherited neurodegenerative diseases. J. Neurochem. 97, 1659e1675.
Gerontol. 35, 1363e1372. Lacor, P.-N., Buniel, M.C., Furlow, P.W., Clemente, A.S., Velasco, P.T.,
Hoyer, S., Nitsch, R., 1989. Cerebral excess release of neurotransmitter amino Wood, M., Viola, K.L., Klein, W.L., 2007. Ab oligomers-induced
acids subsequent to reduced cerebral glucose metabolism in early-onset de- aberrations in synapse composition, shape and density provide a molecular
mentia of Alzheimer type. J. Neural Transm. e Gen. Sect. 75, 227e232. basis for loss of connectivity in Alzheimer’s disease. J. Neurosci. 27,
Hoyer, S., Nitsch, R., Oesterreich, K., 1991. Predominant abnormality in cere- 796e807.
bral glucose utilization in late-onset dementia of the Alzheimer type e Ladner, C.J., Lee, J.M., 1999. Reduced high-affinity agonist binding at the
a cross-sectional comparison against advanced late-onset and incipient M(1) muscarinic receptor in Alzheimer’s disease brain: differential sensi-
early-onset cases. J. Neural Transm. 3, 1e14. tivity to agonists and divalent cations. Exp. Neurol. 158, 451e458.
Hutchin, T., Cortopassi, G., 1995. A mitochondrial DNA clone is associated Lambert, M.P., Barlow, A.K., Chromy, B.A., Edwards, C., Freed, R.,
with increased risk for Alzheimer disease. Proc. Natl. Acad. Sci. U.S.A. Liosatos, M., Morgan, T.E., Rozovsky, I., Trommer, B., Viola, K.L.,
92, 6892e6895. Wals, P., Zhang, C., Finch, C.E., Krafft, G.A., Klein, W.L., 1998.
Izumi, Y., Clifford, D.B., Zorumski, C.F., 1992. Low concentrations of Diffusible, nonfibrillar ligands derived from Abeta1e42 are potent cen-
N-methyl-D-aspartate inhibit the induction of long-term potentiation in tral nervous system neurotoxins. Proc. Natl. Acad. Sci. U.S.A 95,
rat hippocampal slices. Neurosci. Lett. 137, 245e248. 6448e6453.
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 721

Lanctot, K.L., Herrmann, N., Yau, K.K., Khan, L.R., Liu, B.A., Monyer, H., Burnashev, N., Laurie, D.J., Sakmann, B., Seeburg, P.H., 1994.
LouLou, M.M., Einarson, T.R., 2003. Efficacy and safety of cholinesterase Developmental and regional expression in the rat brain and functional
inhibitors in Alzheimer’s disease: a meta-analysis. CMAJ 169, 557e564. properties of four NMDA receptors. Neuron 12, 529e540.
Landfield, P.W., Morgan, G.A., 1984. Chronically elevating plasma Mg2þ im- Morris, R.G.M., Anderson, E., Lynch, G.S., Baudry, M., 1986. Selective
proves hippocampal frequency potentiation and reversal learning in aged impairment of learning and blockade of long-term potentiation by an
and young rats. Brain Res. 322, 167e171. N-methyl-D-aspartate receptor antagonist, AP5. Nature 319, 774e776.
Lanthorn, T.H., Mealing, G.A.R., Morley, P., 2000. Differences in degree of Nakagami, Y., Oda, T., 2002. Glutamate exacerbates amyloid beta1e42-
trapping between AR-R15896 and other uncompetitive NMDA receptor induced impairment of long-term potentiation in rat hippocampal slices.
antagonists. Amino Acids 19, 173e175. Jpn J. Pharmacol. 88, 223e226.
Laube, B., Hirai, H., Sturgess, M., Betz, H., Kuhse, J., 1997. Molecular deter- Nakamura, S., Murayama, N., Noshita, T., Katsuragi, R., Ohno, T., 2006. Cog-
minants of agonist discrimination by NMDA receptor subunits: analysis of nitive dysfunction induced by sequential injection of amyloid-beta and
the glutamate binding site on the NR2B subunit. Neuron 18, 493e503. ibotenate into the bilateral hippocampus; protection by memantine and
Lesne, S., Ali, C., Gabriel, C., Croci, N., MacKenzie, E.T., Glabe, C.G., MK-801. Eur. J. Pharmacol. 548, 115e122.
Plotkine, M., Marchand-Verrecchia, C., Vivien, D., Buisson, A., 2005. Nankai, M., Klarica, M., Fage, D., Carter, C., 1998. The pharmacology of na-
NMDA receptor activation inhibits alpha-secretase and promotes neuronal tive N-methyl-D-aspartate receptor subtypes: different receptors control the
amyloid-beta production. J. Neurosci. 25, 9367e9377. release of different striatal and spinal transmitters. Prog. Neuropsychophar-
Lesne, S., Koh, M.T., Kotilinek, L., Kayed, R., Glabe, C.G., Yang, A., macol. Biol. Psychiatry 22, 35e64.
Gallagher, M., Ashe, K.H., 2006. A specific amyloid-beta protein assembly Noda, M., Nakanishi, H., Akaike, N., 1999. Glutamate release from microglia
in the brain impairs memory. Nature 440, 352e357. via glutamate transporter is enhanced by amyloid-beta peptide. Neurosci-
Li, J.H., Wang, Y.H., Wolfe, B.B., Krueger, K.E., Corsi, L., Stocca, G., ence 92, 1465e1474.
Vicini, S., 1998. Developmental changes in localization of NMDA receptor Nomura, I., Kato, N., Kita, T., Takechi, H., 2005. Mechanism of impairment of
subunits in primary cultures of cortical. Eur. J. Neurosci. 10, 1704e1715. long-term potentiation by amyloid beta is independent of NMDA receptors
Lipton, S.A., 2004. Turning down, but not off. Nature 428, 473. or voltage-dependent calcium channels in hippocampal CA1 pyramidal
Lipton, S.A., 2005. The molecular basis of memantine action in Alzheimer’s neurons. Neurosci. Lett. 391, 1e6.
disease and other neurologic disorders: low-affinity, uncompetitive antag- Novelli, A., Reilly, J.A., Lysko, P.G., Henneberry, R.C., 1988. Glutamate be-
onism. Curr. Alzheimer Res. 2, 155e165. comes neurotoxic via the N-methyl-D-aspartate receptor when intracellular
Lipton, S.A., 2006. Paradigm shift in neuroprotection by NMDA receptor energy levels are reduced. Brain Res. 451, 205e212.
blockade: memantine and beyond. Nat. Rev. Drug Discov. 5, 160e170. Nowak, L., Bregestovski, P., Ascher, P., Herbert, A., Prochiantz, A., 1984.
Lipton, S.A., Chen, H.S., 2004. Paradigm shift in neuroprotective drug devel- Magnesium gates glutamate-activated channels in mouse central neurons.
opment: clinically tolerated NMDA receptor inhibition by memantine. Cell Nature 307, 462e465.
Death Differ. 11, 18e20. Nyakas, C., Szabó, R., Penke, B., Banerjee, P., Luiten, P.G.M., 2006. Meman-
Luhrs, L.B., Banerjee, P.K., LeFerla, F.M., 2006. Memantine reverses cogni- tine rescues cholinergic neurons from the neurotoxic effects of b-amyloid
tive deficits in transgenic mice with both plaques and tangles, P1e001. (Ab1e42) P4e305.
Lukoyanov, N.V., Paula-Barbosa, M.M., 2001. Memantine, but not dizocil- Oliver, D., Ludwig, J., Reisinger, E., Ruppersberg, J.P., Fakler, B., 2001. Mem-
pine, ameliorates cognitive deficits in adult rats withdrawn from chronic antine inhibits efferent cholinergic transmission in the cochlea by blocking
ingestion of alcohol. Neurosci. Lett. 309, 45e48. heteromeric alpha9/alpha10 acetylcholine receptors of outer hair cells.
Maskell, P.D., Speder, P., Newberry, N.R., Bermudez, I., 2003. Inhibition of Pfluegers Arch. Eur. J. Physiol. 441, R203.
human alpha 7 nicotinic acetylcholine receptors by open channel blockers Orgogozo, J.M., Rigaud, A.S., Stoffler, A., Mobius, H.J., Forette, F., 2002. Ef-
of N-methyl-D-aspartate receptors. Br. J. Pharmacol. 140, 1313e1319. ficacy and safety of memantine in patients with mild to moderate vascular
Masliah, E., Raber, J., Alford, M., Mallory, M., Mattson, M.P., Yang, D., dementia: a randomized, placebo-controlled trial (MMM 300). Stroke 33,
Wong, D., Mucke, L., 1998. Amyloid protein precursor stimulates excit- 1834e1839.
atory amino acid transport. Implications for roles in neuroprotection and Ozturk, S., Cillier, A.E., 2006. Magnesium supplementation in the treatment of
pathogenesis. J. Biol. Chem. 273, 12548e12554. dementia patients. Med. Hypotheses 67, 1223e1225.
Mattson, M.P., Barger, S.W., Cheng, B., Lieberburg, I., Smithswintosky, V.L., ParpuraGill, A., Beitz, D., Uemura, E., 1997. The inhibitory effects of beta-
Rydel, R.E., 1993. b-Amyloid precursor protein metabolites and loss of amyloid on glutamate and glucose uptakes by cultured astrocytes. Brain
neuronal Ca2þ homeostasis in Alzheimer’s disease. Trends Neurosci. Res. 754, 65e71.
16, 409e414. Parsons, C.G., Danysz, W., Quack, G., 1998. Glutamate in CNS disorders as
McShane, R., Areosa Sastre, A., Minakaran, N., 2006. Memantine for demen- a target for drug development: an update. Drug News Perspect. 11,
tia. Cochrane Database Syst. Rev. CD003154. 523e569.
Mecocci, P., Macgarvey, U., Beal, M.F., 1994. Oxidative damage to mitochon- Parsons, C.G., Danysz, W., Bartmann, A., Spielmanns, P., Frankiewicz, T.,
drial DNA is increased in Alzheimer’s disease. Ann. Neurol. 36, 747e751. Hesselink, M., Eilbacher, B., Quack, G., 1999a. Amino-alkyl-cyclohexanes
Mielke, R., Herholz, K., Grond, M., Kessler, J., Heiss, W.D., 1992. Differences are novel uncompetitive NMDA receptor antagonists with strong voltage-
of regional cerebral glucose metabolism between presenile and senile de- dependency and fast blocking kinetics: in vitro and in vivo characteriza-
mentia of Alzheimer type. Neurobiol. Aging 13, 93e98. tion. Neuropharmacology 38, 85e108.
Miguel-Hidalgo, J., Paul, I., Wanzo, V., Hachicha, M., Banerjee, P., 2005. Parsons, C.G., Danysz, W., Quack, G., 1999b. Memantine is a clinically well
Memantine inhibits the expression of caspase-8 and improves learning in tolerated N-methyl-D-aspartate (NMDA) receptor antagonist e a review of
rats injected with b-amyloid (Ab1e40). Soc. Neurosci. Abst. 31, 1019.5. preclinical data. Neuropharmacology 38, 735e767.
Minkeviciene, R., Banerjee, P., Tanila, H., 2004. Memantine improves spatial Parsons, C.G., Gilling, K.E., Jatzke, C. Blocking kinetics of memantine on
learning in a transgenic mouse model of Alzheimer’s disease. J. Pharma- NR1a/2A receptors recorded in inside-out and outside-out patches from
col. Exp. Ther. 311, 677e682. Xenopus oocytes. Eur. J. Pharmacol., submitted for publication.
Misztal, M., Frankiewicz, T., Parsons, C.G., Danysz, W., 1996. Learning Parsons, C.G., Gruner, R., Rozental, J., Millar, J., Lodge, D., 1993. Patch
deficits induced by chronic intraventricular infusion of quinolinic acid e clamp studies on the kinetics and selectivity of N-methyl-D-aspartate
protection by MK-801 and memantine. Eur. J. Pharmacol. 296, 1e8. receptor antagonism by memantine (1-amino-3,5-dimethyladamantan).
Mitani, A., Andou, Y., Kataoka, K., 1992. Selective vulnerability of hippocam- Neuropharmacology 32, 1337e1350.
pal CA1 neurons cannot be explained in terms of an increase in glutamate Parsons, C.G., Hartmann, S., Spielmanns, P., 1998. Budipine is a low affinity,
concentration during ischemia in the gerbil: brain microdialysis study. N-methyl-D-aspartate receptor antagonist: patch clamp studies in cultured
Neuroscience 48, 307e313. striatal, hippocampal, cortical and superior colliculus neurones. Neuro-
Molinuevo, J.L., 2003. Memantine. Neurologia 18, 255e261. pharmacology 37, 719e727.
722 C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723

Parsons, C.G., Panchenko, V.A., Pinchenko, V.O., Tsyndrenko, A.Y., Schmitt, F.A., Ashford, J.W., Ferris, S., Mackell, S.J., Schneider, L., et al.,
Krishtal, O.A., 1996. Comparative patch-clamp studies with freshly dissoci- 1996. Severe impairment battery: a potential measure for AD clinical
ated rat hippocampal and striatal neurons on the NMDA receptor antagonistic trials. In: Becker, R., Giacobini, E. (Eds.), Alzheimer Disease: From Mo-
effects of amantadine and memantine. Eur. J. Neurosci. 8, 446e454. lecular Biology to Therapy. Birkhäuser, Boston, pp. 419e423.
Parsons, C.G., Quack, G., Bresink, I., Baran, L., Przegalinski, E., Schmitt, F.A., Cragar, D., Ashford, J.W., Reisberg, B., Ferris, S., Mobius, H.J.,
Kostowski, W., Krzascik, P., Hartmann, S., Danysz, W., 1995. Comparison 2002. Measuring cognition in advanced Alzheimer’s disease for clinical
of the potency, kinetics and voltage-dependency of a series of uncompet- trials. Stoffler ADJ. Neural Transm. Suppl. 62, 135e148.
itive NMDA receptor antagonists in vitro with anticonvulsive and motor Schmitt, F.A., van Dyck, C.H., Wichems, C.H., Olin, J.T., 2006. Cognitive
impairment activity in vivo. Neuropharmacology 34, 1239e1258. response to memantine in moderate to severe Alzheimer disease patients
Periclou, A.P., Ventura, D., Sherman, T., Rao, N., Abramowitz, W.T., 2004. already receiving donepezil: an exploratory reanalysis. Alzheimer Dis.
Lack of pharmacokinetic or pharmacodynamic interaction between mem- Assoc. Disord. 20, 255e262.
antine and donepezil. Ann. Pharmacother. 38, 1389e1394. Schugens, M.M., Egerter, R., Daum, I., Schepelmann, K., Klockgether, T.,
Peskind, E.R., Potkin, S.G., Pomara, N., Ott, B.R., Graham, S.M., Olin, J.T., Loschmann, P.A., 1997. The NMDA antagonist memantine impairs classi-
McDonald, S., 2006. Memantine treatment in mild to moderate Alzheimer cal eyeblink conditioning in humans. Neurosci. Lett. 224, 57e60.
disease: a 24-week randomized, controlled trial. Am. J. Geriatr. Psychiatry Si, A., Helliwell, P., Maleszka, R., 2004. Effects of NMDA receptor antago-
14, 704e715. nists on olfactory learning and memory in the honeybee (Apis mellifera).
Porter, R.H.P., Greenamyre, J.T., 1995. Regional variations in the pharmacol- Pharmacol. Biochem. Behav. 77, 191e197.
ogy of NMDA receptor channel blockers: implications for therapeutic po- Simpson, I.A., Chundu, K.R., Davieshill, T., Honer, W.G., Davies, P., 1994.
tential. J. Neurochem. 64, 614e623. Decreased concentrations of GLUT1 and GLUT3 glucose transporters in
Puzzo, D., Arancio, O., 2006. Fibrillar beta-amyloid impairs the late phase of the brains of patients with alzheimers disease. Ann. Neurol. 35, 546e551.
long term potentiation. Curr. Alzheimer Res. 3, 179e183. Sobolevsky, A., Koshelev, S., 1998. Two blocking sites of amino-adamantane de-
Quack, G., Hesselink, M., Danysz, W., Spanagel, R., 1995. Microdialysis stud- rivatives in open N-methyl-D-aspartate channels. Biophys. J. 74, 1305e1319.
ies with amantadine and memantine on pharmacokinetics and effects on Sobolevsky, A.I., Koshelev, S.G., Khodorov, B.I., 1998. Interaction of memantine
dopamine turnover. J. Neural Transm. Suppl. 46, 97e105. and amantadine with agonist-unbound NMDA-receptor channels in acutely
Rammes, G., Rupprecht, R., Ferrari, U., Zieglgänsberger, W., Parsons, C.G., isolated rat hippocampal neurons. J. Physiol. (Lond.) 512 (pt 1), 47e60.
2001. The NMDA receptor channel blockers memantine, MRZ 2/579 Spanagel, R., Eilbacher, B., Wilke, R., 1994. Memantine-induced dopamine
and other amino-alkyl-cyclohexanes also antagonise 5-HT3 receptor cur- release in the prefrontal cortex and striatum of the rat e a pharmacokinetic
rents in HEK-293 and N1E-115 cells in a non-competitive manner. Neuro- microdialysis study. Eur. J. Pharmacol. 262, 21e26.
sci. Lett. 306, 81e84. Sucher, N.J., Akbarian, S., Chi, C.L., Leclerc, C.L., Awobuluyi, M.,
Rammsayer, T.H., 2001. Effects of pharmacologically induced changes in Deitcher, D.L., Wu, M.K., Yuan, J.P., Jones, E.G., Lipton, S.A., 1995. De-
NMDA-receptor activity on long-term memory in humans. Learn. Mem. velopmental and regional expression pattern of a novel NMDA receptor-
8, 20e25. like subunit (NMDAR-l) in the rodent brain. J. Neurosci. 15, 6509e6520.
Raschetti, R., Maggini, M., Sorrentino, G.C., Martini, N., Caffari, B., Takeda, A., Loveman, E., Clegg, A., Kirby, J., Picot, J., Payne, E., Green, C.,
Vanacore, N., 2005. A cohort study of effectiveness of acetylcholinesterase 2006. A systematic review of the clinical effectiveness of donepezil, riva-
inhibitors in Alzheimer’s disease. Eur. J. Clin. Pharmacol. 61, 361e368. stigmine and galantamine on cognition, quality of life and adverse events
Raymond, C.R., Ireland, D.R., Abraham, W.C., 2003. NMDA receptor regula- in Alzheimer’s disease. Int. J. Geriatr. Psychiatry 21, 17e28.
tion by amyloid-beta does not account for its inhibition of LTP in rat hip- Tariot, P.N., Farlow, M.R., Grossberg, G.T., Graham, S.M., McDonald, S.,
pocampus. Brain Res. 968, 263e272. Gergel, I., 2004. Memantine treatment in patients with moderate to severe
Reisberg, B., Doody, R., Stoffler, A., Schmitt, F., Ferris, S., Mobius, H.J., Alzheimer disease already receiving donepezil: a randomized controlled
2003. Memantine in moderate-to-severe Alzheimer’s disease. N. Engl. J. trial. JAMA 291, 317e324.
Med. 348, 1333e1341. Thompson, S., Lanctot, K.L., Herrmann, N., 2004. The benefits and risks
Rogawski, M.A., 1993. Therapeutic potential of excitatory amino acid antag- associated with cholinesterase inhibitor therapy in Alzheimer’s disease.
onists e channel blockers and 2,3-benzodiazepines. Trends Pharmacol. Expert Opin. Drug Saf. 3, 425e440.
Sci. 14, 325e331. de la Torre, J.C., Stefano, G.B., 2000. Evidence that Alzheimer’s disease is
Rogawski, M.A., Wenk, G.L., 2003. The neuropharmacological basis for the a microvascular disorder: the role of constitutive nitric oxide. Brain.
use of memantine in the treatment of Alzheimer’s disease. CNS Drug Res. Rev. 34, 119e136.
Rev. 9, 275e308. Townsend, M., Shankar, G.M., Mehta, T., Walsh, D.M., Selkoe, D.J., 2006. Ef-
Rogawski, M.A., Yamaguchi, S.I., Jones, S.M., Rice, K.C., Thurkauf, A., fects of secreted oligomers of amyloid beta-protein on hippocampal synap-
Monn, J.A., 1991. Anticonvulsant activity of the low-affinity uncompeti- tic plasticity: a potent role for trimers. J. Physiol. 572, 477e492.
tive N-methyl-D-aspartate antagonist (þ/)-5-aminocarbonyl-10,11-dihy- Van Dam, D., Abramowski, D., Staufenbiel, M., De Deyn, P.P., 2005. Symp-
dro-5H-dibenzo<a,d>cyclohepten-5, 10-imine (ADCI) e comparison tomatic effect of donepezil, rivastigmine, galantamine and memantine on
with the structural analogs dizocilpine (mK-801) and carbamazepine. cognitive deficits in the APP23 model. Psychopharmacology (Berl.) 180,
J. Pharmacol. Exp. Ther. 259, 30e37. 177e190.
Rosen, W.G., Mohs, R.C., Davis, K.L., 1984. A new rating scale for Alzheim- Van Dam, D., De Deyn, P.P., 2006. Cognitive evaluation of disease-modifying
er’s disease. Am. J. Psychiatry 141, 1356e1364. efficacy of galantamine and memantine in the APP23 model. Eur. Neuro-
Rowan, M.J., Klyubin, I., Cullen, W.K., Anwyl, R., 2003. Synaptic plasticity psychopharmacol. 16, 59e69.
in animal models of early Alzheimer’s disease. Philos. Trans. R. Soc. Vignini, A., Nanetti, L., Moroni, C., Tanase, L., Bartolini, M., Luzzi, S.,
Lond. B Biol. Sci. 358, 821e828. Provinciali, L., Mazzanti, L., 2006. Modifications of platelet from Alz-
Rowan, M.J., Klyubin, I., Wang, Q., Anwyl, R., 2004. Mechanisms of the heimer disease patients: a possible relation between membrane properties
inhibitory effects of amyloid beta-protein on synaptic plasticity. Exp. and NO metabolites. Neurobiol. Aging 28 (7), 987e994.
Gerontol. 39, 1661e1667. Walsh, D.M., Klyubin, I., Fadeeva, J.V., Cullen, W.K., Anwyl, R., Wolfe, M.S.,
Rowan, M.J., Klyubin, I., Wang, Q., Anwyl, R., 2005. Synaptic plasticity Rowan, M.J., Selkoe, D.J., 2002. Naturally secreted oligomers of amyloid
disruption by amyloid beta protein: modulation by potential Alzheimer’s beta protein potently inhibit hippocampal long-term potentiation in vivo.
disease modifying therapies. Biochem. Soc. Trans. 33, 563e567. Nature 416, 535e539.
Saxton, J., McGonigle, K.L., Swihart, A.A., Boller, F., 1993. The severe Walsh, D.M., Selkoe, D.J., 2004. Deciphering the molecular basis of memory
impairment battery. In: Suffolk (Ed.), The University of Pittsburgh of failure in Alzheimer’s disease. Neuron 44, 181e193.
the Commonwealth System of Higher Education. Thames Valley Test Walsh, D.M., Townsend, M., Podlisny, M.B., Shankar, G.M., Fadeeva, J.V., El
Company, pp. 1e16. Agnaf, O., Hartley, D.M., Selkoe, D.J., 2005. Certain inhibitors of
C.G. Parsons et al. / Neuropharmacology 53 (2007) 699e723 723

synthetic amyloid beta-peptide (Abeta) fibrillogenesis block oligomeriza- neurons of rats can be attenuated by glutamatergic antagonism or cycloox-
tion of natural Abeta and thereby rescue long-term potentiation. J. Neuro- ygenase-2 inhibition. Exp. Brain Res. 134, 58e65.
sci. 25, 2455e2462. Winblad, B., Poritis, N., 1998. Clinical improvement in a placebo-controlled
Wang, H.W., Pasternak, J.F., Kuo, H., Ristic, H., Lambert, M.P., Chromy, B., trial with memantine in care-dependent patients with severe dementia.
Viola, K.L., Klein, W.L., Stine, W.B., Krafft, G.A., Trommer, B.L., 2002. Neurobiol. Aging 19, S303.
Soluble oligomers of beta amyloid (1e42) inhibit long-term potentiation Wu, J.Q., Anwyl, R., Rowan, M.J., 1995. beta-amyloid selectively augments
but not long-term depression in rat dentate gyrus. Brain Res. 924, 133e140. NMDA receptor-mediated synaptic transmission in rat hippocampus. Neu-
Wang, Q., Rowan, M.J., Anwyl, R., 2004a. Beta-amyloid-mediated inhibition roreport 6, 2409e2413.
of NMDA receptor-dependent long-term potentiation induction involves Xiong, H., McCabe, L., Costello, J., Anderson, E., Weber, G., Ikezu, T., 2004.
activation of microglia and stimulation of inducible nitric oxide synthase Activation of NR1a/NR2B receptors by soluble factors from APP-stimu-
and superoxide. J. Neurosci. 24, 6049e6056. lated monocyte-derived macrophages: implications for the pathogenesis
Wang, Q., Walsh, D.M., Rowan, M.J., Selkoe, D.J., Anwyl, R., 2004b. Block of Alzheimer’s disease. Neurobiol. Aging 25, 905e911.
of long-term potentiation by naturally secreted and synthetic amyloid beta- Yamada, K., Takayanagi, M., Kamei, H., Nagai, T., Dohniwa, M.,
peptide in hippocampal slices is mediated via activation of the kinases Kobayashi, K., Yoshida, S., Ohhara, T., Takauma, K., Nabeshima, T.,
c-Jun N-terminal kinase, cyclin-dependent kinase 5, and p38 mitogen- 2005. Effects of memantine and donepezil on amyloid beta-induced mem-
activated protein kinase as well as metabotropic glutamate receptor type ory impairment in a delayed-matching to position task in rats. Behav. Brain
5. J. Neurosci. 24, 3370e3378. Res. 162, 191e199.
Wang, Q., Wu, J., Rowan, M.J., Anwyl, R., 2005. Beta-amyloid inhibition of Yang, L.S., Ksiezak-Reding, H., 1999. Ca2þ and Mg2þ selectively induce
long-term potentiation is mediated via tumor necrosis factor. Eur. J. Neuro- aggregates of PHF-Tau but not normal human Tau. J. Neurosci. Res. 55,
sci. 22, 2827e2832. 36e43.
Wary, C., Brillault-Salvat, C., Bloch, G., Leroy-Willig, A., Roumenov, D., Ye, C., Walsh, D.M., Selkoe, D.J., Hartley, D.M., 2004. Amyloid beta-protein
Grognet, J.M., Leclerc, J.H., Carlier, P.G., 1999. Effect of chronic magne- induced electrophysiological changes are dependent on aggregation state:
sium supplementation on magnesium distribution in healthy volunteers N-methyl-D-aspartate (NMDA) versus non-NMDA receptor/channel acti-
evaluated by 31P-NMRS and ion selective electrodes. Br. J. Clin. Pharma- vation. Neurosci. Lett. 366, 320e325.
col. 48, 655e662. Ye, L., Qiao, J.T., 1999. Suppressive action produced by beta-amyloid peptide
Wenk, G.L., Danysz, W., Mobley, S.L., 1994. Investigations of neurotoxicity and fragment 31e35 on long-term potentiation in rat hippocampus is N-
neuroprotection within the nucleus basalis of the rat. Brain Res. 655, 7e11. methyl-D-aspartate receptor-independent: it’s offset by ()huperzine A.
Wenk, G.L., Danysz, W., Roice, D.D., 1996. The effects of mitochondrial Neurosci. Lett. 275, 187e190.
failure upon cholinergic toxicity in the nucleus basalis. Neuroreport 7, Zajaczkowski, W., Frankiewicz, T., Parsons, C.G., Danysz, W., 1997. Uncom-
1453e1456. petitive NMDA receptor antagonists attenuate NMDA-induced impairment
Wenk, G.L., Parsons, C.G., Danysz, W., 2006. Potential role of NMDA recep- of passive avoidance learning and LTP. Neuropharmacology 36, 961e971.
tors as executors of neurodegeneration resulting from diverse insults focus Zajaczkowski, W., Hetman, M., Nikolaev, E., Quack, G., Danysz, W.,
on memantine. Behav. Pharmacol. 17, 411e424. Kaczmarek, L., 2000. Behavioural evaluation of long-term neurotoxic
Wesemann, W., Ekenna, O., 1982. Effect of 1-aminoadamantanes on the MAO effects of NMDA receptor antagonists. Neurotox. Res. 1, 299e310.
activity in brain, liver, and kidney of the rat. Arzneimittelforschung 32, Zajaczkowski, W., Moryl, E., Papp, M., 1996a. Discriminative stimulus effects
1241e1243. of the NMDA receptor antagonists MK-801 and CGP 37849 in rats. Phar-
Wesemann, W., Schollmeyer, J.D., Sturm, G., 1982. Distribution of Meman- macol. Biochem. Behav. 55, 163e168.
tine in brain, Liver, and blood of the rat. Arzneimittelforschung 32, Zajaczkowski, W., Quack, G., Danysz, W., 1996b. Infusion of (þ)-MK-801
1243e1245. and memantine e contrasting effects on radial maze learning in rats
Wesemann, W., Sturm, G., Funfgeld, E.W., 1980. Distribution of metabolism with entorhinal cortex lesion. Eur. J. Pharmacol. 296, 239e246.
of the potential anti-parkinson drug memantine in the human. J. Neural Zakharova, E., Danysz, W., Bespalov, A., 2005. Drug discrimination analysis
Transm. Suppl. 16, 143e148. of NMDA receptor channel blockers as nicotinic receptor antagonists in
Wilcock, G., Mobius, H.J., Stoffler, A., 2002. A double-blind, placebo- rats. Psychopharmacology 179, 128e135.
controlled multicentre study of memantine in mild to moderate vascular Zoladz, P.R., Campbell, A.M., Park, C.R., Schaefer, D., Danysz, W.,
dementia (MMM500). Int. Clin. Psychopharmacol. 17, 297e305. Diamond, D.M., 2006. Enhancement of long-term spatial memory in adult
Willard, L.B., Hauss-Wegrzyniak, B., Danysz, W., Wenk, G.L., 2000. The cy- rats by the noncompetitive NMDA receptor antagonists, memantine and
totoxicity of chronic neuroinflammation upon basal forebrain cholinergic neramexane. Pharmacol. Biochem. Behav. 85, 298e306.

You might also like