Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Drug Evaluation

A brief review of the


pharmacologic and therapeutic
aspects of memantine in
1. Introduction Alzheimer’s disease
2. Overview of
pharmacodynamic properties
Frederick Schmitt†, Melody Ryan & Gregory Cooper
†University
of Kentucky, Sanders-Brown Center on Aging, 800 S. Limestone Street, Lexington,
3. Pharmacokinetic characteristics: KY 40536-0230, USA
absorption and distribution
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

4. Metabolism and elimination The past decade has seen an increase in therapeutic options for Alzheimer’s
5. Food interactions
disease (AD) that target neurotransmitters, such as acetylcholine, and
research continues to target abnormal proteins in the AD brain. Recently,
6. Drug interactions
glutamate excitotoxicity has also become a target for AD treatment with the
7. Special populations advent of memantine. Clinical trial data reviewed for memantine show good
8. Safety and tolerability tolerability, low side-effect profiles and a positive therapeutic impact in mod-
9. Therapeutic use including erate-to-severe AD, both as monotherapy and in conjunction with donepezil.
dosage and administration However, additional data suggest variable benefits in the mild stages of AD.
10. Conclusion Furthermore, published reports support reduced dosing in patients with sig-
nificant renal disease. However, the opportunity to target a second mecha-
11. Expert opinion
nism in the treatment of AD, thereby providing added symptomatic benefit,
appears to be a useful consideration for clinicians who treat this devastating
For personal use only.

neurodegenerative disorder.

Keywords: Alzheimer's disease, clinical trials, dementia, excitotoxicity, glutamate, memantine

Expert Opin. Drug Metab. Toxicol. (2007) 3(1):135-141

1. Introduction

Alzheimer’s disease (AD) is a chronic, debilitating neurodegenerative disorder. It is


estimated that > 4 million individuals suffer from this illness in the US alone, with
numbers expected to rise dramatically in the coming decades. Approximately 10%
of all people aged > 65 years are afflicted with this condition. Despite the increasing
prevalence and importance of this disease, available treatments are limited. For the
past decade, the mainstays of treatment have been the acetylcholinesterase inhibitors
donepezil, rivastigmine and galantamine. These agents all afford a modest degree of
symptomatic benefit. They are based on the cholinergic hypothesis of AD, which
posits that the symptoms of this disease result, at least in part, from a deficiency in
the cholinergic system within the brain. However, over time, it has become clear
that additional pathways, neurotransmitter systems and biochemical processes also
play important roles in this disease. Key pathologic features of AD include changes
in amyloid precursor protein and tau protein [1-4] that lead to neuritic plaques and
For reprint orders, neurofibrillary tangles.
please contact: Recent attention has focused on the role of excitotoxicity within the CNS. It has
ben.fisher@informa.com
been shown that excitatory neurotransmitters, such as glutamate, can serve both a
physiologic, as well as neurotoxic, role in the brain. Such a neurotoxic role has been
implicated in a number of neurologic conditions ranging from ischemic stroke and
status epilepticus to neurodegenerative illnesses, including Huntington’s disease and
AD [5]. This has led to interest in the development of compounds capable of modu-
lating activity at glutamate receptors and, thereby, potentially attenuating excito-
toxicity, given emerging evidence that neurodegenerative diseases such as AD result,
in part, from derangements in glutamate homeostasis [5-7].

10.1517/17425255.3.1.135 © 2007 Informa UK Ltd ISSN 1742-5255 135


Memantine

Although a complete review of the glutamate hypothesis and studies, for example, have shown reduced cell loss in in vivo
neurodegenerative disease (e.g., AD) is beyond the scope of this rat models with administration of memantine [11,15-17].
review, other reviews have appeared in the literature regarding
glutamate and excitotoxicity [8,9]. However, some of the findings 2.1 Glutamate, neurotoxicity and
that are relevant to glutamate toxicity, long-term potentiation long-term potentiation
(LTP), β-amyloid toxicity and AD are briefly reviewed. Much of the hypothesized action of memantine hinges on
indications that glutamate excitotoxicity is involved in AD
2. Overview of pharmacodynamic properties and other dementing disorders [18,19]. In AD, the early obser-
vations that β-amyloid can reduce glial uptake of glutamate
Glutamate is the principal excitatory neurotransmitter within [20] and enhance glutamate toxicity [21] support the concept of
the CNS, in which up to 40% of synapses are glutamatergic. excitotoxicity as a mechanism in this disease. NMDA recep-
The NMDA receptor is a voltage-sensitive, glutamate-gated tors and glutamate may also have another role through cellu-
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

ion channel that, when activated, allows entry of Ca2+ into the lar mechanisms involving the promotion of calpain
neuron. Activation typically occurs only after depolarization degradation of the tau protein, leading to neuronal death [7].
by other ionotropic glutamate receptors (AMPA and kainate). The preclinical evidence for the effects of memantine is
At rest, the NMDA receptor channel is blocked by a magne- summarized in detail by Danysz and Parsons [22] and, there-
sium ion. It is been postulated that chronic overactivity at the fore, will not be repeated here. However, given the proposed
NMDA receptor, allowing excessive calcium influx into the mechanism of action of this compound, a brief commentary
cell, may play a role in the progression of AD. It is felt that on LTP seems warranted. LTP is viewed as a cellular substrate
excessive calcium influx may result in synaptic or dendritic of memory and involves NMDA receptors and intracellular
damage, necrosis or apoptosis [10,11]. calcium levels [23,24]. For example, animal studies suggest that
Memantine is a noncompetitive NMDA receptor anta- β-amyloid inhibits NMDA receptor-dependent LTP [25].
gonist. It acts selectively at NMDA receptors, with no activity Work by Parsons and colleagues [26-29] has investigated LTP
at AMPA/kainate receptors. It binds at, or near, the magne- blockades in different in vitro and in vivo animal models. This
For personal use only.

sium-binding site and thereby blocks Ca2+ entry into the cell work in the CA1 region of the hippocampus, using direct ago-
through the NMDA receptor at times of partial neuronal depo- nists of NMDA receptors as well as decreased magnesium
larization, but allows entry during full neuronal depolarization. concentration, reduced LTP. This ‘loss’ of LTP, however, is
Memantine is dependent on prior activation of the NMDA reversed with memantine administration. These and other
receptor by glutamate in order to access its binding site. There- observations have led to the hypothesis that memantine
fore, the NMDA channel must be open in order for meman- blocks the tonic overactivation of the NMDA receptors in
tine to gain access and exhibit its blocking activity [10,11]. AD, but allows the receptor (and LTP) to function in the
Memantine can rapidly occupy its receptor site and can quickly presence of a ‘relevant synaptic signal’ [22].
disassociate with the receptor. This property allows for normal
physiologic signal transmission during times of learning or 2.2 Clinical trials
recall, along with inhibiting pathologic, tonic overactivation Multiple large-scale, multisite, double-blind, placebo-control-
[10]. Finally, memantine exhibits a low affinity for the NMDA led trials have now been completed with memantine in people
receptor (> 500 nmol/l), a characteristic that likely allows for its with AD (for a review of clinical trial measures used in the eval-
good tolerability profile. Other NMDA antagonists, such as uation of AD therapies see [30]). In a study of 252 individuals
ketamine and phencyclidine, have high receptor affinities with moderate-to-severe AD (Mini-Mental State Exam
(< 100 nmol/l); therefore, they have had unacceptably high [MMSE]: 3 – 14, Global Deterioration Scale: 5 – 6, Functional
rates of neurotoxicity and, at times, have induced a psycho- Assessment Staging ≥ 6a), subjects were randomized to meman-
sis-like state. It is felt that this difference in receptor affinity tine, titrating to 20 mg/day, or placebo. Subsequent analysis
accounts for the relative lack of toxicity with memantine, possi- favored the memantine-treated group on multiple measures
bly by allowing for more dynamic adjustments of this including the Alzheimer’s Disease Cooperative Study’s
compound to ongoing synaptic activity [11]. (ADCS-ADL19) [31] measure of activities of daily living and the
Memantine may have neuroprotective qualities, as seen in a Severe Impairment Battery (SIB) [32] as a measure of cognition.
number of model systems, both in vivo and in vitro. Preven- Although the between-group difference in the global clinician
tion of NMDA- and glutamate-induced cell death has been rating (CIBIC-Plus) did not reach significance by the
shown in a number of culture systems, including rat retinal intent-to-treat analysis (p = 0.06), a significant difference favor-
ganglion, cerebellar, cortical, mesencephalic and hippocampal ing the memantine-treated group was seen in the observed cases
neurons. Interestingly, in other tissue culture experiments, analysis [33]. In addition, recently published results from this
memantine reduced tau hyperphosphorylation [12] and clinical trial’s open-label extension [34] appear to support the
promoted non-amyloidogenic amyloid precursor protein relative safety of this compound.
processing [11,13,14]. These effects may contribute to the In a larger study, 403 moderate-to-severe AD (MMSE:
observed efficacy of memantine in patients with AD. Other 5 – 14) patients taking stable dosages of donepezil were

136 Expert Opin. Drug Metab. Toxicol. (2007) 3(1)


Schmitt, Ryan & Cooper

randomized to the addition of memantine, titrating to 10 mg AD, the results of this trial revealed no discernable differ-
b.i.d., versus placebo [35]. In this study, significant ences due to the addition of memantine over placebo on
between-group differences favoring the memantine-treated either of the primary clinical outcome measures (ADAScog or
group were seen in the primary end point measures CIBIC-Plus) or for secondary outcomes of daily living skills
ADCS-ADL19 and SIB. Significant differences were noted on or behavioral symptoms. Of importance, however, are the
the secondary outcome measures, including the CIBIC-Plus, safety data that also showed no differences in treat-
Behavioural Rating Scale for Geriatric Patients (a measure of ment-emergent adverse events between the two groups [101].
patient dependence on the caregiver) and the Neuropsychiatric Therefore, these data lend themselves to speculations regard-
Inventory (a measure of behavioral disturbances) [35]. ing the nature of AD neurotransmitter therapies based on
Although both of the above trials involved only out-patients disease stage or progression, as well as the timing of different
with moderate-to-severe AD, many clinicians are faced with types of therapy for AD. Additional data from the
patients who may be in a facility or who may have other types open-label extensions of this trial, providing memantine for
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

of dementia. An earlier trial examined the efficacy of meman- up to 2 years, reveal the potential for slowing of AD pro-
tine, titrated to a dosage of 10 mg once daily, in 166 nursing gression (e.g., MMSE scores only declined by an average of
home patients with dementia [36]. A subsequent reanalysis of 4.6 points over 104 weeks), but without placebo controls no
this study separated these patients into two subgroups based clear conclusions can be derived from these unpublished
on their modified Hachinski Ischaemia Scale (HIS) scores. data regarding maintenance of symptomatic effects much
One group had risk factors for vascular dementia (VaD; HIS less disease modification.
score > 4) and the other with relatively ‘pure’ AD (HIS score
< 4) [37]. This reanalysis included 41 AD patients treated with 3.Pharmacokinetic characteristics:
memantine and 38 AD patients treated with placebo. In this absorption and distribution
study, significant between-group differences favoring the
memantine-treated group were seen on both a measure of glo- Memantine is well absorbed following oral dosing with a bio-
bal change and behavior/dependency (Behavioural Rating availability of ∼ 100% [39]. Following a linear dose–concentra-
For personal use only.

Scale for Geriatric Patients) for both AD and VaD patient tion pattern [40], the maximum serum concentration is
groups, although the placebo/drug difference was numerically achieved in 3 – 7 h, with food having no influence on the rate
larger for the AD group. or extent of absorption [102]. Steady-state serum memantine
Other clinical trial data involving memantine as either concentrations after an oral daily dose of 20 mg are in the
monotherapy or in combination with cholinesterase inhibi- range of 0.5 – 1.0 µM [41].
tors in milder stages of AD have appeared in publications and Memantine is not protein bound to a clinically significant
in abstract form. Summaries of the unpublished trials, includ- degree (45%) [103,104]. The relatively high volume of dis-
ing safety data from open-label extensions plus different titra- tribution (9 – 11 l/kg) demonstrates extensive distribution
tion and daily dosing schedules up to 20 mg (once daily) are throughout the body in the unionized state; memantine easily
available on the Forest Laboratories clinical trials website [101]. diffuses across biological membranes [40]. Importantly,
Results of a single trial of memantine monotherapy memantine has been shown to cross the blood–brain barrier
(20 mg/day) versus placebo in mild-to-moderate AD and to distribute to many areas within the brain [40,41].
(MMSE: 10 – 22) have recently been published [38]. In this In an attempt to determine the specific brain distribution
study, 403 subjects were randomized to memantine, titrating of memantine, radiolabeled [18F]-memantine was adminis-
to 10 mg b.i.d., versus placebo in a 24-week multi-center trial. tered intravenously to five healthy volunteers. Positron emis-
Significant differences favoring the memantine-treated group sion tomography scans were obtained over a 2-h period.
were seen on both primary measures, the CIBIC-Plus and the During this time, the level of radioactivity in the brain tissues
AD Assessment Scale (cognitive subscale or ADAScog, a meas- continued to increase, but never attained a peak or plateau.
ure of cognitive function). Benefit was also seen on the Neu- The amount of radioactivity in the brain tissue was dependent
ropsychiatric Inventory (a measure of behavioural on perfusion and possibly demonstrated nonspecific binding;
disturbance), although not on the ADCS-ADL23 (a measure only slightly related to the regional NMDA receptor concen-
of activities of daily living). Treatment discontinuations due trations in the various brain areas [42]. Although this study
to adverse events during the trial were 9.5% in the memantine determined that radiolabeling of memantine is not helpful for
and 5.0% in the placebo arms [38]. research purposes, it did demonstrate that memantine is
A second trial in mild-to-moderate AD has also been distributed to the brain.
completed. This trial randomized 427 mildly impaired AD A small, six-patient study demonstrated the achievement of
patients (baseline MMSE mean: 17), who were already on memantine cerebrospinal fluid (CSF) concentrations that
stable cholinesterase inhibitors (e.g., donepezil, galantamine, were linearly correlated with serum concentrations (R = 0.99,
rivastigmine) therapy for at least 3 months, to receive either p = 0.0018). The mean CSF/serum ratio was 0.52 ± 0.0904,
memantine or placebo for 24 weeks. Unlike the study that because only unbound memantine was available to cross the
combined memantine and donepezil in moderate-to-severe blood–brain barrier [102].

Expert Opin. Drug Metab. Toxicol. (2007) 3(1) 137


Memantine

4. Metabolism and elimination donepezil did not result in alterations of the pharmacokinetics
of either agent [44]. Memantine produces minimal inhibition
Memantine does not undergo extensive metabolism, with of CYP enzymes (CYP1A2, -2A6, -2C9, -2D6, -2E1 and
57 – 82% of dose eliminated unchanged in the urine [105]. -3A4), suggesting a lack of interaction with other drugs
Three memantine metabolites are formed: N-gludantan con- metabolized through this system [45]. Furthermore, although
jugate, 6-hydroxy memantine and 1-nitroso-deaminated memantine is predominantly excreted through the urine,
memantine. These metabolites have minimal NMDA recep- in vivo studies did not show a significant affect in AUC for
tor activity [102]. The CYP enzyme system has little involve- memantine after multiple doses of the diuretic hydrochloro-
ment in the metabolism of mematine, thus, the potential for thiazide/triamterene. Because of memantine’s low level of
drug–drug interactions is largely decreased [102]. The clearance plasma protein binding, any interaction with other highly
of memantine is ∼ 170 ml/min/1.73 m2. Clearance is reduced protein-bound medications, such as warfarin or digoxin, is
in the elderly and in those with decreased creatinine clearance unlikely [103].
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

(CrCl). The elimination half-life of memantine is 60 – 80 h The combined use of memantine with other NMDA
[105,106]. The correlation of memantine and CrCl emphasizes antagonists (e.g., amantadine, ketamine, dextromethorphan)
the role of urinary elimination for this compound. Meman- has not been systematically studied. Therefore, caution is
tine also undergoes renal tubular secretion and reabsorption urged when coadministering these agents [103].
via cationic transport proteins. Because memantine is a weak
base (pKa = 10.27), there are possible concerns with changes 7. Special populations
in the rate of elimination due to urine pH or urine flow
alterations [43]. Caution is also recommended in individuals with moder-
To study the effects of variations in urine pH and urine flow ate-to-severe renal insufficiency, with dosing adjustments
on memantine elimination, Freudenthaler et al. [43] designed a made in patients with moderately reduced CrCl. Different
crossover study. In this trial, 12 healthy males were randomized sources have different recommendations on the appropriate
to different sequences of the following groups: i) acidified use of memantine in renal insufficiency. The product infor-
For personal use only.

urine pH and reduced urinary flow; ii) acidified urine pH and mation provided by Forest Laboratories does not recommend
increased urinary flow; iii) alkalinized urine pH and reduced a dose reduction in moderate renal insufficiency, but does
urinary flow; and iv) alkalinized urine pH and increased uri- recommend a decrease to 5 mg b.i.d. if CrCl is in the range of
nary flow. Subjects were maintained at steady-state concentra- 5 – 29 ml/min [103]. The European Agency for Evaluation of
tions of memantine and then were subjected to the altered Medicinal Products suggest that memantine not be used in
urinary state for a 1-day period. Each period was separated by severe renal insufficiency and that a dose reduction to 10 mg
a wash-out phase. Alkalinization of the urine resulted in once daily is used if CrCl is in the range of 40 – 60
decreased renal excretion and renal clearance, probably stem- ml/min/1.73m2 [106]. A recent report from Periclou et al. [45]
ming from reduced renal reabsorption. The renal clearance directly examined the impact of renal insufficiency on
decreased to a mean of 19.4 – 30.5 ml/min versus a mean of memantine clearance. They enrolled 32 subjects in the age
148.6 ml/min without alterations. Acidification increased the range of 18 – 80 years (mean = 62.1, standard deviation [SD]
renal clearance to 223.3 – 234.3 ml/min [43]. Some patients = 10.6) and subdivided these individuals based on CrCl into
who may be taking memantine might undergo changes in four groups of eight subjects. Mean CrCl for the healthy and
their urine pH as a result of medications or diet. Alkaline urine mild groups were 93.5 (SD = 13.4) and 60.9 (SD = 7.9)
conditions can be caused by a pure vegetarian diet and acidic ml/min, respectively. Moderate impairment was in the range
urine conditions can result from a very protein-rich diet [43]. In of 50 – 75 ml/min (mean = 41.6, SD = 5.0) for eight subjects
this study, increasing urinary flow rate increased renal clear- and severe impairment was defined as 10 – 27 ml/min (mean
ance ∼ 9 ml/min. This increase was statistically significant, but = 20.1, SD = 5.7) in the remaining eight. Subjects received a
is unlikely to be clinically significant [43]. single 20-mg oral dose of memantine followed by blood draws
and urine specimen collections (up to 504 h post-dose). Urine
5. Food interactions samples were analysed for memantine plus its glucuronic acid
conjugate. Mean plasma concentrations were increased by
As noted above, absorption of oral memantine is not affected 60% in the moderate and 115% in the severe impairment
by food. No significant interactions with food have been groups compared with healthy controls. Elimination half-life
reported [103]. was increased by 95% in subjects with severe renal impair-
ment and 41% in moderately impaired subjects compared
6. Drug interactions with healthy subjects. Based on these data, Periclou et al. sim-
ulated the steady-state pharmacokinetic parameters across the
No in vitro [16] or in vivo [44] interactions with acetylcho- four renal groups. This simulation suggests that no change in
linesterase inhibitors have been noted. Furthermore, in dosing (e.g., 10 mg b.i.d.) is needed in persons with mild or
healthy volunteers, the coadministration of memantine and moderate renal insufficiency. However, based on their

138 Expert Opin. Drug Metab. Toxicol. (2007) 3(1)


Schmitt, Ryan & Cooper

modeling, they recommend a reduction of dosing to 5 mg pain (30 mg) [4], but no data with doses higher than
b.i.d. in persons with severe renal impairment. 20 mg/day are available for dementia. Although these doses
might be safe, it is unknown if increased efficacy would result.
8. Safety and tolerability Dosing typically starts at 5 mg daily and increases at weekly
intervals to a target of 10 mg b.i.d. As noted above and based
Memantine has demonstrated an excellent safety and on European Agency for the Evaluation of Medicinal Prod-
tolerability profile. In a 12-week nursing home-based trial of ucts guidelines, doses may be reduced for patients with mod-
memantine versus placebo, adverse events were noted in 22% erate renal insufficiency (CrCl: 40 – 60 ml/min/1.73m2) to
of memantine-treated subjects compared with 21% of pla- 10 mg once daily [106]. If memantine use is desired in patients
cebo-treated subjects [38]. In a 28-week study involving sub- with severe renal impairment (CrCl: 5 – 29 ml/min), the dose
jects with moderate-to-severe AD, 84% of subjects receiving should be reduced to 5 mg b.i.d.
memantine experienced an adverse event versus 87% of sub-
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

jects receiving placebo [33]. In patients with moder- 10. Conclusion


ate-to-severe AD already taking donepezil and, subsequently,
randomized to the addition of memantine versus placebo, Excitotoxicity has been hypothesized as an important mecha-
78% receiving memantine reported an adverse event versus nism in the pathogenesis of AD. Memantine is a non-
72% receiving placebo [35]. Most of these events were rated as competitive, low-affinity, voltage-dependent NMDA
mild-to-moderate and unrelated to treatment. Discontinua- receptor antagonist. It is thought to exert its activity by
tion in these trials was more common in placebo-treated sub- blocking calcium influx into neurons during time of partial,
jects. Similarly, in an integrated database of 587 patients with pathologic depolarization, and allowing normal receptor
dementia [102] serious adverse events were noted in 7% of channel functioning at times of full, physiologic neuronal
memantine recipients versus 10% of placebo recipients. Only depolarization. Animal models suggest a potential for neuro-
2% of memantine recipients versus 1% of placebo recipients protection, although this effect has not been confirmed in
experienced serious adverse events considered possibly related human studies. Emerging evidence from a number of large,
For personal use only.

to treatment. multi-center, double-blinded, placebo-controlled clinical


Adverse events noted in ≥ 5% of subjects receiving meman- trials demonstrate a clinical benefit [46] in the treatment of
tine with a greater than twofold incidence of that seen in sub- moderate-to-severe AD either as monotherapy or add-on
jects receiving placebo includes confusion (8 versus 2%) [35], therapy to a cholinesterase inhibitor.
somnolence (7 versus 1%) [44] and headache (6 versus 3%)
[34]. It should also be noted that a significantly lower incidence 11. Expert opinion
of agitation as an adverse event has been noted in memantine
treated subjects versus placebo (18 versus 32%) [33]. The introduction of memantine to the clinician’s armamen-
Finally, no clinically significant abnormalities have been tarium for moderate-to-severe AD has had an impact on the
noted on laboratory, electrocardiogram and vital sign treatment of this devastating neurodegenerative disorder.
measurements [33,35]. Although it has thus far been approved for only moder-
ate-to-severe disease, the clinical trial evidence is equivocal
9.Therapeutic use including dosage regarding its efficacy in mild AD. Its use as add-on therapy
and administration with the cholinesterase inhibitors is becoming commonplace,
and is supported by recent clinical trials in advanced AD.
Memantine has been approved for use in patients with mod- Over time, the use of this compound may increase and its role
erate-to-severe AD in both Europe and the US. Applications may grow in other related disorders, such as vascular dementia
for an expanded indication to include mild-to-moderate AD and other neurodegenerative illnesses, if the glutamate excito-
were submitted, but not approved, in the US [107]. The toxicity hypothesis gains in strength. Perhaps, based on the
present recommended dosage is 20 mg/day divided into data involving animal models, we may also see trials of this or
twice-daily dosing. Although the long half-life of 60 – 80 h similar agents that evaluate whether or not it serves a
would appear to support once-daily dosing, only one study neuroprotective role in addition to its symptomatic effects.
has used a once-daily regimen for dementia patients [37].
Other daily dosing and titration schemes have been studied in Conflicts of interest
open-label extension trials, but only safety data are available
on the Forest Laboratories website [101]. Higher daily doses of This work was supported by National Institute on Aging
memantine have been used in patients with phantom limb grants P50 AG05144 and R01 AG19241.

Expert Opin. Drug Metab. Toxicol. (2007) 3(1) 139


Memantine

Bibliography Alzheimer type abnormal involving reactive oxygen species and


1. SELKOE DJ: Alzheimer’s disease is a hyperphosphorylation of tau and associated calcium. Brain Res. (1995) 676(1):219-224.
synaptic failure. Science (2002) neurodegeneration. FEBS Lett. (2004) 22. DANYSZ W, PARSONS CG: The NMDA
298(5594):789-791. 566(1-3):261-269. receptor antagonist memantine as a
2. SELKOE DJ: Defining molecular targets to 13. KIMURA M, KOMATSU H, OGURA H, symptomatological and neuroprotective
prevent Alzheimer’s disease. Arch. Neurol. SAWADA K: Comparison of donepezil and treatment for Alzheimer’s disease: preclinical
(2005) 62(2):192-195. memantine for protective effect against evidence. Int. J. Geriatr. Psychiatry (2003)
amyloid-beta(1-42) toxicity in rat septal 18(Suppl. 1):S23-S32.
3. SELKOE DJ, SCHENK D: Alzheimer’s
neurons. Neurosci. Lett. (2005) 23. WOODSIDE BL, BORRONI AM,
disease: molecular understanding predicts
391(1-2):17-21. HAMMONDS MD, TEYLER TJ:
amyloid-based therapeutics. Ann. Rev.
Pharmacol. Toxicol. (2003) 43:545-584. 14. MIGUEL-HIDALGO JJ, ALVAREZ XA, NMDA receptors and voltage-dependent
CACABELOS R, QUACK G: calcium channels mediate different aspects
4. WIECH K, KIEFER RT, TOPFNER S
Neuroprotection by memantine against of acquisition and retention of a spatial
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

et al.: A placebo-controlled randomized


neurodegeneration induced by memory task. Neurobiol. Learn. Mem.
crossover trial of the N-methyl-D-aspartic
beta-amyloid(1-40). Brain Res. (2002) (2004) 81(2):105-114.
acid receptor antagonist, memantine, in
958(1):210-221. 24. HARNEY SC, ROWAN MJ, ANWYL R:
patients with chronic phantom limb pain.
Anesth. Analg. (2004) 98(2):408-413. 15. SEIF El NASR M, PERUCHE B, Long-term depression of NMDA
ROSSBERG C, MENNEL HD, receptor-mediated synaptic transmission is
5. GREENAMYRE JT, MARAGOS WF,
KRIEGLSTEIN J: Neuroprotective effect dependent on activation of metabotropic
ALBIN RL, PENNEY JB, YOUNG AB:
of memantine demonstrated in vivo and glutamate receptors and is altered to
Glutamate transmission and toxicity in
in vitro. Eur. J. Pharmacol. (1990) long-term potentiation by low intracellular
Alzheimer’s disease. Prog.
185(1):19-24. calcium buffering. J. Neurosci. (2006)
Neuropsychopharmacol. Biol. Psychiatry
16. WENK GL, ZAJACZKOWSKI W, 26(4):1128-1132.
(1988) 12(4):421-430.
DANYSZ W: Neuroprotection of 25. WANG Q, ROWAN MJ, ANWYL R:
6. AMADORO G, CIOTTI MT,
acetylcholinergic basal forebrain neurons by Beta-amyloid-mediated inhibition of
COSTANZI M, CESTARI V,
For personal use only.

memantine and neurokinin B. NMDA receptor-dependent long-term


CALISSANO P, CANU N: NMDA
Behav. Brain Res. (1997) 83(1-2):129-133. potentiation induction involves activation
receptor mediates tau-induced neurotoxicity
17. TREMBLAY R, CHAKRAVARTHY B, of microglia and stimulation of inducible
by calpain and ERK/MAPK activation.
HEWITT K et al.: Transient NMDA nitric oxide synthase and superoxide.
Proc. Natl. Acad. Sci. USA (2006)
receptor inactivation provides long-term J. Neurosci. (2004) 24(27):6049-6056.
103(8):2892-2897.
protection to cultured cortical neurons from 26. FRANKIEWICZ T, POTIER B,
7. RIEDERER P, HOYER S: From benefit to
a variety of death signals. J. Neurosci. (2000) BASHIR ZI, COLLINGRIDGE GL,
damage. Glutamate and advanced glycation
20(19):7183-7192. PARSONS CG: Effects of memantine and
end products in Alzheimer brain. J. Neural
18. TROTTI D, GIBB SL: Glutamate MK-801 on NMDA-induced currents in
Transm. (2006) 113(11):1671-1677.
transporters. In: Neurodegenerative diseases: cultured neurons and on synaptic
8. LIPTON SA: The molecular basis of transmission and LTP in area CA1 of rat
neurobiology, pathogenesis, and therapeutics.
memantine action in Alzheimer’s disease hippocampal slices. Br. J. Pharmacol. (1996)
Beal MF, Lang AE, Ludolph AC (Eds),
and other neurologic disorders: low-affinity, 117(4):689-697.
Cambridge University Press, Cambridge,
uncompetitive antagonism.
UK (2005):57-64. 27. ZAJACZKOWSKI W, FRANKIEWICZ T,
Curr. Alzheimer Res. (2005) 2(2):155-165.
19. IKONOMIDOU C, TURSKI L: PARSONS CG, DANYSZ W:
9. LIPTON SA: Paradigm shift in Uncompetitive NMDA receptor antagonists
Excitotoxicity and excitatory amino acid
neuroprotection by NMDA receptor attenuate NMDA-induced impairment of
antagonists in chronic neurodegenerative
blockade: memantine and beyond. passive avoidance learning and LTP.
diseases. In: Neurodegenerative diseases:
Nat. Rev. Drug Discov. (2006) Neuropharmacology (1997) 36(7):961-971.
neurobiology, pathogenesis, and therapeutics.
5(2):160-170.
Beal MF, Lang AE, Ludolph AC (Eds), 28. FRANKIEWICZ T, PARSONS CG:
10. LIPTON SA, CHEN HS: Paradigm shift in Cambridge University Press, Cambridge, Memantine restores long-term potentiation
neuroprotective drug development: UK (2005):44-56. impaired by tonic N-methyl-D-aspartate
clinically tolerated NMDA receptor (NMDA) receptor activation following
20. HARRIS ME, HENSLEY K,
inhibition by memantine. Cell Death Differ. reduction of Mg2+ in hippocampal slices.
BUTTERFIELD DA, LEEDLE RA,
(2004) 11(1):18-20. Neuropharmacology (1999)
CARNEY JM: Direct evidence of oxidative
11. ROGAWSKI MA, WENK GL: The injury produced by the Alzheimer’s 38(9):1253-1259.
neuropharmacological basis for the use of beta-amyloid peptide (1-40) in cultured 29. FRANKIEWICZ T, PILC A,
memantine in the treatment of Alzheimer’s hippocampal neurons. Exp. Neurol. (1995) PARSONS CG: Differential effects of
disease. CNS Drug Rev. (2003) 131(2):193-202. NMDA-receptor antagonists on long-term
9(3):275-308. potentiation and hypoxic/hypoglycaemic
21. MATTSON MP, GOODMAN Y:
12. LI L, SENGUPTA A, HAQUE N, Different amyloidogenic peptides share a excitotoxicity in hippocampal slices.
GRUNDKE-IQBAL I, IQBAL K: similar mechanism of neurotoxicity Neuropharmacology (2000) 39(4):631-642.
Memantine inhibits and reverses the

140 Expert Opin. Drug Metab. Toxicol. (2007) 3(1)


Schmitt, Ryan & Cooper

30. SCHMITT FA, WICHEMS CH: A Alzheimer’s type. American Association for Websites
systematic review of assessment and Geriatric Psychiatry 27th Annual Meeting.
treatment of moderate to severe Alzheimer's Baltimore, USA (2004). 101. http://www.forestclinicaltrials.com
disease. Prim. Care Companion J. 38. PESKIND E, POTKIN S, POMARA N: Forest Laboratories clinical trials website.
Clin. Psychiatry (2006) 8:158-169. Memantine monotherapy is effective and 102. http://www.emea.eu.int/humandocs/PDFs/
31. GALASKO D, SCHMITT F, sage for the treatment of mild-to-moderate EPAR/axura/094802en6.pdf
THOMAS R, JIN S, BENNETT D; Alzheimer’s disease: a randomized EMEA. Scientific Discussion (2004).
ALZHEIMER’S DISEASE controlled trial. J. Eur. Neuropharmacol. 103. http://www.fda.gov/cder/foi/label/2003/0214
COOPERATIVE STUDY: Detailed (2004) 14(Suppl. 3):332. 87lbl.pdf
assessment of activities of daily living in 39. JAIN KK: Evaluation of memantine for Approval Labelling Text (2003).
moderate-to-severe Alzheimer’s disease. neuroprotection in dementia. Expert Opin. 104. http://www.fda.gov/cder/foi/nda/2003/
J. Int. Neuropsychol. Soc. (2005) Investig. Drugs (2000) 9(6):1397-1406. 21-487_Namenda.htm
11(4):446-453.
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by RMIT University on 09/09/14

40. KORNHUBER J, QUACK G: Drug Approval Package (2003).


32. SCHMITT FA, ASHFORD W, Cerebrospinal fluid and serum 105. http://www.fda.gov/ohrms/dockets/ac/03/
ERNESTO C et al.: The severe impairment concentrations of the N-methyl-D-aspartate briefing/3979B1_01_ForestLabs-
battery: concurrent validity and the (NMDA) receptor antagonist memantine in Memantine.pdf
assessment of longitudinal change in man. Neurosci. Lett. (1995) Memantine HCl (2003).
Alzheimer’s disease. The Alzheimer’s disease 195(2):137-139.
Cooperative Study. Alzheimer Dis. Assoc. 106. http://www.emea.europa.eu/humandocs/PD
41. PARSONS CG, DANYSZ W, QUACK G: Fs/EPAR/ebixa/H-463-PI-en.pdf
Disord. (1997) 11(Suppl. 2):S51-S56.
Memantine is a clinically well tolerated European Public Assessment Report (2002).
33. REISBERG B, DOODY R, STOFFLER A, N-methyl-D-aspartate (NMDA) receptor
SCHMITT F, FERRIS S, MOBIUS HJ; 107. http://www.frx.com/news/pressrelease.aspx?id
antagonist – a review of preclinical data.
MEMANTINE STUDY GROUP: =734345
Neuropharmacology (1999) 38(6):735-767.
Memantine in moderate-to-severe TRIANO CE: Forest Laboratories
42. AMETAMEY SM, BRUEHLMEIER M, Announces FDA Decision on Supplemental
Alzheimer’s disease. N. Engl. J. Med. (2003)
KNEIFEL S et al.: PET studies of New Drug Application for Namenda®
For personal use only.

348(14):1333-1341.
18F-memantine in healthy volunteers. (2005).
34. REISBERG B, DOODY R, STOFFLER A, Nucl. Med. Biol. (2002) 29(2):227-231.
SCHMITT F, FERRIS S, MOBIUS HJ:
A 24-week open-label extension study of
43. FREUDENTHALER S, MEINEKE I, Affiliation
SCHREEB KH, BOAKYE E, Frederick Schmitt†1, Melody Ryan2 &
memantine in moderate-to-severe
GUNDERT-REMY U, GLEITER CH: Gregory Cooper3,4
Alzheimer’s disease. Arch. Neurol. (2006) †Author for correspondence
Influence of urine pH and urinary flow on
63(1):49-54. 1University of Kentucky, Sanders-Brown Center
the renal excretion of memantine. Br. J.
35. TARIOT PN, FARLOW MR, Clin. Pharmacol. (1998) 46(6):541-546. on Aging, and Department of Neurology, and
GROSSBERG GT, GRAHAM SM, Departments of Psychiatry, Psychology, and
44. PERICLOU AP, VENTURA D,
MCDONALD S, GERGEL I; Behavioural Sciences, 800 S. Limestone Street,
SHERMAN T, RAO N,
MEMANTINE STUDY GROUP: Lexington, KY 40536-0230, USA
ABRAMOWITZ WT: Lack of
Memantine treatment in patients with E-mail: fascom@email.uky.edu
pharmacokinetic or pharmacodynamic
moderate-to-severe Alzheimer’s disease 2University of Kentucky, Department of
interaction between memantine and
already receiving donepezil: a randomized Neurology, and Department of Pharmacy
donepezil. Ann. Pharmacother. (2004)
controlled trial. JAMA (2004) Practice and Science, 800 S. Limestone Street,
38(9):1389-1394.
291(3):317-324. Lexington, KY 40536-0230, USA
45. PERICLOU A, VENTURA D, RAO N, 3University of Kentucky, Sanders-Brown Center
36. WINBLAD B, PORITIS N: Memantine in
ABRAMOWITZ W: Pharmacokinetic
severe dementia: results of the 9M-Best on Aging, and Department of Neurology,
study of memantine in healthy and renally
Study (Benefit and efficacy in severely 800 S. Limestone Street, Lexington, KY
impaired subjects. Clin. Pharmacol. Ther.
demented patients during treatment with 40536-0230, USA
(2006) 79(1):134-143. 4Lexington Clinic, Division of Neurology,
memantine). Int. J. Geriatr. Psychiatry
(1999) 14(2):135-146. 46. SMITH M, WELLS J, BORRIE M: Lexington, KY, USA
Treatment effect size of memantine therapy
37. WINBLAD B, GRAHAM SM, LEE G:
in Alzheimer disease and vascular dementia.
Efficacy and tolerability of memantine in
Alzheimer Dis. Assoc. Disord. (2006)
nursing home patients with
20(3):133-137.
moderate-to-severe dementia of the

Expert Opin. Drug Metab. Toxicol. (2007) 3(1) 141

You might also like