You are on page 1of 7

Mechanism of action of memantine

Jon W Johnson and Shawn E Kotermanski

Memantine is a clinically useful drug in many neurological effective in the treatment of other dementias [5] and
disorders, including Alzheimer’s disease. The principal neurodegenerative diseases, including Parkinson’s [5,6]
mechanism of action of memantine is believed to be the and Huntington’s [7] diseases. Memantine also shows
blockade of current flow through channels of N-methyl-D- promise as a treatment for other diseases associated with
aspartate (NMDA) receptors — a glutamate receptor subfamily excessive NMDAR activation in the central nervous
broadly involved in brain function. Surprisingly, other drugs that system (CNS), including glaucoma, multiple sclerosis,
block NMDA receptor channels, such as ketamine, exhibit epilepsy and neuropathic pain [2,5,6,8]. Further under-
serious deleterious effects. The unusual therapeutic utility of standing of the mechanism of action of memantine might
memantine probably results from inhibitory mechanisms help identify the properties responsible for its high clin-
shared with ketamine, combined with actions specific to ical potential, and hasten the development of other
memantine. These potentially important differences between therapeutic NMDAR antagonists.
memantine and ketamine include effects on gating of blocked
channels and binding of memantine to two sites on NMDA Sites of action of memantine
receptors. Because modulation of NMDA receptor activity can Memantine was first synthesized in the 1960s, and was
increase or decrease excitability of neuronal circuits, subtle found in the 1970s to affect the CNS [6]. In 1989,
differences in the mechanisms of action of NMDA receptor memantine was found to inhibit NMDARs [9,10] with
antagonists can strongly impact on their clinical effects. an IC50 of approximately 1 mM [11–19], which corre-
sponds well with its therapeutic concentration range. In
Addresses the treatment of AD, memantine is typically adminis-
Department of Neuroscience, University of Pittsburgh, Pittsburgh, tered at 20 mg/d [4]. Following administration of 5–
Pennsylvania, 15260, USA
30 mg/d of memantine in humans, cerebrospinal fluid
Corresponding author: Johnson, Jon W (johnson@bns.pitt.edu) concentrations of 0.05–0.31 mM were measured 2–3 hours
after the last dose; serum concentrations were about
twofold higher [20]. NMDARs are ubiquitous within
Current Opinion in Pharmacology 2006, 6:61–67
the vertebrate CNS, and are involved in a remarkable
This review comes from a themed issue on variety of physiological functions and pathological states
Neurosciences [21–23]. The idea that the principal target of memantine
Edited by Alan Foster and John Kemp is a receptor involved in the etiology of many nervous
Available online 20th December 2005 system diseases was, and remains, highly attractive [6,24–
26,27]. Thus, the discovery that memantine inhibits
1471-4892/$ – see front matter NMDARs initiated a continuing flood of studies.
# 2006 Elsevier Ltd. All rights reserved.

DOI 10.1016/j.coph.2005.09.007 At high concentrations (10–500 mM), memantine affects


many CNS targets, including serotonin and dopamine
uptake, nicotinic acetylcholine receptors (nAChRs), ser-
Introduction otonin receptors, sigma-1 receptors, and voltage-activated
Although overactivation of N-methyl-D-aspartate recep- Na+ channels [28,29]. In addition, higher affinity actions
tors (NMDARs) has been implicated in many neurode- have been observed on specific subtypes of nAChRs and
generative diseases [1], attempts to use NMDAR 5-hydroxytryptamine (5-HT) receptors. The nAChR sub-
antagonists as therapeutic agents often failed because unit combinations that have been shown to be most
of debilitating side effects [2,3]. These side effects occur sensitive to memantine (with the memantine IC50, esti-
because of the critical roles that NMDARs play in brain mated at 70 mV if possible, given in parentheses) are:
functions, including synaptic communication and mem- a4/b2 heteromers (14 mM) [30], a9/a10 heteromers
ory formation. Memantine (Figure 1a) has been hypothe- (1.6 mM) [31] and a7 homomers (5 mM [32] or 0.34 mM
sized to allow physiological activation of NMDARs while [33]). The significant discrepancy between the IC50
inhibiting pathological overactivation as a result of its estimates for homomeric a7 receptors might result from
mechanism of NMDAR inhibition. experimental differences (recombinant human receptors
expressed in Xenopus oocytes [32] versus native receptors
Memantine has been a focus of attention because of its in rat hippocampal neurons [33]) or inclusion of sub-
recent approval in Europe and the US for use in moderate units other than a7 in the native receptors [33]. Inhibi-
to severe Alzheimer’s disease (AD) [4]. However, mem- tion of nAChRs is unlikely to augment the therapeutic
antine might have far broader therapeutic utility, and is action of memantine, as loss of cholinergic neurons is a

www.sciencedirect.com Current Opinion in Pharmacology 2006, 6:61–67


62 Neurosciences

Figure 1 Properties of NMDARs


NMDARs are heteromeric ligand-gated ion channels
composed of four subunits (Figure 1b) [21]. Three
NMDAR subunit families have been described: NR1,
NR2 and NR3. Functional glutamate-dependent
NMDARs must contain NR1 and NR2 subunits; NR3
subunits are not obligatory and modulate NMDAR prop-
erties [37]. NMDARs require occupation by two types of
agonists for channel activation: a glutamate site agonist at
NR2 subunits, and a glycine site agonist at NR1 subunits
(Figure 1b).

The channels of NMDARs exhibit two unusual proper-


ties: voltage-dependent channel block by endogenous
Mg2+, and high permeability to Ca2+. Channel block by
Mg2+ inhibits more than 90% of the NMDAR-mediated
current at typical neuronal resting potentials. However,
postsynaptic depolarization changes the voltage field
across the channel, forcing Mg2+ out and enhancing
NMDAR-mediated currents. Thus, effective conduction
of current through NMDARs requires two coincident
signals — presynaptic release of glutamate and postsy-
naptic depolarization — allowing NMDARs to act as
‘coincidence detectors’.

The second unusual property of NMDAR channels —


high Ca2+ permeability — endows NMDAR activity with
profound physiological and pathological significance.
Coincident pre- and post-synaptic activity stimulates
Ca2+ influx through NMDARs, activating an array of
intracellular signaling pathways with diverse potential
consequences, including stabilization of synaptic connec-
tions, long-term depression or long-term potentiation of
synaptic strength, and either necrotic or apoptotic neu-
Memantine and NMDARs. (a) Structure of memantine. (b) Diagram ronal death [22,38–41]. Thus, inhibition of NMDAR
depicting the four-subunit quaternary structure of NMDARs (upper activity can have vastly divergent consequences, which
panel), and the transmembrane structure (lower panel) of two NMDAR can be either deleterious or beneficial.
subunits (shown in blue, with the front and back subunits removed for
clarity; note that the actual arrangement of subunits is not firmly
established), with glycine (green) bound to the NR1 subunit, glutamate Channel block of NMDA receptors by
(also green) bound to the NR2 subunit, and memantine (red) blocking the memantine
channel. The N-sites, asparagine residues critically involved in channel Memantine works by blocking the channel of NMDARs
blockade by Mg2+ and many organic blockers, are identified with (Figure 1b) [10–17,42–44]. Memantine is classified as an
asterisks. Locations of amino acids at which mutations cause greater
than a threefold change in memantine IC50 are identified by black dots.
‘open channel blocker’ because it can enter the channel
These amino acids, based on experiments with receptors composed of and block current flow only after channel opening. Over-
NR1 and NR2B (one of the four NR2 subunit subtypes) subunits [19], are lap of the sites where memantine and Mg2+ bind is
N616 (the N-site), A645, Y647, A653 and V656 in NR1; and W607, N615 suggested by the following two observations: Mg2+
(the N-site) and N616 in NR2B. Identical or homologous amino acids
decreases blockade by memantine [17], and mutation
confirmed as important in memantine channel blockade based on
experiments with receptors composed of NR1 and NR2A subunits [45] of ‘N-site’ asparagines — residues in the M2 region of
are N616 and A645 in NR1; N614 (the N-site) and N615 in NR2A. NR1 and NR2 subunits that are critical for Mg2+ block —
also strongly affects memantine block [19]. Other muta-
tions [19,45,46] have distinct effects on the block by
hallmark of AD [25,33] (but see [34]) and behavioral Mg2+ and memantine (Figure 1b), implying differences in
studies suggest that memantine does not have anticholi- access or binding sites.
nergic effects at clinically useful concentrations [35].
Memantine also inhibits steady-state 5-HT3 receptor After memantine blocks the channel of NMDARs, the
responses with an IC50 of 2.3 mM, an action that might channel can close and agonists can unbind, ‘trapping’
contribute to its therapeutic efficacy [36]. memantine inside the channel (Figure 2a) [14,15]. Many

Current Opinion in Pharmacology 2006, 6:61–67 www.sciencedirect.com


Mechanism of action of memantine Johnson and Kotermanski 63

Figure 2 antagonists with potentially diverse characteristics (see


below). By the first definition, memantine is not an
uncompetitive antagonist, because it can be trapped.
By the second definition, memantine is an uncompetitive
antagonist. Both definitions of uncompetitive are used
commonly to describe the mechanism of memantine
action (e.g. [6,13,15,25,26,42,43]). Because of the confu-
sion created by the various definitions of non-competitive
and uncompetitive, we find it best to avoid their applica-
tion to channel blockers.

Mechanistic differences between


memantine and ketamine
A critical and unresolved question is why memantine,
Schematic models of channel blockade of NMDARs. Colours of symbols
among the many NMDAR antagonists that have been
correspond to Figure 1. (a) Trapping model: an NMDAR (R) must bind
multiple agonist molecules (A), probably two glutamate and two tested in humans, is of unusual therapeutic utility. A host
glycines, to reach state AnR. Subsequently, multiple structural of intersecting hypotheses have been proposed to identify
rearrangements lead to channel opening (AnR*). The blocker molecule properties of memantine that endow it with clinical safety
then can bind in the open channel (AnR*B); the channel can close on the and efficacy, including the ability to bind only (or pre-
blocker (AnRB); and agonist can unbind, trapping the blocker (RB). (b)
‘Sequential’ or ‘foot-in-the-door’ model: because blocker binding
ferentially) to open channels; the tendency to inhibit
prevents channel closure, AnRB and RB states are not accessible to this faster, or with higher affinity, at higher agonist concen-
type of blocker. trations; a relatively low affinity of inhibition; relatively
fast unblocking kinetics; relatively strong voltage depen-
dence; an ability to be trapped in some but not all
‘trapping channel blockers’ of NMDARs have been receptors; an ability to inhibit at two different sites;
described, including MK-801, phencyclidine, ketamine and NMDAR subtype specificity, or lack thereof. The
and amantadine [2]. Several other channel blockers are clinical utility of memantine probably results from a
thought to prevent channel closure, and thus are bound in combination of these properties, and numerous reviews
the NMDAR channel only when it is open (Figure 2b) have explored the hypothetical relationship between the
[44,47]. Such blockers are called ‘sequential’ or ‘foot-in- properties of memantine and its therapeutic effects
the-door’ blockers. [3,6,8,25,26,28]. To limit and focus the rest of this review,
we concentrate on differences between memantine and
Channel blockers also are sometimes, although inconsis- ketamine, two drugs that appear to have strikingly similar
tently, referred to as non-competitive or uncompetitive. channel blocking properties, but dissimilar clinical
Because channel blockers do not compete for binding to effects. Both drugs are trapping blockers of NMDARs,
agonist binding sites, they can be generically called non- and thus bind exclusively or preferentially to open chan-
competitive antagonists (in contrast to competitive nels and inhibit with faster kinetics at higher agonist
antagonists such as D-2-amino-5-phosphonovaleric acid concentrations. The IC50 of ketamine is about half that
(APV), which binds to the glutamate site). However, of memantine; the blocking and unblocking kinetics and
these channel blockers can be further divided into two voltage dependence also are similar [12,13,43,48], and
separate subgroups: uncompetitive and, again, non-com- neither drug appears to exhibit strong NMDAR subtype
petitive. This second use of non-competitive creates specificity [11,18,49].
obvious potential for confusion. The distinction between
uncompetitive and non-competitive mechanisms derives It is likely that some of the properties shared by mem-
from enzyme kinetics: inhibitors that can be bound only antine and ketamine are required for the clinical utility of
to an enzyme–substrate complex (analogous to state AnR* memantine. However, despite their similarities, the ther-
in Figure 2) are uncompetitive, whereas inhibitors that apeutic potential of these drugs is strongly divergent.
bind equally well to all enzymatic states are non-compe- Ketamine causes memory deficits; reproduces with
titive. Further confusion of antagonist terminology has impressive accuracy the symptoms of schizophrenia; is
resulted from two subtly different applications of ‘uncom- widely abused; and induces vacuoles in neurons at mod-
petitive’ to channel blockers. The first definition — ‘an erate concentrations and cell death at higher concentra-
antagonist that can be bound to the receptor only when the tions [23,50]. Memantine, on the other hand, is well
channel is open’ — corresponds to uncompetitive enzyme tolerated; although instances of psychotic side effects have
inhibitors, and includes only sequential blockers been reported, in placebo-controlled clinical studies the
(Figure 2a). The second — ‘antagonists that can bind to incidence of side effects is remarkably low [4,6,25,51].
the receptor only when the channel is open’ — includes Memantine improves memory in AD patients and in some
both sequential and trapping blockers (Figure 2b), (but not all) studies in animals; does not appear to have

www.sciencedirect.com Current Opinion in Pharmacology 2006, 6:61–67


64 Neurosciences

abuse potential, but rather has shown promise in treating of channel closure [47]. The measurements, however,
addiction; and, although both drugs can be neuroprotective vary considerably. This variability might arise because
at low doses and neurotoxic at high doses, memantine channel blocking kinetics increase with agonist concen-
exhibits much weaker neurotoxicity [4,6,13,25,26,52,53]. tration: at low agonist concentrations, kinetics can be slow
and inhibition can appear weak if measurements are
How can two drugs that act by such similar mechanisms made too early. The observation of ‘partial trapping’ of
have such different effects? One possible explanation is memantine is also consistent with partial inhibition of
pharmacokinetic differences: memantine is eliminated, for channel closure. When agonist and a high concentration
example, much more slowly than ketamine. The elimina- of memantine are applied to a neuron, nearly all NMDAR
tion half-lives in human serum are: memantine (oral), 60– channels become blocked. If agonist and memantine are
80 h [51]; ketamine (imtravenous or oral), 2.5 h [54,55]. then simultaneously removed, some blocked channels do
The faster pharmacokinetics of ketamine might lead to the not trap memantine [14]. Partial trapping can result from
use of doses that cause a concentration spike resulting in partial inhibition of channel closure because the resulting
psychotomimetic effects. However, the psychotomimetic increase in channels in the open, blocked state (AnR*B in
effects of ketamine have been demonstrated [56] by a Figure 2a) allow more blocker to unbind before channel
continuous 40 min intravenous administration of ketamine closure [14]. However, alternative explanations for partial
at 0.0125 mg/kg/min (total dose, 0.5 mg/kg), a procedure trapping have been proposed, including closed-channel
that should minimize concentration spikes. Furthermore, egress of blocker [60] (see [61,62] for related discussion)
phencyclidine exhibits even stronger psychotomimetic and the presence of a second memantine binding site (see
effects than does ketamine [23,25], despite its longer below). The mechanism that underlies memantine partial
elimination half-life (7–51 hours via multiple routes of trapping might be important to its therapeutic use, as
administration [57]). Thus, we find it unlikely that phar- incomplete trapping can increase the number of
macokinetic differences can fully explain the different NMDARs available to support repetitive synaptic inputs.
clinical utilities of memantine and ketamine. We will Interestingly, a higher percentage of blocked channels
explore two other possible explanations for the clinical trap ketamine than they do memantine [43], suggesting
disparity of memantine and ketamine: differences in their the two drugs may have differential effects on channel
effects on NMDAR transitions among blocked states, and gating. Divergent effects of chemical modification of
differences in their ability to bind at multiple sites on NMDARs on the inhibition by memantine and ketamine
NMDARs. [46] also support the idea that these drugs affect channel
gating differently.
Transitions among blocked states of a receptor (states
AnR*B, AnRB and RB in Figure 2) have a surprisingly The second characteristic to be discussed here that could
powerful effect on the inhibitory properties of a blocker. be important in explaining the functional differences
For example, if a trapping blocker (Figure 2a) and a between memantine and ketamine is the ability of mem-
sequential blocker (which does not permit transitions antine to bind at a shallow, as well as a deep, site
among blocked states; Figure 2b) have equal affinities [14,16,17]. There is not general agreement on the char-
for the open channel, the trapping blocker can inhibit acteristics of the shallow site; although memantine can
responses with a much lower IC50 [47,58]. Furthermore, interact with a shallow site in the absence of agonist [14],
because a sequential blocker can only be bound to open other data suggest that a shallow site can be occupied only
channels, it inhibits most effectively (with the lowest when the channel is open [16,44]. This conflict might be
IC50) channels that spend longest in the open state partly resolved by the suggestion that occupation of the
(i.e. receptors with the highest Popen). A potential benefit shallow site in the absence of agonist can induce channel
is that, as glutamate concentration increases (e.g. during opening [17]. Although the IC50 of memantine for this
an ischemic insult), the antagonist becomes more effec- shallow site appears to be >100 mM, the ability of mem-
tive. However, depending upon a blocker’s kinetics, it antine to bind to two sites (an ability that has not been
might inhibit synaptic transmission most effectively, as reported for ketamine) might have important conse-
glutamate concentrations are highest during synaptic quences for its inhibitory effects. Further investigation
responses. Trapping blockers, conversely, can partly inhi- of the functional implications of this shallow memantine
bit, have no effect on, or even encourage channel closure. binding site is needed.
The clinically useful drug amantadine, an analog of
memantine, is a trapping blocker that encourages channel The importance of Mg2+ to understanding
closure [59]. channel-blocking drugs
Most studies of memantine block have been performed in
Data on the effects of memantine and ketamine on the absence of Mg2+. It will be important in future studies
channel gating are limited. Memantine inhibition has to examine carefully the interaction between Mg2+ and
been observed to increase with agonist concentration memantine. If Mg2+ and memantine cannot bind simul-
[12,33,42], an effect consistent with partial inhibition taneously, as appears likely, then Mg2+ and memantine

Current Opinion in Pharmacology 2006, 6:61–67 www.sciencedirect.com


Mechanism of action of memantine Johnson and Kotermanski 65

must compete for binding. Thus, at resting potential in to those of memantine. The beneficial effects of mem-
physiological Mg2+ concentrations, the IC50 of memantine antine and the neurotoxic effects of ketamine might
(or any similar channel blocker) should be strongly derive, in part, from shifts in the balance of the powerful
increased, and therapeutic memantine concentrations influences of NMDARs on excitation and inhibition in
should have negligible effects on NMDAR responses. the brain. Thus, apparently minor differences in the
Depolarizations that relieve Mg2+ block should increase mechanism of action of a drug can translate into great
inhibition by memantine because the voltage dependence differences in its clinical effects. Potentially important
of memantine inhibition is weaker than that of Mg2+. Large mechanistic differences between memantine and keta-
depolarization, however, would increase the IC50 of mem- mine include effects on channel function and the ability
antine to values far above therapeutic concentrations. of memantine to bind to two distinct sites on NMDARs.
Thus, significant effects of channel blockers at physiolo- However, a clear explanation for the clinical effectiveness
gical Mg2+ concentrations would be predicted only in of memantine is yet to be developed. A characteristic of
moderately depolarized neurons, and not during ‘normal’ memantine that deserves further investigation is its prob-
synaptic transmission at potentials near resting potential. able competition with Mg2+, which should allow it (like
other channel blocking drugs) to have significant effects
Enigmatic clinical effects of memantine only in moderately depolarized neurons. Deeper under-
It is surprising that a glutamate receptor antagonist can standing of the mechanisms of action of NMDAR chan-
improve the symptoms of AD, a disease characterized by nel blockers could lead to improved therapeutic strategies
deficits in the levels of both glutamate and glutamate for numerous neurological disorders.
receptors [27]. The idea that memantine can slow the
long-term progression of AD through neuroprotective Acknowledgements
actions is consistent with the ability of memantine to The authors thank P Ascher, TA Blanpied, RJ Clarke and BA Siegler for
insightful comments on the manuscript. This work was supported by
inhibit NMDAR responses. However, memantine also NIH grant MH45817.
exhibits therapeutic effects, such as improvements in
cognitive abilities, after as little as two weeks of treatment References and recommended reading
[2]. Several explanations for the cognitive benefits of Papers of particular interest, published within the annual period of
review, have been highlighted as:
memantine in AD have been proposed, including a
decrease in synaptic ‘noise’ resulting from excessive  of special interest
NMDAR activation [6], inhibition of b-amyloid produc-  of outstanding interest
tion or toxicity [25], and readjustment of the balance
1. Lodge D, Danysz W, Parsons CG: Ionotropic glutamate receptors
between inhibition and excitation [27]. The last hypoth- as therapeutic targets. Johnson City: FP Graham Pub; 2002.
esis, which relies on NMDAR actions on neuronal net- 2. Palmer GC: Neuroprotection by NMDA receptor antagonists
works, might also be relevant to neurotoxic NMDAR in a variety of neuropathologies. Curr Drug Targets 2001,
channel blockers such as MK-801, phencyclidine and 2:241-271.
ketamine. These antagonists have been proposed to kill 3. Lipton SA: Failures and successes of NMDA receptor
antagonists: molecular basis for the use of open-channel
neurons by reducing activity of inhibitory interneurons that blockers like memantine in the treatment of acute and chronic
depend heavily on NMDARs for their activation, resulting neurologic insults. NeuroRx 2004, 1:101-110.
in disinhibition of excitatory neurons [50]. Because the 4. Witt A, Macdonald N, Kirkpatrick P: Memantine hydrochloride.
activity and plasticity of both excitatory and inhibitory Nat Rev Drug Discov 2004, 3:109-110.
neurons depend critically on NMDARs, subtle variations 5. Kilpatrick GJ, Tilbrook GS: Memantine. Merz. Curr Opin Investig
in the action of an NMDAR inhibitor can cause great Drugs 2002, 3:798-806.
variations in its network effects. Furthermore, the balance 6. Parsons CG, Danysz W, Quack G: Memantine is a clinically
well tolerated N-methyl-D-aspartate (NMDA) receptor
between synaptic and extrasynaptic NMDAR activation antagonist — a review of preclinical data. Neuropharmacology
can determine whether neuron survival or death is encour- 1999, 38:735-767.
aged [22]. Thus, it is not surprising that inhibitors which 7. Beister A, Kraus P, Kuhn W, Dose M, Weindl A, Gerlach M:
appear to differ mechanistically even as little as memantine The N-methyl-D-aspartate antagonist memantine retards
progression of Huntington’s disease. J Neural Transm Suppl
and ketamine can exhibit such divergent therapeutic 2004:117-122.
potential. 8. Lipton SA: Paradigm shift in NMDA receptor antagonist drug
development: molecular mechanism of uncompetitive
Conclusions inhibition by memantine in the treatment of Alzheimer’s
disease and other neurologic disorders. J Alzheimer’s Dis 2004,
Memantine is an NMDAR antagonist with clinical utility 6:S61-S74.
in many nervous system diseases, of which the most 9. Bormann J: Memantine is a potent blocker of N-methyl-D-
prominent currently is AD, a disease characterized by aspartate (NMDA) receptor channels. Eur J Pharmacol 1989,
deficits in glutamatergic neurotransmission. The surpris- 166:591-592.
ing therapeutic effects of memantine contrast strongly 10. Kornhuber J, Bormann J, Retz W, Hubers M, Riederer P:
Memantine displaces [3H]MK-801 at therapeutic
with the deleterious effects of ketamine, an NMDAR concentrations in postmortem human frontal cortex.
antagonist with channel-blocking characteristics similar Eur J Pharmacol 1989, 166:589-590.

www.sciencedirect.com Current Opinion in Pharmacology 2006, 6:61–67


66 Neurosciences

11. Bresink I, Benke TA, Collett VJ, Seal AJ, Parsons CG, Henley JM, 29. Brau ME, Dreimann M, Olschewski A, Vogel W, Hempelmann G:
Collingridge GL: Effects of memantine on recombinant rat Effect of drugs used for neuropathic pain management on
NMDA receptors expressed in HEK 293 cells. Br J Pharmacol tetrodotoxin-resistant Na(+) currents in rat sensory neurons.
1996, 119:195-204. Anesthesiology 2001, 94:137-144.
12. Parsons CG, Gruner R, Rozental J, Millar J, Lodge D: Patch 30. Buisson B, Bertrand D: Open-channel blockers at the human
clamp studies on the kinetics and selectivity of N-methyl-D- alpha4beta2 neuronal nicotinic acetylcholine receptor.
aspartate receptor antagonism by memantine (1-amino-3,5- Mol Pharmacol 1998, 53:555-563.
dimethyladamantan). Neuropharmacology 1993, 32:1337-1350.
31. Oliver D, Ludwig J, Reisinger E, Zoellner W, Ruppersberg JP,
13. Parsons CG, Quack G, Bresink I, Baran L, Przegalinski E, Fakler B: Memantine inhibits efferent cholinergic transmission
Kostowski W, Krzascik P, Hartmann S, Danysz W: Comparison of in the cochlea by blocking nicotinic acetylcholine receptors of
the potency, kinetics and voltage-dependency of a series of outer hair cells. Mol Pharmacol 2001, 60:183-189.
uncompetitive NMDA receptor antagonists in vitro with
anticonvulsive and motor impairment activity in vivo. 32. Maskell PD, Speder P, Newberry NR, Bermudez I: Inhibition of
Neuropharmacology 1995, 34:1239-1258. human alpha 7 nicotinic acetylcholine receptors by open
channel blockers of N-methyl-D-aspartate receptors.
14. Blanpied TA, Boeckman FA, Aizenman E, Johnson JW: Trapping Br J Pharmacol 2003, 140:1313-1319.
channel block of NMDA-activated responses by amantadine
and memantine. J Neurophysiol 1997, 77:309-323. 33. Aracava Y, Pereira EF, Maelicke A, Albuquerque EX: Memantine
 blocks alpha7 nicotinic acetylcholine receptors more potently
15. Chen HS, Lipton SA: Mechanism of memantine block of NMDA- than n-methyl-D-aspartate receptors in rat hippocampal
activated channels in rat retinal ganglion cells: uncompetitive neurons. J Pharmacol Exp Ther 2005, 312:1195-1205.
antagonism. J Physiol 1997, 499:27-46. Memantine at clinically relevant concentrations is reported to inhibit
native a7 nAChR responses.
16. Sobolevsky A, Koshelev S: Two blocking sites of amino-
adamantane derivatives in open N-methyl-D-aspartate 34. Banerjee P, Samoriski G, Gupta S: Comments on ‘‘Memantine
channels. Biophys J 1998, 74:1305-1319. blocks alpha7* nicotinic acetylcholine receptors more
potently than N-methyl-D-aspartate receptors in rat
17. Sobolevsky AI, Koshelev SG, Khodorov BI: Interaction of
hippocampal neurons’’. J Pharmacol Exp Ther 2005,
memantine and amantadine with agonist-unbound
313:928-929 author reply 930-923.
NMDA-receptor channels in acutely isolated rat hippocampal
neurons. J Physiol 1998, 512:47-60. 35. Zakharova ES, Danysz W, Bespalov AY: Drug discrimination
18. Parsons CG, Danysz W, Bartmann A, Spielmanns P, Frankiewicz analysis of NMDA receptor channel blockers as nicotinic
T, Hesselink M, Eilbacher B, Quack G: Amino-alkyl- receptor antagonists in rats. Psychopharmacology (Berl) 2005,
cyclohexanes are novel uncompetitive NMDA receptor 179:128-135.
antagonists with strong voltage-dependency and fast 36. Rammes G, Rupprecht R, Ferrari U, Zieglgansberger W, Parsons
blocking kinetics: in vitro and in vivo characterization. CG: The N-methyl-D-aspartate receptor channel blockers
Neuropharmacology 1999, 38:85-108. memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes
19. Kashiwagi K, Masuko T, Nguyen CD, Kuno T, Tanaka I, Igarashi K, antagonise 5-HT(3) receptor currents in cultured HEK-293 and
Williams K: Channel blockers acting at N-methyl-D-aspartate N1E-115 cell systems in a non-competitive manner. Neurosci
receptors: differential effects of mutations in the vestibule and Lett 2001, 306:81-84.
ion channel pore. Mol Pharmacol 2002, 61:533-545.
37. Sasaki YF, Rothe T, Premkumar LS, Das S, Cui J, Talantova MV,
20. Kornhuber J, Quack G: Cerebrospinal fluid and serum Wong HK, Gong X, Chan SF, Zhang D et al.: Characterization and
concentrations of the N-methyl-D-aspartate (NMDA) receptor comparison of the NR3A subunit of the NMDA receptor in
antagonist memantine in man. Neurosci Lett 1995, 195:137-139. recombinant systems and primary cortical neurons.
J Neurophysiol 2002, 87:2052-2063.
21. Dingledine R, Borges K, Bowie D, Traynelis SF: The glutamate
receptor ion channels. Pharmacol Rev 1999, 51:7-61. 38. Aamodt SM, Constantine-Paton M: The role of neural activity in
synaptic development and its implications for adult brain
22. Hardingham GE, Bading H: The Yin and Yang of NMDA receptor function. Adv Neurol 1999, 79:133-144.
signalling. Trends Neurosci 2003, 26:81-89.
39. Nicoll RA, Malenka RC: Expression mechanisms underlying
23. Krystal JH, D’Souza DC, Petrakis IL, Belger A, Berman RM, NMDA receptor-dependent long-term potentiation.
Charney DS, Abi-Saab W, Madonick S: NMDA agonists and Ann N Y Acad Sci 1999, 868:515-525.
antagonists as probes of glutamatergic dysfunction and
pharmacotherapies in neuropsychiatric disorders. 40. Olney JW: Excitotoxicity, apoptosis and neuropsychiatric
Harv Rev Psychiatry 1999, 7:125-143. disorders. Curr Opin Pharmacol 2003, 3:101-109.

24. Smith PF: Therapeutic N-methyl-D-aspartate receptor 41. Perez-Otano I, Ehlers MD: Homeostatic plasticity and NMDA
antagonists: will reality meet expectation? Curr Opin Investig receptor trafficking. Trends Neurosci 2005, 28:229-238.
Drugs 2003, 4:826-832.
42. Chen HS, Pellegrini JW, Aggarwal SK, Lei SZ, Warach S,
25. Rogawski MA, Wenk GL: The neuropharmacological basis for Jensen FE, Lipton SA: Open-channel block of N-methyl-D-
the use of memantine in the treatment of Alzheimer’s disease. aspartate (NMDA) responses by memantine: therapeutic
CNS Drug Rev 2003, 9:275-308. advantage against NMDA receptor-mediated neurotoxicity.
J Neurosci 1992, 12:4427-4436.
26. Lipton SA: Paradigm shift in NMDA receptor antagonist drug
development: molecular mechanism of uncompetitive 43. Mealing GA, Lanthorn TH, Murray CL, Small DL, Morley P:
inhibition by memantine in the treatment of Alzheimer’s Differences in degree of trapping of low-affinity uncompetitive
disease and other neurologic disorders. J Alzheimers Dis 2004, N- methyl-D-aspartic acid receptor antagonists with
6:S61-S74. similar kinetics of block. J Pharmacol Exp Ther 1999,
288:204-210.
27. Schmitt HP: On the paradox of ion channel blockade
 and its benefits in the treatment of Alzheimer disease. 44. Bolshakov KV, Gmiro VE, Tikhonov DB, Magazanik LG:
Med Hypotheses 2005, 65:259-265. Determinants of trapping block of N-methyl-d-aspartate
The effects of NMDA response inhibition on the balance between synap- receptor channels. J Neurochem 2003, 87:56-65.
tic excitation and inhibition are hypothesized to explain the surprising
therapeutic utility of memantine in AD. 45. Chen HSV, Lipton SA: Pharmacological implications of
 two distinct mechanisms of interaction of memantine
28. Danysz W, Parsons CG, Kornhuber J, Schmidt WJ, Quack G: with N-methyl-D-aspartate-gated channels. J Pharmacol Exp
Aminoadamantanes as NMDA receptor antagonists and Ther 2005, 314:961-971.
antiparkinsonian agents–preclinical studies. Neurosci Covalent modification of mutant NMDARs with methanethiosulfonate
Biobehav Rev 1997, 21:455-468. reagents is used to investigate the approximate location of the external

Current Opinion in Pharmacology 2006, 6:61–67 www.sciencedirect.com


Mechanism of action of memantine Johnson and Kotermanski 67

memantine binding site, and mutagenesis is used to confirm the impor- 54. Persson J, Hasselström J, Maurset A, Öye I, Svensson J-O,
tance of some NMDAR amino acids to the deep memantine blocking site. Almqvist O, Scheinin H, Gustafsson LL, Almqvist O:
Pharmacokinetics and non-analgesic effects of S- and R-
46. Yuan H, Erreger K, Dravid SM, Traynelis SF: Conserved structural ketamine in healthy volunteers with normal and reduced
 and functional control of N-methyl-D-aspartate receptor metabolic capacity. Eur J Clin Pharmacol 2002, 57:869-875.
gating by transmembrane domain M3. J Biol Chem 2005,
280:29708-29716. 55. Grant IS, Nimmo WS, Clements JA: Pharmacokinetics and
After methanethiosulfonate modification of mutant NMDA receptors, analgesic effects of i.m. and oral ketamine. Br J Anaesth 1981,
receptor-mediated currents are observed even after removal of agonists. 53:805-810.
Interaction of channel blockers, including memantine and ketamine, with
modified receptors suggests differences in the mechanism of block. 56. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R,
Bremner JD, Heninger GR, Bowers MB, Charney DS:
47. Johnson JW, Qian A: Interaction between channel blockers and Subanesthetic effects of the noncompetitive NMDA
channel gating of NMDA receptors. Biologicheskie Membrany antagonist, ketamine, in humans. Arch Gen Psychiatry 1994,
2002, 19:17-22. 51:199-214.
48. Parsons CG, Panchenko VA, Pinchenko VO, Tsyndrenko AY, 57. Busto U, Bendayan R, Sellers EM: Clinical pharmacokinetics of
Krishtal OA: Comparative patch-clamp studies with freshly non-opiate abused drugs. Clin Pharmacokinetics 1989, 16:1-26.
dissociated rat hippocampal and striatal neurons on the
NMDA receptor antagonistic effects of amantadine and 58. Bolshakov KV, Kim KH, Potapjeva NN, Gmiro VE, Tikhonov DB,
memantine. Eur J Neurosci 1996, 8:446-454. Usherwood PN, Mellor IR, Magazanik LG: Design of antagonists
for NMDA and AMPA receptors. Neuropharmacology 2005,
49. Yamakura T, Mori H, Masaki H, Shimoji K, Mishina M: Different
49:144-155.
sensitivities of NMDA receptor channel subtypes to
non-competitive antagonists. Neuroreport 1993, 4:687-690. 59. Blanpied TA, Clarke RJ, Johnson JW: Amantadine inhibits NMDA
50. Sharp FR, Tomitaka M, Bernaudin M, Tomitaka S: Psychosis:  receptors by accelerating channel closure during channel
pathological activation of limbic thalamocortical circuits by block. J Neurosci 2005, 25:3312-3322.
psychomimetics and schizophrenia? Trends Neurosci 2001, Occupation of the channel of NMDARs by the clinically useful memantine
24:330-334. analog amantadine is shown to cause the channel to close more quickly.
This effect of amantadine on channel gating lowers its IC50.
51. Sonkusare SK, Kaul CL, Ramarao P: Dementia of Alzheimer’s
disease and other neurodegenerative disorders-memantine, a 60. Mealing GA, Lanthorn TH, Small DL, Murray RJ, Mattes KC,
new hope. Pharmacol Res 2005, 51:1-17. Comas TM, Morley P: Structural modifications to an
N-methyl-D-aspartate receptor antagonist result in large
52. Rogawski MA: Low affinity channel blocking (uncompetitive) differences in trapping block. J Pharmacol Exp Ther 2001,
NMDA receptor antagonists as therapeutic agents — toward 297:906-914.
an understanding of their favorable tolerability.
Amino Acids 2000, 19:133-149. 61. Sobolevsky AI, Beck C, Wollmuth LP: Molecular rearrangements
of the extracellular vestibule in NMDAR channels during
53. Danysz W, Parsons CG: The NMDA receptor antagonist gating. Neuron 2002, 33:75-85.
memantine as a symptomatological and neuroprotective
treatment for Alzheimer’s disease: preclinical evidence. 62. Qian A, Johnson JW: Channel gating of NMDA receptors.
Int J Geriatr Psychiatry 2003, 18:S23-S32. Physiol Behav 2002, 77:577-582.

www.sciencedirect.com Current Opinion in Pharmacology 2006, 6:61–67

You might also like