Pharmacological Research 51 (2005) 1–17
Dementia of Alzheimer’s disease and other neurodegenerative disorders—memantine, a new hope S.K. Sonkusare, C.L. Kaul, P. Ramarao∗ Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali 160 062, India Accepted 19 May 2004
Abstract Alzheimer’s disease is the fourth largest cause of death for people over 65 years of age. Dementia of Alzheimer’s type is the commonest form of dementia, the other two forms being vascular dementia and mixed dementia. At present, the therapy of Alzheimer’s disease is aimed at improving both, cognitive and behavioural symptoms and thereby, quality of life for the patients. Since the discovery of Alzheimer’s disease by Alois Alzheimer, many pathological mechanisms have been proposed which led to the testing of various new treatments. Until recently the available drugs for the treatment of Alzheimer’s disease are cholinesterase inhibitors, which have limited success because these drugs improve cognitive functions only in mild dementia and cannot stop the process of neurodegeneration. Moreover, drugs of this category show gastrointestinal side effects. As the cells of central and peripheral nervous system cannot regenerate, newer strategies are aimed at preserving the surviving neurons by preventing their degeneration. NMDA-receptor-mediated glutamate excitotoxicity plays a major role in A-induced neuronal death. Hence, it was thought that NMDA receptors could be a promising target for preventing the progression of Alzheimer’s disease. All the compounds synthesized initially in this category showed toxicity mainly because of their high affinity for NMDA receptors. Memantine (1-amino adamantane derivative), NMDA-receptor antagonist was reported to be effective therapeutically in Alzheimer’s disease. It was available in Germany as well as European Union and has been approved for moderate to severe dementia in United States of America recently. It is an uncompetitive, moderate affinity antagonist of NMDA receptors that inhibits the pathological functions of NMDA receptors while physiological processes in learning and memory are unaffected. Memantine is also reported to have beneficial effects in other CNS disorders viz., Parkinson’s disease (PD), stroke, epilepsy, CNS trauma, amyotrophic lateral sclerosis (ALS), drug dependence and chronic pain. Mechanisms of neuroprotection, preclinical and clinical evidence for effectiveness of memantine have been provided. Pharmacology and pharmacokinetics of memantine and other NMDA-receptor antagonists in comparison with currently approved drugs for dementia treatment have been discussed. The focus is on ‘glutamate excitotoxicity’ and glutamate receptors as drug target. Various other novel strategies for the treatment of dementia of neurodegenerative disorders have also been discussed. © 2004 Elsevier Ltd. All rights reserved. Keywords: Dementia; Memantine; Neuroprotection; NMDA-receptor antagonist; Preclinical and clinical studies
1. Alzheimer’s disease—pathology and therapeutic options 1.1. Introduction Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by irreversible, progressive loss of memory followed by complete dementia [1]. The cognitive decline is accompanied by impaired performance of daily activities, behavior, speech and visual-spatial perception [2]. The disease, along with vascular and mixed dementia, is the ∗ Corresponding
author. Tel.: +91 172 2214683; fax: +91 172 2214692. E-mail address:
[email protected] (P. Ramarao).
1043-6618/$ – see front matter © 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.phrs.2004.05.005
commonest form of dementia affecting older people and accounts for 60–65% of dementia cases whereas vascular dementia and mixed dementia account for 15–20% of the cases each [3]. Neuritic plaques (consisting of a core of -amyloid aggregates covered by dead neurons, microglia and apolipoprotein E) and neurofibrillary tangles (paired helical filaments of microtubules and hyperphosphorylated tau proteins) are the major pathological lesions in an AD brain [4]. Differential diagnosis of the disease is difficult and AD can be confirmed only when autopsy reveals characteristic pathological changes in brain morphology and histology. Various hypotheses have been proposed for the pathogenesis of AD. These include glutamate excitotoxicity as a result of blockade of glutamate uptake into the astrocytes
2
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
by A aggregates, oxidative stress and membrane lipid peroxidation induced by A-aggregates, membrane lipid peroxidation due to C-terminal fragment of amyloid precursor protein (APP), microglial activation by A-aggregates and molecular pathways activated by A-induced stimulation of various kinases including MAP kinases and JNK (Jun amino-terminal kinase) [5]. Cognitive impairment in AD is caused mainly by death of cholinergic neurons in basal forebrain area, though other neurotransmitter systems could well be involved. A deficit of acetylcholine (ACh) in an Alzheimer’s disease brain is well known. At the same time, extent of dementia correlates well with the extent of neuronal death caused by excess of glutamate, the most prevalent excitatory neurotransmitter in the brain [6] (Fig. 1).
1.2. Therapeutic options for treatment of Alzheimer’s disease Alzheimer’s disease is the fourth largest cause of death for people over the age of 65 years. Prevalence of the disease is shown by the United Nations Population Projections estimate, which states that more than 100 million people will be affected by the disease in coming 50 years [7]. Since the last decade or so, AD patients are being treated with replacement of neurotransmitters that are deficient in AD brain, based on ‘cholinergic hypothesis’ of AD. Currently approved drugs for AD treatment are cholinesterase inhibitors that include tacrine (Cognex® ), rivastigmine (Exelon® ), donepezil (Aricept® ) and galantamine (Reminyl® ) (Tables 1
Fig. 1. (A) Steps involved in neurodegeneration of AD and targets therein for novel treatment strategies; (B) glutamate excitotoxicity in AD and its prevention by memantine. GSK: glycogen synthase kinase; CTF: C-terminal fragment; APP: amyloid precursor protein; MAPK: mitogen activated protein kinase, JNK: Jun amino-terminal kinase, NFT: neurofibrillary tangles; NSAID: non-steroidal anti-inflammatory agents; KA: kainic acid (A: calcium channel; B: membrane ion pore).
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
3
Fig. 1. (Continued ).
and 2). The approach is to inactivate acetyl cholinesterase (AChE), the enzyme that cleaves synaptic ACh and terminates neuronal signaling. These drugs have limited success as they only improve memory in mild dementia but cannot stop the process of neurodegeneration. The magnitude of response to AChE inhibitors depends on integrity of presynaptic neurons. Obviously, effectiveness of these agents will decrease with increase in severity of AD. Moreover, their use is associated with gastro-intestinal side effects [8]. First generation of cholinesterase inhibitors includes tacrine, physostigmine and NIK-247. These agents were non-selective and inhibited butyryl cholinesterase (plasma cholinesterase) as well. NIK-247 and tacrine also block potassium channels. Tacrine is known to cause hepatotoxicity and sedation. Selective inhibition of AChE markedly reduces peripheral adverse effects [9]. Second generation anticholinesterases were developed using this principle and include donepezil (Pfizer and Eisai) [10], galantamine (Janssen) [11] and eptastigmine (Mediolanum) [12]. Another approach is to develop prodrugs, which release the active component slowly in the blood giving long-term inhibition of cholinesterase. Metrifonate (Bayer) is one such drug that releases dimethyl-2,2-dichlorvinylphosphate. Huperazine A, a plant alkaloid having both, AChE as well as
antioxidant activity, is under investigation as a potential therapeutic option for treatment of AD [13]. As depression is commonly seen in AD patients, dual inhibitors of AChE-SERT (serotonin transporters) would be a better therapeutic option. Inhibition of serotonin transporters may also reduce dose-related side effects of AChE inhibitors. Such dual inhibitors were designed by hybridization of rivastigmine and fluoxetine [14]. 1.3. Novel therapeutic approaches for treatment of Alzheimer’s disease As the cells of central and peripheral nervous system cannot regenerate, newer strategies are aimed at preserving the surviving neurons by reducing their degeneration [15]. Some of these potentially disease modifying treatments include NMDA-receptor blockade, use of -sheet breakers, antioxidant strategies, amyloid--peptide vaccination, secretase inhibitors, cholesterol-lowering drugs, metal chelators, anti-inflammatory agents [16], inhibitors of tau phosphorylation, activators of phosphatases, neuroregeneration by neurotrophic factors and immunophylline ligands, supply of neuronal cells by gene therapy and human embryonic stem cells [17]. A brief description of these newer therapeutic strategies has been given below.
4
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
Table 1 Comparative pharmacology of currently approved drugs Drug Tacrine
(Cognex® )
Donepezil (Aricept® )
Rivastigmine (Exelon® )
Galantamine (Reminyl® ) Memantine (Ebixa® , Axura® , Akatinol® , Namenda® )
Mechanism
Adverse effects
Special comments
Non-selective cholinesterase inhibitor, more affinity towards synaptic G4 form of AChE than G1 form Selective AChE inhibitor affinity for G1 and G4 forms varies from region to region Non-selective inhibitor of cholinesterases, has higher affinity for cytoplasmic G1 Form of AChE Specifically inhibits AChE
Hepatotoxicity and gastro-intestinal symptoms such as nausea, anorexia, diarrhoea
Monitoring of liver transaminase levels is required; also inhibits K+ -channels weakly
Diarrhoea, nausea, anorexia, vomiting, muscle cramps, fatigue in some cases Nausea, vomiting and diarrhoea
Reversible inhibitor, high oral bioavailability and no hepatotoxicity Pseudo-irreversible inhibitor inactivated by enzymatic cleavage at the active site of enzyme and not by metabolism Also binds to presynaptic nicotinic receptors, stimulating ACh release Inhibits pathological but not physiological functions of NMDA receptors, also has antioxidant action and increases BDNF production
Moderate affinity, non-competitive inhibitor of NMDA receptors
Nausea, vomiting, anorexia, weight loss acutely at higher doses Mild and not common, constipation, confusion, headache, dizziness, tiredness
BDNF: brain-derived neurotrophic factor; G1 and G4 forms: isoforms of AChE; G1 form is a cytoplasmic whereas G4 form is synaptic.
• Inhibition of - and ␥-secretase reduces A levels in brain. Activation of ␣-secretase stimulates non-amyloidogenic pathway of APP proteolysis that forms harmless p3, thereby decreasing A levels. Inhibition of ␥-secretase inhibits cleavage of APP and Notch. Inhibition of notch cleavage affects embryogenesis, haematopoiesis, and thymocyte maturation. Partial inhibition of ␥-secretase does not inhibit its physiological functions [18]. • A fragments are cleared by phagocytosis and intra- and extracellular proteolysis. Insulin degrading enzyme and neprilysin [19] degrade A-fragments extracellularly. Activation of these enzymes or administration of exogenous enzymes can be beneficial. Delivery of genes that express neprilysin into neurons of frontal cortex and hippocampus reduces A-levels and neuronal death [20] in transgenic mouse model expressing human form of A.
• In AD brain, presence of oxidative stress is well known [21]. A-aggregates can directly insert into cell membrane and cause membrane lipid peroxidation. After internalization into the cell, A-aggregates damage mitochondrial membrane leading to electron leakage from mitochondrial electron transport system. This results into formation of intracellular free radicals. C-terminal fragment of APP also results into intracellular free radical formation in a similar way. These free radicals cause membrane lipid peroxidation and disturb the integrity of neuronal membrane. This finally results in neuronal death. Free radical formation also couples with calcium excitotoxicity and both these factors act synergistically to cause neuronal death. In this regard, antioxidants, particularly those, which can cross cell membrane, can prove beneficial in AD patients [5].
Table 2 Comparative clinical pharmacology of currently available drugs Characteristic
Tacrine
Rivastigmine
Donepezil
Galantamine
Memantine
Introduced Chemical class Maximum dose (mg per day) Times per day Initial dose (mg) tmax (h) Plasma protein binding (%) Elimination t1/2 (h) Metabolism by CYP450 system Interaction with food Drug–drug interactions Metabolites Bioavailability (%)
1993 Aminoacridine 160 4 40 1–2 55 2–3 Yes (CYP1A2) Not observed Yes Active 10–30
2000 Phenylcarbamate 12 2 3 1 40 1.5 No To be taken with mealsb None known Inactive 40
1996 Piperidine 10 1 5 3–4 96 70 Yes (CYP3A4) Not observed Yes Active 99
2001 Phenanthrene Alkaloid 24 2 8 1 18 7 Yes (CYP3A4) To be taken with mealsb Yes, some Active 90
2002a Amino Adamantine 20 1 10 3–8 45 60–80 No Not observed No Inactive 100
a The drug is being used over 10 years in Germany for the treatment of dementia but was approved for this purpose by the European Medicines Evaluation Agency in February 2002. b Food delays absorption, lowers C max and reduces adverse effects.
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
• Only insoluble aggregates of A are neurotoxic. -folded oligomeric forms of A are responsible for formation of insoluble A-fibrils. Compounds that prevent the formation of -folded forms (-sheet breakers) will be an obvious choice. Many such agents, e.g. laminin derivatives, rifampicin, daunomycin, etc. are being worked upon [22]. • A clinical study has shown that log-term treatment with NSAIDS decreases the risk of AD. Their effectiveness is mainly because of reduced generation of A1–42 which is more amyloidogenic. Anti-inflammatory activity plays a little role (by preventing microglial activation and release of neurotoxins) [23], as all anti-inflammatory agents do not show benefit in AD [24]. Unfortunately, results from clinical trials with NSAIDs are not encouraging [7,25]. • High blood cholesterol is a risk factor for AD. High cholesterol levels favour APP processing by -secretase pathway that is amyloidogenic, whereas low cholesterol favours processing by non-amyloidogenic ␣-secretase pathway [26]. Cholesterol depletion inhibits A-formation in brain hippocampus [27]. Moreover, cholesterol-lowering statins reduce A-levels in vitro and in vivo [28]. Statins also block interferon-␥-induced T-cell activation, thus reduces inflammation and has neuroprotective effect [29]. • Antibodies against A may enhance its excretion as observed in animal model of peripheral amyloidosis [30]. Antibodies can cross blood–brain barrier and trigger microglia to phagocytose A [24]. Such passive vaccination reduces the extent of amyloid plaque formation in transgenic mouse model of AD. • Active immunisation with A-peptide was well tolerated in phase I trials, but many patients developed meningoencephalitis in phase II. These trials confirmed the production of antibodies to A in humans, though the trials were halted [31–33]. • A in brain exists as membrane associated, soluble and aggregated forms. In AD, later two fractions are aggravated [34]. Zn2+ precipitates soluble A to form insoluble aggregate. Cu2+ and Fe3+ induce A-aggregation at acidic pH. All these ions are constitutively present in neocortex, which is the area most susceptible to AD. Depletion of biometal causes A-deposits to dissolve. Clioquinol prevents metal ions from binding to A and prevents A-deposition in transgenic mouse model for AD [35]. Metal ions catalyze H2 O2 production by A. Metal chelator act by preventing this process [36]. • Neuronal growth factor can prevent death due to excitotoxin and oxidative stress, but its delivery to brain is a problem as it cannot cross blood–brain barrier [37]. Induction of neurotrophic factors is another option. Nerve growth factor has a survival and growth promoting effect on basal forebrain neurons. • Co-localization of tau with microtubules is required for transport of essential nutrients and organelles and neurotransmitters along the microtubules. When tau protein is hyperphosphorylated, as in AD brain, co-localisation of tau and microtubules cannot take place and the transport
5
is hampered. GSK (glycogen synthase kinase)-3 and CDK5 (cyclin-dependent kinases) are implicated in tau-phosphorylation at sites that are phosphorylated in neurofibrillary tangles. These enzymes also accumulate in neurons with NFTs. Overexpression of GSK-3 results into tau-hyperphosphorylation. This indicates that inhibition of GSK-3 could be beneficial in AD [38]. • Oestrogen protects against risk of AD in ageing women by enhancing the outgrowth of nerve processes and formation of synaptic connections. It prevents neuronal death caused by oxidative stress and excitotoxicity and upregulates cholinergic and monoamine neurotransmitter transport system. In controlled trials, estrogen replacement therapy was protective against decline in verbal memory [39]. • Some of the disease modifying agents that are under phase II trials include phenserine of Axonyx (AChEI and A formation inhibitor), clioquinol of Prana Biotechnology (A aggregation inhibitor) and Alzhemed of Neurochem (A aggregation inhibitor) [40]. • Finally, if selective antagonists at NMDA receptors are used, death of cholinergic neurons in basal forebrain can be prevented [41].
2. Memantine—a new hope 2.1. Memantine vis-à-vis other NMDA-receptor antagonists NMDA-receptor-mediated glutamate excitotoxicity is a major factor responsible for A-induced neuronal death. NMDA receptors therefore appear promising target for preventing progression of neurodegeneration. Theoretically, any disorder of central nervous system characterized by glutamate excitotoxicity-induced neuronal death, should be cured with treatment of NMDA-receptor antagonists. These include diseases like Alzheimer’s disease (in which glutamate excitotoxicity is one of the causative factors for neuronal death), cerebral ischemia (dying neurons at the core release glutamate to overstimulate the neurons nearby), Parkinson’s and Huntington’s disease (in which compromised neurons are sensitized to glutamate excitotoxicity, though excess of glutamate is not seen). However, clinical trials with NMDA antagonists, before introduction of memantine, have failed, the reason being side effects of these drugs that occur because of high binding affinity towards NMDA receptors, differential actions on neurons from various regions of brain and interaction with neurotransmitter receptors other than NMDA receptors [42]. Mechanism-based side effects of these agents on central nervous system include hallucination, agitation, catatonia, centrally mediated increase in blood pressure and anaesthesia. In fact, NMDA antagonists like ketamine and phencyclidine were developed as anaesthetics only. Competitive NMDA-receptor antagonists bind to the same receptor site as glutamate and include midafotel and selfotel,
6
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
whose development has been discontinued because of their dose-related psychotomimetic side effects. Uncompetitive NMDA-receptor antagonists block the NMDA-receptor channel and prevent excessive flux of calcium inside the cell. The agents have been categorized on the basis of their in vitro binding affinities as low to moderate affinity antagonists (memantine, dextromethorphan and amantadine) and high-affinity antagonists (dizocilpine or MK-801, phencyclidine, ketamine). Uncompetitive antagonism is a type of non-competitive block i.e. it can not be overcome by increasing the concentration of glutamate [43]. High-affinity blockers like PCP and MK-801 block open ion channels and get trapped when the ion channels close, which means there is no unblocking. NMDA receptors (NR) are composed of an NR1 subunit in combination with NR2 subunit/s and a less common NR3 subunit. NR1 subunit is invariably present whereas NR2A, -B, -C or -D subunits determine the kinetics of channel opening and effect of various antagonists [44]. Subunit selective compounds therefore provide a better option for neuroprotection. Ifenprodil, an NR2B antagonist, has a less affinity for NMDA receptors and leaves non-activated receptors. Moreover, it acts mainly at persistently activated receptor ion-channels, sparing the transiently activated receptors (Table 3). Several subtypes selective NMDA antagonists have been studied that produce minimal adverse effects at neuroprotective doses. Agents selective for NR2B have shown their effectiveness in animal and primate models of Parkinson’s disease [45]. One such agent, traxoprodil is devoid of the side effects of non-selective NMDA antagonists in humans and clinical trials with traxoprodil in traumatic brain injury have shown some promise [46]. However, development of Gavestinel, a glycine-site antagonist, has been halted. Amantadine, a low-affinity NMDA-receptor blocker, is used in the treatment of Parkinson’s disease. Dextromethorphan (known for decades as a non-opioid cough suppressant),
another low-affinity NMDA antagonist, has been demonstrated to reduce epileptiform discharges reversibly in mouse cortical slices [47] and is neuroprotective in glutamate-induced excitotoxicity in retina [48]. It also has an analgesic effect in patients with diabetic neuropathy and neuropathic pain of traumatic origin [49]. Dextromethorphan reduces the amount of analgesics required perioperatively by 50% without serious side effects [50,51]. Low-affinity NMDA-receptor ion channels therefore may have upper hand over the high-affinity blockers in disorders like AD. Main reason behind this is a transient blockade of activated NMDA receptors by low-affinity agents, inhibiting their pathological functions only, leaving physiological functions unaltered. Memantine is one such agent that has been approved in European Union and Australia, whereas, US-FDA has approved it very recently in October, 2003. Above all, this agent does not show the side effects typically associated with high-affinity NMDA-blockers and its usefulness has been proved in several clinical trials across the globe. The drug has regenerated the interest in this category of drugs when NMDA antagonists were being thought of as “too toxic to be used therapeutically” in humans. Therapeutic goals in AD should be to improve the cellular energy status and the membrane functioning [52]. Memantine meets both these goals and is the first drug that can be used for the treatment of more advanced AD. 2.2. Development of memantine as a treatment for dementia Memantine is the first drug to be licensed in UK for treatment of severe AD [53] and the only treatment approved for more advanced AD [54]. Synthesized for the first time by Lilly (USA) in 1963 as a postulated hypoglycaemic agent, this drug was disappointing, as it did not have any blood sugar lowering activity. Merz Co. (Germany) was the first to identify the potential of this compound as a moderate
Table 3 NMDA antagonists at different stages of development Name of drug Agmatine Eliprodil Amantadine Amisulpiride Aptiganel Dizocilpine (MK-801) Fanapanel Ifenprodil Ketamine HCl Methoxy idazoxan Neramexane HCl Phencyclidine HCl Remacemide HCl Safinamide mesilate Selfotel a b
Ki . IC50 value.
Licensee Harvard University Pharmacia, Sanofi-Synth DuPont Pharmaceuticals Sanofi-Synth,Labo Cambridge NeuroScience Merck Sharp and Dohme Novo Nordisk, Schering AG Sanofi-Synth, Labo Pfizer, Bristol-Myer’s Squibb Pierre Fabre, Reckitt & Colman Forest, Merz Pfizer Astra Zeneca Newron Novartis
Developmental Stage
Ki or IC50
Preclinical Phase III Launched Launched Phase II Phase II Phase I Launched in 1973 Launched in 1966 Phase I Phase II Unspecified phase Phase III Phase II Phase III
219 M in rat cerebral 0.277 ± 0.103 in hippocampusb 219 M in cortexa >10 M in cortexb 0.029 ± 0.005 M in cortexa 0.0086 M in cortexa 11 ± 2.1 M in cortexb 10 M in cortexb 3.9 ± 0.5 M in entorhinal cortexb 190 M cortexa 1.47 ± 0.13 M in cortex a 1.06 ± 0.07 M in hippocampusb 968 ± 45 M in hippocampusb >100 M in hippocampusb 22.4 M in rat brainb
Reference cortexa
[142] [143] [142] [144] [145] [146] [147] [148] [124] [142] [60] [124] [124] [149] [150]
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
affinity non-competitive antagonist at NMDA receptors in 1972. It launched the drug in 1989 for the treatment of dementia. Merz and Lundbeck submitted an NDA for memantine in AD in November 2000. The compound was in phase II trials in the US for the treatment of AIDS-related dementia and pain by August 1996 and phase III trials for glaucoma and neuroprotection by 1999. Analysts at Merrill Lynch predicted in October 2001 that Allergan would make regulatory filings in the US for memantine in glaucoma and ocular hypertension in 2005 [55]. At the same time, the drug is under development by Lundbeck, Neurobiological Technologies Inc. (NTI), Suntory for the treatment of AD, AIDS-related dementia, pain in patients with neuropathy and by Allergan for the treatment of ocular diseases. Memantine was also found beneficial in patients with vascular dementia [56,57] and in Wernicke–Korsakoff syndrome [58]. This well-tolerated drug (as shown by several clinical trials) is being used over 10 years in Germany where it is being marketed by Merz Co. under the trade name of Axura® for the treatment of dementia and was approved for this purpose by the European Medicines Evaluation Agency in February 2002. Namenda® of Forest Laboratories was approved by US-FDA in October 2003 for use in moderate to severe AD. Compared with acetylcholine-esterase inhibitors, memantine can be beneficial not only for mild but advanced dementia as well. Before the approval of memantine by US-FDA, it was predicted that the approval would make it a blockbuster drug. After US-FDA approval, memantine looks all set to cross US$ 1 billion mark in annual sales by the year 2005 [59]. Ebixa® , launched by Lundbeck Ltd. in October 2002, has been licensed for the treatment of moderately severe to severe AD. Memantine treatment leads to reduction in overall cost for management of AD patients by reducing utilization of resources. 2.3. Chemical structure of memantine Memantine (1-amino-3,5-dimethyladamantane), an amino-alkyl cyclohexane derivative (1-amino adamantine series), is a white bitter solid that is soluble in DMSO (Fig. 2). Aminoadamantanes are atypical drug compounds with non-planar, three dimensional tricyclic structure [46]. Memantine hydrochloride is readily soluble in water. The NH 3+ Cl-
R6
R5
R1
H 3C CH 3
(A)
R4
R2
R3
(B)
Fig. 2. Structure of memantine hydrochloride (A) and basic structure of aminoalkyl cyclohexane (B).
7
aminoadamantanes represent a class of drugs, which may be largely free of side effects of other NMDA antagonists, and which have already been used clinically as antiviral and antiparkinsonian agents. It has been predicted that amino-alkyl-cyclohexanes having methyl substitutions at R1, R2, and R5, at least one methyl or ethyl at R3 or R4 and a charged amino-containing substitution at R6 (given in the diagram below), could be useful therapeutics in a wide range of CNS disorders involving disturbances in glutamatergic neurotransmission [60]. Kinetic analysis of the molecule shows that amino adamantane-induced block of open NMDA channels is mediated by two distinct blocking sites. These sites are located in the depth of the channel pore and can be simultaneously occupied by two blocking molecules [61]. Memantine has a log P value of 3.28 and pKa of 10.27 which make this drug suitable for oral drug delivery. • Molecular formula of memantine: C12 H21 N, • Molecular weight of memantine: 179.30, • CAS registry number: 41100-52-1.
3. Neuroprotection by memantine 3.1. Mechanisms involved in neuroprotection About 70% of all excitatory synapses in the central nervous system are stimulated by glutamate. Glutamate release from the vesicles in presynaptic neuron occurs via a calcium-dependent mechanism that involves N- and P/Q-type calcium channels. Glutamate concentration in a vesicle is around 100 mmol/L. Glutamate acts at three types of ionotropic receptors on post-synaptic membrane. These receptors have been named after their agonists as N-methyl-d-aspartate (NMDA), ␣-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and kainate receptors. NMDA receptors are present mainly in neurons and their activation results into Ca2+ influx [62] whereas AMPA- and kainate receptors are present in neurons as well as in glia and cause Na+ -influx and K+ -efflux on activation [44]. In the last decade, molecular biology of NMDA receptors has also been explored. It is now known that NMDA receptors have an NR1 subunit [63] along with one or more NR2 subunits and less commonly, NR3 subunit [64]. Four NR2 subunits (A–D) determine the biophysical and pharmacological characteristics of NMDA receptors though alternative splicing of NR1, especially in exon 5 also plays some role [45]. Glutamate is the most potent agonist at NMDA receptors in hippocampus, its EC50 being 2.3 mol/L. Two special features of NMDA receptors are voltage-dependent blockade by Mg2+ ions at physiological concentrations [65] and requirement of glycine as a coagonist [44]. Each receptor unit has two binding sites for glutamate and two for glycine. Opening of NMDA receptor ion channel and calcium entry
8
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
is a complex process, which requires binding of both, glutamate and glycine. Glutamate is released from the presynaptic terminal whereas glycine is present constitutively in the extracellular fluid at a constant level. Schaffer-collateral pathway between CA1 and CA3 regions of hippocampus plays a major role in storage of learning and memory. Long-term potentiation (LTP), which can be defined as long lasting enhancement of post-synaptic potential in response to a brief stimulus of high frequency, is responsible for long-term memory and changes in synaptic structure and strength in the hippocampus. Glutamate is the most important neurotransmitter involved in expression of LTP. Glutamate receptors could be ionotropic as well as metabotropic, but ionotropic NMDA receptors are more important for LTP. Metabotropic glutamate receptors are broadly divided into three groups—group I receptors act via stimulation of phospholipase C and production of diacylglycerol and inositol triphosphate as second messengers whereas groups II and III receptors inhibit adenyl cyclase. Transient release of high concentration of glutamate in synaptic cleft activates AMPA-receptors on post-synaptic membrane, leading to membrane depolarization that removes voltage-dependent Mg2+ -block of NMDA receptors [66]. This results into Ca2+ -influx inside the neuron. Ca2+ -calmodulin dependent kinases then induce long-term activation of neighbouring AMPA and kainate receptors. Sustained activation of NMDA receptors results into activation of MAP kinases (mitogen activated protein kinases), which translocate to the nucleus and phosphorylate CREB (cyclic AMP response element binding protein). This leads to rapid induction of several immediate early genes that give way to late response genes. This is the mechanism underlying synaptic plasticity and increased synaptic strength [45]. In short, long-term memory involves orderly activation of genes. Calcium is required in the brain for synaptic transmission, neuronal development, synaptic plasticity and several metabolic processes, but improper regulation of intracellular calcium ions is associated with pathological features of AD i.e. neuritic plaques and neurofibrillary tangles. Glutamate excitotoxicity cannot be an initiating factor for neurodegeneration, but plays an important role in events triggered by some other processes like aggregates of -amyloid (A) and reduced energy metabolism in the neurons [67]. Chronic excess of glutamate causes overactivation of NMDA receptors and excess of intracellular calcium ions and leads to excitotoxicity (the mechanism responsible for neuronal death because of glutamate excitotoxicity in neurodegenerative disorders). Recently, it has been proposed that increased calcium influx into the cytoplasm results into accumulation of calcium inside mitochondria [68]. This leads to generation of reactive oxygen species that destroy Ca2+ -ATPase and hence to reduced ability of cell membrane to expel Ca2+ -ions. This causes further increase in mitochondrial calcium ions and when a threshold is reached, mitochondrial permeability transition pores (a way out for both calcium ions and com-
pounds with a molecular weight <1.5 kDa) open irreversibly. This is followed by exit of Ca2+ -ions and macromolecular compounds that predetermine the cell death like cyt c (amplifier for apoptotic signal), Apaf-1 (trigger for apoptosis) and caspase activators from the mitochondria [69]. This explains the role played by glutamate excitotoxicity in apoptotic cell death. Overloading of a cell with calcium ions also results into overactivation of calcium-calmodulin-dependent nitric oxide synthase (NOS). NO formed by this enzyme then combines with superoxide radicals to form peroxynitrite, which may cause DNA-fragmentation. Lastly, excess of calcium ions inside the cell overactivates various kinases, including those responsible for tau-phosphorylation and JNK (Jun amino-terminal kinase). NMDA receptors are, therefore, promising targets for neuroprotective drugs as high permeability of these receptors for Ca2+ ions is responsible for their neurotoxic potential [70,71]. Radioligand binding studies have proved selective interaction of A with recognition sites for glutamate and glycine on NMDA receptors. Glutamate transporters on the glial membrane are Na+ -dependent and transmembrane gradient of Na+ and K+ is the driving force for the transport. A-induced elevation of cytosolic Ca2+ causes further Ca2+ release from endoplasmic reticulum by IP3 -signalling. These changes in glial membrane conductance reduces glial glutamate uptake by blockade of Na+ -dependent glutamate transporters [72]. This increases glutamate concentration in synaptic cleft beyond physiological levels [73,74] causing calcium excitotoxicity-induced neuronal death by activating NMDA receptors on post-synaptic neuronal membrane. Calcium overload may result in protein hyperphosphorylation such as that of tau proteins [75], activation of proteolytic enzymes and even DNA breakdown leading to apoptotic cell death. A has also been found to increase the voltage-dependant release of glutamate in vitro [76]. This glutamate then exerts its excitotoxicity by acting on glutamate receptors (docking sites) on the cell surface resulting into calcium influx into the neurons [77]. Sustained elevation of glutamate leads to overloading of the cell with calcium and even after the transient release of glutamate that occurs during learning and memory process, the signal is not detected because of already elevated Ca2+ inside the cells. This results into impairment of learning and memory and symptoms of dementia. Though NMDA receptors are involved in long-term potentiation, continuous activation of NMDA receptors increases noise and probability of arriving signal to be detected is reduced. This couples with neuronal damage caused by Ca2+ excitotoxicity and ultimately produces neuronal dysfunction. Memantine blocks NMDA receptors non-competitively in use-dependent manner i.e. only when the channels are open. Memantine blocks NMDA receptor when there is sustained release of low glutamate concentrations. The influx of calcium is prevented (neuroprotection) [78]. The intracellular calcium pool is reduced. During learning and memory processes (i.e. transient high glutamate release), meman-
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
tine—the fast, voltage-dependent NMDA-receptor antagonist—leaves the NMDA receptor for a short time and a signal is produced that can be recognized and processed (symptomatic improvement in dementia symptoms). Thus, memantine blocks the neurotoxicity of glutamate without interfering with its physiological actions required for learning and memory [3]. Memantine was found to have antioxidant property which is useful since calcium excitotoxicity is associated with oxidative stress. Moreover, only non-competitive NMDA-receptor blockers showed such property [79]. Brain-derived neurotrophic factor (BDNF) is a neurotrophin, which also enhances hippocampal synaptic transmission by increasing NMDA receptor activity [80]. Memantine increases the levels of BDNF mRNA in the limbic cortex and this effect increases with increase in dose. Effects of memantine on BDNF mRNA result in increased production of BDNF. At the same time, memantine induces isoforms of the BDNF receptor trkB. Both these factors may also mediate the neuroprotective and memory enhancing effect of memantine [81]. The N-methyl-d-aspartate (NMDA) receptors are involved in long-term potentiation (LTP) and are phosphorylated by several tyrosine kinases including Src-family tyrosine kinases Fyn. Fyn links the BDNF receptor trkB with NMDA receptors, which play an important role in spatial memory formation [82]. BDNF activates hippocampal extracellular signal-regulated kinase (ERK) and Ras-signalling, both of which are responsible for synaptic plasticity in hippocampus [83]. Calcium ions inside the cells have been found to increase BDNF expression [84]. Other actions of memantine include non-competitive, voltage-independent inhibition of 5HT3 receptor currents with an IC50 value of 2 m by inducing receptor desensitization [85]. Memantine was found to reduce the peak amplitude and duration of endplate current in frog sartorious muscle preparation and it was postulated that it reacts with ACh receptor ion channel complex (IC50 value of 10 m) in open and closed state and inhibits neuromuscular transmission [86]. It has also been shown to block human neuronal nicotinic cholinergic receptors (IC50 = 6.6 m) [87]. Redox-modulatory sites consisting of cystein residues are present on NMDA-receptor ion channel complex. These sites act in a manner similar to memantine by blocking NMDA-receptor ion channel when it is open for an excessively longer period (pathological activation). Redox-related forms of NO act with these cystein residues by a reaction called S-nitrosylation and this reaction protects neurons against excitotoxic damage. [88,89]. Nitroglycerin supplies NO group in the form of nitrosonium ion (NO+ ), which acts with critical sulphydryl groups at the redox modulatory site and facilitates the formation of disulphide bonds. This reduces the activity of NMDA-receptor ion channel. Long-term use of nitroglycerin produces tolerance to its effects on cardiovascular system, but not to its effect on NMDA-receptor-mediated neurotoxicity in brain [90]. Clinically safer NMDA antagonists such as memantine and
9
nitroglycerin and the combination drug nitro-memantine are promising as drugs in treating neurodegenerative diseases [91]. There are several other agents acting on glutamate system, apart from memantine, in the pipeline. Sabeluzole is one such agent known to block glutamate-induced calcium influx and to stabilize neuronal cytoskeleton [22]. Taurine also protects against glutamate excitotoxicity by stabilizing calcium homeostasis and energy metabolism [92]. Dimebon, an antihistaminic agent, shows dual activity, as an NMDA antagonist (ED50 = 42 mg kg−1 , i.p.) and an AChE and BuChE inhibitor (IC50 values of 42 and 7.9 M, respectively). 3.2. Memantine in neurological disorders other than dementia Besides dementias, memantine can be an effective alternative for treatment of numerous CNS disorders that include stroke, CNS trauma, Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), epilepsy, drug dependence, glaucoma and chronic pain [55]. Glaucoma is a neurodegenerative disease showing progressive loss of retinal ganglion cells (RGCs). It is a leading cause of blindness and second leading cause of irreversible blindness worldwide [93]. Safety and efficacy of memantine in glaucoma is being tested in clinical trials. Mild excitotoxicity is one of the factors contributing to RGC death. This type of excitotoxic cell death is, in part, due to overactivation of N-methyl-d-aspartate (NMDA)-type glutamate receptors. In glaucoma, glutamate levels are increased in the vitreous humour [94]. Memantine may be beneficial as it acts by blocking excessive activity of NMDA receptors without interfering with physiological activity [95,96]. NMDA-receptor excitotoxicity contributes towards neurodegeneration of Parkinson’s disease. Memantine shows antiparkinsonian activity in animal models and patients of Parkinson’s disease [97]. It also prevents MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced neuronal excitotoxicity and death [98]. Neuroprotective activity of memantine has also been seen in neonatal as well as adult stroke models [91,99]. Since there is evidence that NMDA-excitotoxicity may play an important role in epilepsy, NMDA-receptor antagonists have become the compounds of interest. Memantine might be a lead for therapeutically promising compounds for symptomatic treatment of epilepsy [53,100]. Memantine was found to be effective and safe in amyotrophic sclerosis as well [59,101], suggesting NMDA receptors as a target for future treatment of multiple sclerosis. Drug dependence on exposure to opiates relates to neuronal plasticity and NMDA receptors are implicated in this. Memantine administration prevented the development of morphine dependence in rats. Memantine has also been found to be effective in the treatment of addiction [102–104]. Memantine showed antinociceptive effect in experimental models of inflammatory and neuropathic pain. It prevented carageenan-induced mechanical
10
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
Table 4 Memantine—preclinical evidence Experimental model
Parameter
Effect of memantine
Reference
Adult (9–12 months) male F-344 rats
(a) LTP or field excitatory post-synaptic potential (f-EPSP) (b) Morris water maze task
[109]
Injection of antigen or lipoplysaccharides in NBM NMDA-receptor overactivation by an agonist or reduced Mg2+ concentration Systemic NMDA-administration (25 mg kg−1 ) Incubation of hippocampal slices with NMDA (10 M) Lesioning of entorhinal cortex in rats by quinolinic acid AF64A-induced lesions to cholinergic neurons A-injection in hippocampal fissure
Inflammation, neuronal death
Increased maintenance of LTP of f-EPSP at 30 mg kg−1 Increased tendency towards selective spatial search 1 M memantine provided neuroprotection Attenuated NMDA-induced deficit in passive avoidance and LTP Antagonized the impairment produced by NMDA Antagonized NMDA-induced decrease in expression of LTP Decreased lesion-induced impairment
Incubation of rat cortical cell culture with human HIV type 1 coat protein gp120
Passive avoidance in vivo and LTP in CA1 in vitro LTP in vitro and passive avoidance LTP expression, AMPA-mediated field potential in CA1 Radial Arm Maze performance Active avoidance and Morris water maze Neuronal death, astrocytic and microglial activation, spatial discrimination DNA fragmentation, percentage of viable cells
and thermal analgesia and reduced nociception during late phase of formalin test [105,106]. Memantine exhibited an antinociceptive effect in diabetic neuropathy in rats [107]. 3.3. Memantine in experimental models for Alzheimer’s disease and dementia—preclinical evidence Memantine inhibits binding of (3 H)MK-801 to both rat and human cortical neuronal membranes with an affinity that is less than MK-801. Unblocking kinetics of memantine (Koff =0.2 s−1 ) is 40 times faster than that of MK-801 (Koff = 0.005 s−1 ) [108]. Memantine improves memory and spatial learning in aged rats and increases the expression of hippocampal LTP. Memantine-treated rats showed more selective spatial search patterns in the training quadrant of the Morris water maze. Electrophysiological studies have shown the ability of memantine to increase the durability of synaptic plasticity in moderately aged rats and improved performance in Morris water maze task [109,110]. Incubation of rat hippocampal slices with exogenous NMDA decreased AMPA-receptor-mediated currents and impaired LTP in CA1 region. This latter effect was prevented by co-incubation with memantine but not by MK-801 [111] indicating a basic difference in mode of action. It also improves reference memory based learning in rats with lesions in entorhinal cortex, the area that is affected in early stages of AD [112]. Memantine cured the memory impairment caused by AF64A-induced lesion of cholinergic system [113]. Memantine protected neurons from glutamate-induced neurotoxicity (EC50 of 1.1–1.4 m) [60] and reduced A-induced apoptotic death and neuroinflammation in the hippocampus [114]. It also exerted antiapoptotic effects against DNA fragmentation induced by the HIV type I coat
Attenuated the deficit in memory due to the lesion Protected against A-induced neuronal death Prevented apoptosis caused by gp120
[110] [110] [111] [111] [112] [113] [114] [115]
protein gp 120 [115]. Apoptosis induced by gp120 may contribute to the neurological complications associated with the immunodeficiency syndrome. The cytoprotective effect of memantine as observed in cortical cell cultures may help in the treatment of AIDS-related dementia. There was no evidence of carcinogenicity, genotoxicity and impairment of fertility with memantine in animal toxicity studies. Table 4 summarizes the preclinical findings with memantine. 4. Clinical studies with memantine 4.1. Clinical trials of memantine in dementia patients The efficacy of oral memantine has been evaluated in three recently concluded short-term random clinical trials (RCT) [116] and one post-marketing surveillance (PMS) [117] study in patients of age 60–80 years already diagnosed with moderate to severe AD (Table 5). One RCT and one PMS showed improved global symptoms and functional task performance in patients whereas the other RCT showed reduced deterioration of global functions in memantine-treated patients. Three recent trials of memantine have already been completed—one in Latvia, one US phase III trial testing memantine as a single drug therapy, and another phase III trial testing memantine in combination with donepezil (Aricept® ), a cholinesterase inhibitor marketed by Eisai, Inc. and Pfizer, Inc. Data from the combination therapy trial suggests that individuals who received both memantine and donepezil performed better in terms of their thinking skills and ability to perform daily activities than those who took donepezil and a placebo. Combination therapy of memantine with donepezil was well tolerated (no adverse drug reactions reported), safe and effective [118]
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
11
Table 5 Clinical trials on memantine About the trial
Number and type of patients
Dose (mg per day)
Duration
Assessment
Reference
Randomized, double blind, placebo-controlled Double-blind, randomized, parallel-group study Post-marketing surveillance, combination therapy (memantine + donepezil) Double-blind, placebo-controlled Double-blind, placebo-controlled, parallel-group, randomized, unconfounded Double-blind, parallel group, randomized placebo controlled Randomised, double blind, placebo-controlled Placebo-controlled, multi-centred Post-marketing surveillance Randomized, two-centred, placebo-controlled Randomised, double blind, placebo controlled
Outpatients with moderate to severe AD patients, n = 252 Moderate to severe AD or VaD patients, n = 166 Moderate to severe AD out-patients, n = 158
20
28 weeks
ADCS-ADL, CIBIC-plus
[121]
10
12 weeks
BGP, CGI-C
[116]
20
16 weeks
SIB, ADCS-ADL, CIBIC-plus
[118]
20
28 weeks
[123]
20
28 weeks
ADAS-Cog, CGI-C ADL, CGI-C, CIBIC-plus
20
28 weeks
CGI-C, MMSE, ADCS-ADL
[58]
20
28 weeks
CGI-C, BGP
[122]
20
28 weeks
[57]
20
6 weeks
ADAS-Cog, CIBIC-plus CGI-C, ADL
[117]
10 from D1 to D3, 20 from D4
42 days
SCAG, GBS, DL
[151]
Starting dose 5 mg per day, increase to 20 mg per day
24 weeks
SIB, ADCS-ADL, CIBIC-plus, BGP
[120]
Patients with mild-to moderate VaD, n = 900 Moderate to severe AD or VaD
Patients with Wernicke–Korsakoff syndrome, n = 16 Out-patients with moderate to severe AD Patients with VaD, n = 321 Patients with advanced dementia, n = 531 Patients with mild to moderate dementia, n = 88 Patients with moderate to severe AD, n = 404
[3]
ADCS: Alzheimer’s Disease Cooperative Study; ADL: activities of daily living; CIBIC-plus: clinician’s interview based impression of change; BGP: Beurteilungsskala fur Geriatrische Patienten, rating scale for geriatric patients; CGI-C: clinical global impression of change; SIB: severe impairment battery; ADAS-Cog: Alzheimer’s Disease Assessment Scale—Cognitive subscale; MMSE: Mini-Mental Status Examination; SCAG: total sum Scores of Clinical Assessment of Geriatric scale; GBS: Gottfries–Brane–Steen scale; D1: day 1.
and there was no drug–drug interaction between these two [119]. The results from a study in patients with moderate to severe dementia already receiving stable treatment with donepezil, memantine resulted in significantly better outcomes than placebo on measures of cognition, activities of daily living, global outcome and behavior and was well tolerated [120]. Whether effects of memantine in combination with cholinesterase inhibitors are complementary or synergistic needs to be determined [121]. A placebo-controlled study of memantine in dementia of Wernicke–Korsakoff syndrome in Vienna, Austria showed significant and clinically relevant benefit for memantine-treated group as compared to placebo-treated group as demonstrated by results of MMSE, CGI and ADCS-ADL [58]. In a 28-week, randomized, double-blind, placebo-controlled, multicentred study at Auckland, New Zealand, memantine at 20 mg per day slowed deterioration in patients with moderate to severe AD [122]. Another study of similar kind reported slowing of deterioration with memantine at 20 mg per day in patients
with moderate to severe AD, the variables studied were CIBIC-plus, ADCS-ADL [121]. In a 28-week multicentred study in UK, memantine (20 mg per day) was found to improve cognition relative to placebo in vascular dementia and was well tolerated [123]. In all these trials, memantine was found safe and well tolerated. The tolerability of an NMDA antagonist depends upon its affinity towards the receptors, unbinding kinetics and voltage dependency. Multiple clinical trials using NMDA receptor antagonists (phencyclidine, MK-801) have been stopped mainly due to the severe psychomimetic adverse effects. 4.2. Adverse effects of memantine observed in clinical studies Memantine has shown acceptable safety and tolerability in 2297 patients and 27 clinical trials. In contrast to high-affinity NMDA antagonists, side effects associated
12
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
with the use of memantine are of mild intensity and include hallucinations, confusion, dizziness, headache and tiredness. Binding of dizocilpine to phencyclidine-binding site of NMDA receptors (MK-801) is associated with severe psychomimetic effects of this compound. Memantine also binds to the same site at NMDA receptors. This is the reason why many physicians are cautious while using memantine for patients of AD. In clinical trials of memantine in moderate to severe dementia, commonly seen adverse reactions as compared to placebo-treated group were (1–10% and more frequent than placebo) hallucinations (2.0% versus 0.7%), dizziness (1.7% versus 1.0%), headache (1.7% versus 1.4%), confusion (1.3% versus 0.3%) and tiredness (1.0% versus 0.3%). Other adverse effects seen during clinical trials include (as reported in Approval Labeling Text for Namenda® ) syncope, cardiac failure, vertigo, transient ischemic attacks, ataxia, hypokinesia, anaemia, increased alkaline phosphatase activity, skin rash and frequent micturition.
5. Pharmacokinetics of memantine Memantine HCl is available as a 10 mg tablet and as a 10 mg mL−1 oral solution. The maximum daily dose is 20 mg. To reduce the risk of side effects, the maintenance dose is achieved by an upward titration of 5 mg per week over first three weeks. From fourth week onwards, the treatment can be continued with maintenance dose of 20 mg per day (Lundbeck Ltd., Ebixa, Summary of Product Characteristics, 2002). In patients with moderate renal impairment, the dose can be reduced to 10 mg per day. At current prices, treatment with memantine for a year at 20 mg per day costs £890. 5.1. Animal pharmacokinetics Regional binding characteristics of memantine and other NMDA antagonists have been studied using quantitative receptor autoradiography. Drugs that are well-tolerated showed higher affinity in cerebellar region than in forebrain whereas the drugs known to cause neurobehavioral or psychomimetic side effects showed higher affinity in forebrain. This also showed that cerebellar NMDA-receptor ion channels are different from those in forebrain [124]. In a study in rats, it was shown that same dose of memantine results into about two fold higher serum levels of memantine in female rats as compared to male rats [125]. It has been reported that memantine does not have any effect on dopamine release from prefrontal cortex and does not have any psychotomimetic effects [126]. 5.2. Human pharmacokinetics Memantine crosses blood–brain barrier rapidly and within 30 min of 20 mg intravenous infusion, the drug can be de-
tected in cerebrospinal fluid (CSF). At a dose of 20 mg per day, the CSF levels match the inhibition constant of memantine at the phencyclidine-binding site of NMDA receptors, which is 0.5 m in human frontal cortex [127]. CSF level attained by memantine was lower than serum level in each patient and a mean CSF/serum ratio of 0.52 has been reported [128]. The drug shows linear pharmacokinetics in the dose range of 5–40 mg. It undergoes little hepatic metabolism, so clearance of memantine may not be affected in patients with hepatic impairment. Memantine does not inhibit CYP450 isoenzymes in vitro. In humans, major metabolites are N-3,5-dimethyl gludantan which is an isomeric mixture of 4- and 6-hydroxy memantine and 1-nitroso-3,5-dimethyl adamantine, none of which is NMDA antagonist [124]. Memantine is 100% bioavailable after an oral dose and shows a tmax of 3–8 h. There are no reports showing effect of food, sex and age on absorption of the drug. It shows minimal metabolism and a terminal elimination half-life of 60–80 h. In patients with normal kidney function, total clearance (Cltot ) is around 170 mL/min/1.73 m2 . A daily dose of 20 mg takes plasma steady-state concentration to 70–150 ng mL−1 [104]. No difference was observed in steady-state plasma concentrations of memantine between healthy subjects and patients with dementia. About 45% of the drug in plasma is bound to plasma proteins [129] and volume of distribution is 10 L kg−1 . About 75% or more of the drug is excreted unchanged via urine. Acidification of urine increases the renal clearance of memantine but urine flow does not affect its renal clearance significantly [130]. In case of overdosage, excretion of memantine can be hastened by acidification of urine. 5.3. Therapeutic index Memantine has a high therapeutic index as it inhibits NMDA-receptor-mediated currents with an IC50 of 3 M whereas the expression of LTP is inhibited at a much higher concentration with an IC50 of 11.6 M. This is in contrast with MK-801 that inhibits both, NMDA-receptor currents as well as LTP with an IC50 of 0.14 M [131,132]. 5.4. Interactions with other drugs Memantine has the potential to interact with a number of drugs. It releases dopamine in a dose-dependent manner [133,134] and hence enhances the actions of l-DOPA, dopaminergic agonists and anticholinergics. A study has shown that memantine enhances anti-Parkinsonism activity of l-DOPA when the two drugs are co-administered in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)treated mice [135]. NMDA-receptor-mediated events are involved in MPTP-induced neurotoxicity. This is another mechanism by which memantine may be helpful in Parkinsonism [98]. At the same time, memantine suppresses dopamine decarboxylase and this may render l-DOPA less
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
effective for Parkinsonism treatment [136]. It reduces the activity of neuroleptics and barbiturates [137]. Memantine along with other low-affinity NMDA antagonists enhance anti-nociceptive activity of morphine recorded from the tail in rats [138]. It may restore the analgesic activity of morphine in acutely tolerant rats [139]. A role for excitatory amino acids has been implicated in epilepsy. Memantine and similar NMDA antagonists potentiate the activity of conventional antiepileptics and this combination has a potential clinical importance [98]. Memantine blocks 5-HT3 receptor currents and this activity may be responsible for clinical depression seen with memantine [85]. Concomitant administration of memantine and amantadine or ketamine should be avoided as all are chemically related NMDA antagonists, affect dopaminergic system [140] and may compete for the probenecid-sensitive organic cation transporters in renal tubules. Drugs like cimetidine, ranitidine, procainamide, quinidine, and nicotine use same cationic renal transport system [141] as amantadine and may interact with memantine increasing its plasma concentration. Drugs that increase urinary pH may result in accumulation of memantine. No clinical data regarding safety in pregnancy are available for memantine but the drug has shown a potential for reducing intrauterine growth in animals (Lundbeck Ltd., Ebixa, Summary of Product Characteristics, 2002). Hence, it is better to avoid use of the drug during pregnancy unless very necessary. It is not known whether memantine is excreted in breast milk but being a lipophillic compound, it may be excreted. Hence, women taking memantine should not breast feed. 5.5. Conclusion Discovery of memantine has shown for the first time that NMDA antagonists can also be used therapeutically in AD and other neurodegenerative diseases without causing the adverse effects of NMDA antagonists discovered earlier, in the therapeutic dose range. Memantine is a well-tolerated, moderate affinity, non-competitive NMDA-receptor antagonist. It blocks the neurotoxicity of glutamate, but not its physiological functions. The drug exerts its therapeutic effect in various conditions mainly by blocking glutamate excitotoxicity, although it has antioxidant property and also increases production of BDNF. As far as AD is concerned, once diagnosed, further neurodegeneration can be prevented by memantine treatment. At the same time, it needs to be emphasized that memantine is just a disease modifying agent and cannot revert the process of neurodegeneration. In short, though this drug is way ahead of other approved drugs for treatment of AD, in no way it is a solution to our search for cure to AD. Memantine will enjoy exclusivity for an initial period of 1–2 years. Later on, it may face competition from the emerging agents that have potential to modify underlying pathology of AD and are already under clinical trials.
13
References [1] Filley CM. Alzheimer’s disease: it’s irreversible but not untreatable. Geriatrics 1995;50:18–23. [2] Khachaturian ZS. Diagnosis of Alzheimer’s disease. Arch Neurol 1985;42:1097–105. [3] Areosa SA, Sherriff F. Memantine for dementia. Cochrane Database Syst Rev 2003;CD003154. [4] Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev 2001;81:741–66. [5] Suh YH, Checler F. Amyloid precursor protein, presenilins, and alpha-synuclein: molecular pathogenesis and pharmacological applications in Alzheimer’s disease. Pharmacol Rev 2002;54:469–525. [6] Wenk GL, Danysz W, Roice DD. The effects of mitochondrial failure upon cholinergic toxicity in the nucleus basalis. Neuroreport 1996;7:1453–6. [7] Scorer CA. Preclinical and clinical challenges in the development of disease-modifying therapies for Alzheimer’s disease. Drug Discov Today 2001;6:1207–19. [8] Hake AM. Use of cholinesterase inhibitors for treatment of Alzheimer’s disease. Cleveland Clin J Med 2001;68:608–16. [9] Cocabelos R, Nordberg A, Caamano J, Franco-Maside A, Fernandez-Novoa L, Gomez MJ, et al. Molecular strategies for the first generation of antidementia drugs(I). Tacrine and related compounds. Drugs Today 1994;30:259–337. [10] Bryson HM, Benfield P. Donepezil Drugs Aging 1997;10:234–9 [discussion 231–40]. [11] Scott LJ, Goa KL. Galantamine: a review of its use in Alzheimer’s disease. Drugs 2000;60:1095–122. [12] Imbimbo BP, Martelli P, Troetel WM, Lucchelli F, Lucca U, Thal LJ. Efficacy and safety of eptastigmine for the treatment of patients with Alzheimer’s disease. Neurology 1999;52:700–8. [13] Bai DL, Tang XC, He XC, Huperzine A. a potential therapeutic agent for treatment of Alzheimer’s disease. Curr Med Chem 2000;7:355–74. [14] Toda N, Tago K, Marumoto S, Takami K, Ori M, Yamada N, et al. A conformational restriction approach to the development of dual inhibitors of acetylcholinesterase and serotonin transporter as potential agents for Alzheimer’s disease. Bioorg Med Chem 2003;11:4389–415. [15] Clausen T, Bullock R. Medical treatment and neuroprotection in traumatic brain injury. Curr Pharm Des 2001;7:1517–32. [16] Helmuth L. New therapies. New Alzheimer’s treatments that may ease the mind. Science 2002;297:1260–2. [17] Scarpini E, Scheltens P, Feldman H. Treatment of Alzheimer’s disease: current status and new perspectives. Lancet Neurol 2003;2:539–47. [18] Doerfler P, Shearman MS, Perlmutter RM. Presenilin-dependent gamma-secretase activity modulates thymocyte development. Proc Natl Acad Sci USA 2001;98:9312–7. [19] Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard NP, et al. Metabolic regulation of brain Abeta by neprilysin. Science 2001;292:1550–2. [20] Brown M. Gene therapy success for Alzheimer’s? Drug Discov Today 2003;8:474–5. [21] Ames BN, Shigenaga MK, Hagen TM. Oxidants antioxidants and the degenerative diseases of ageing. Proc Natl Acad Sci USA 1993;90:7915–22. [22] Bachurin SO. Medicinal chemistry approaches for the treatment and prevention of Alzheimer’s disease. Med Res Rev 2003;23:48–88. [23] McGeer EG, McGeer PL. Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry 2003;27: 741–9. [24] Dominguez DI, De Strooper B. Novel therapeutic strategies provide the real test for the amyloid hypothesis of Alzheimer’s disease. Trends Pharmacol Sci 2002;23:324–30.
14
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
[25] Breitner JC. NSAIDs Alzheimer’s disease: how far to generalise from trials? Lancet Neurol 2003;2:527. [26] Kojro E, Gimpl G, Lammich S, Marz W, Fahrenholz F. Low cholesterol stimulates the non-amyloidogenic pathway by its effect on alpha-secretase ADAM 10. Proc Natl Acad Sci USA 2001;98: 5815–20. [27] Simons M, Keller P, De Strooper B, Beyreuther K, Dotti CG, Simons K. Cholesterol depletion inhibits the generation of beta-amyloid in hippocampal neurons. Proc Natl Acad Sci USA 1998;95:6460–4. [28] Fassbender K, Simons M, Bergmann C, Stroick M, Lutjohann D, Keller P, et al. Simvastatin strongly reduces levels of Alzheimer’s disease beta-amyloid peptides Abeta 42 and Abeta 40 in vitro and in vivo. Proc Natl Acad Sci USA 2001;98:5856–61. [29] Kwak B, Mulhaupt F, Myit S, Mach F. Statins as a newly recognized type of immunomodulator. Nat Med 2000;6:1399–402. [30] Hrncic R, Wall J, Wolfenbarger DA, Murphy CL, Schell M, Weiss DT, et al. Antibody-mediated resolution of light chain-associated amyloid deposits. Am J Pathol 2000;157:1239–46. [31] Schenk D. Amyloid-beta immunotherapy for Alzheimer’s disease: the end of the beginning. Nat Rev Neurosci 2002;3:824–8. [32] Hirschfield GM, Hawkins PN. Amyloidosis: new strategies for treatment. Int J Biochem Cell Biol 2003;35:1608–13. [33] McGeer PL, McGeer E. Is there a future for vaccination as a treatment for Alzheimer’s disease? Neurobiol Aging 2003;24: 391–5. [34] Cherny RA, Legg JT, McLean CA, Fairlie DP, Huang X, Atwood CS, et al. Aqueous dissolution of Alzheimer’s disease Abeta amyloid deposits by biometal depletion. J Biol Chem 1999;274:23223–8. [35] Bush AI. The metallobiology of Alzheimer’s disease. Trends Neurosci 2003;26:207–14. [36] Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JD, Hanson GR, et al. Cu(II) potentiation of alzheimer abeta neurotoxicity. Correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem 1999;274:37111–6. [37] Kordower JH, Isacson O, Emerich DF. Cellular delivery of trophic factors for the treatment of Huntington’s disease: is neuroprotection possible? Exp Neurol 1999;159:4–20. [38] Mandelkow E. Alzheimer’s disease The tangled tale of tau. Nature 1999;402:588–9. [39] Sherwin BB. Estrogen and cognitive aging in women. Trends Pharmacol Sci 2002;23:527–34. [40] Witt A, Macdonald N, Kirkpatrick P. Memantine hydrochloride. Nat Rev Drug Discov 2004;3:109–10. [41] Wenk GL. Neuropathologic changes in Alzheimer’s disease. J Clin Psychiatry 2003;64(Suppl 9):7–10. [42] Kornhuber J, Weller M. Psychotogenicity and N-methyl-d-aspartate receptor antagonism: implications for neuroprotective pharmacotherapy. Biol Psychiatry 1997;41:135–44. [43] Doraiswamy PM. Alzheimer’s disease and the glutamate NMDA receptor. Psychopharmacol Bull 2003;37:41–9. [44] Meldrum BS. Glutamate as a neurotransmitter in the brain: review of physiology and pathology. J Nutr 2000;130:1007S–15S. [45] Kemp JA, McKernan RM. NMDA receptor pathways as drug targets. Nat Neurosci 2002;5(Suppl):1039–42. [46] Rogawski MA, Wenk GL. The neuropharmacological basis for the use of memantine in the treatment of Alzheimer’s disease. CNS Drug Rev 2003;9:275–308. [47] Naish HJ, Marsh WL, Davies JA. Effect of low-affinity NMDA receptor antagonists on electrical activity in mouse cortical slices. Eur J Pharmacol 2002;443:79–83. [48] Calzada JI, Jones BE, Netland PA, Johnson DA. Glutamate-induced excitotoxicity in retina: neuroprotection with receptor antagonist, dextromethorphan, but not with calcium channel blockers. Neurochem Res 2002;27:79–88. [49] Carlsson KC, Hoem NO, Moberg ER, Mathisen LC. Analgesic effect of dextromethorphan in neuropathic pain. Acta Anaesthesiol Scand 2004;48:328–36.
[50] Weinbroum AA, Ben-Abraham R. Dextromethorphan and dexmedetomidine: new agents for the control of perioperative pain. Eur J Surg 2001;167:563–9. [51] Palmer GC. Neuroprotection by NMDA receptor antagonists in a variety of neuropathologies. Curr Drug Targets 2001;2:241–71. [52] Hoyer S, Riederer P. Pathomechanisms and hypothesis-guided therapeutic strategies for late-onset Alzheimer’s disease. Fortschr Neurol Psychiatry 2003;71(Suppl 1):S16–26. [53] Molinuevo JL. Memantine. Neurologia 2003;18:255–61. [54] Wilcock GK. Memantine for the treatment of dementia. Lancet Neurol 2003;2:503–5. [55] Kilpatrick GJ, Tilbrook GS. Memantine. Merz Curr Opin Investig Drugs 2002;3:798–806. [56] Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine in probable vascular dementia and Alzheimer’s disease combined with cerebrovascular disease: a randomised trial. Lancet 2002;359:1283–90. [57] Orgogozo JM, Rigaud AS, Stoffler A, Mobius HJ, Forette F. Efficacy and safety of memantine in patients with mild to moderate vascular dementia: a randomized, placebo-controlled trial (MMM 300). Stroke 2002;33:1834–9. [58] Rustembegovic A, Kundurovic Z, Sapcanin A, Sofic E. A placebo-controlled study of memantine (Ebixa) in dementia of Wernicke–Korsakoff syndrome. Med Arh 2003;57:149–50. [59] Jain KK. Evaluation of memantine for neuroprotection in dementia. Expert Opin Investig Drugs 2000;9:1397–406. [60] Parsons CG, Danysz W, Bartmann A, Spielmanns P, Frankiewicz T, Hesselink M, et al. Amino-alkyl-cyclohexanes are novel uncompetitive NMDA receptor antagonists with strong voltage-dependency and fast blocking kinetics: in vitro and in vivo characterization. Neuropharmacology 1999;38:85–108. [61] Sobolevsky A, Koshelev S. Two blocking sites of amino-adamantane derivatives in open N-methyl-d-aspartate channels. Biophys J 1998;74:1305–19. [62] Headley PM, Grillner S. Excitatory amino acids and synaptic transmission: the evidence for a physiological function. Trends Pharmacol Sci 1990;11:205–11. [63] McBain CJ, Mayer ML. N-Methyl-d-aspartic acid receptor structure and function. Physiol Rev 1994;74:723–60. [64] Das S, Sasaki YF, Rothe T, Premkumar LS, Takasu M, Crandall JE, et al. Increased NMDA current and spine density in mice lacking the NMDA receptor subunit NR3A. Nature 1998;393:377–81. [65] Nowak L, Bregestovski P, Ascher P, Herbet A, Prochiantz A. Magnesium gates glutamate-activated channels in mouse central neurones. Nature 1984;307:462–5. [66] Clements JD, Lester RA, Tong G, Jahr CE, Westbrook GL. The time course of glutamate in the synaptic cleft. Science 1992;258:1498– 501. [67] Beal MF. Does impairment of energy metabolism result in excitotoxic neuronal death in neurodegenerative illnesses? Ann Neurol 1992;31:119–30. [68] Nicholls DG, Ward MW. Mitochondrial membrane potential and neuronal glutamate excitotoxicity: mortality and millivolts. Trends Neurosci 2000;23:166–74. [69] Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309–12. [70] Molecule of the month. Memantine hydrochloride. Drug News Perspect 2003;16:5. [71] Meldrum B, Garthwaite J. Excitatory amino acid neurotoxicity and neurodegenerative disease. Trends Pharmacol Sci 1990;11:379–87. [72] Harkany T, Hortobagyi T, Sasvari M, Konya C, Penke B, Luiten PG, et al. Neuroprotective approaches in experimental models of beta-amyloid neurotoxicity: relevance to Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry 1999;23:963–1008. [73] Greenamyre JT, Young AB. Excitatory amino acids and Alzheimer’s disease. Neurobiol Aging 1989;10:593–602.
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17 [74] Harris ME, Wang Y, Pedigo Jr NW, Hensley K, Butterfield DA, Carney JM. Amyloid beta peptide (25-35) inhibits Na+-dependent glutamate uptake in rat hippocampal astrocyte cultures. J Neurochem 1996;67:277–86. [75] Couratier P, Lesort M, Sindou P, Esclaire F, Yardin C, Hugon J. Modifications of neuronal phosphorylated tau immunoreactivity induced by NMDA toxicity. Mol Chem Neuropathol 1996;27:259– 73. [76] Arias C, Arrieta I, Tapia R. Beta-amyloid peptide fragment 25-35 potentiates the calcium-dependent release of excitatory amino acids from depolarized hippocampal slices. J Neurosci Res 1995;41: 561–6. [77] Lipton SA, Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 1994;330:613–22. [78] Doraiswamy PM. Non-cholinergic strategies for treating and preventing Alzheimer’s disease. CNS Drugs 2002;16:811–24. [79] Lupp A, Kerst S, Karge E. Evaluation of possible pro- or antioxidative properties and of the interaction capacity with the microsomal cytochrome P450 system of different NMDA-receptor ligands and of taurine in vitro. Exp Toxicol Pathol 2003;54:441–8. [80] Lu B. BDNF and activity-dependent synaptic modulation. Learn Mem 2003;10:86–98. [81] Marvanova M, Lakso M, Pirhonen J, Nawa H, Wong G, Castren E. The neuroprotective agent memantine induces brain-derived neurotrophic factor and trkB receptor expression in rat brain. Mol Cell Neurosci 2001;18:247–58. [82] Mizuno M, Yamada K, He J, Nakajima A, Nabeshima T. Involvement of BDNF receptor TrkB in spatial memory formation. Learn Mem 2003;10:108–15. [83] Sweatt JD. The neuronal MAP kinase cascade: a biochemical signal integration system subserving synaptic plasticity and memory. J Neurochem 2001;76:1–10. [84] Orban PC, Chapman PF, Brambilla R. Is the Ras-MAPK signalling pathway necessary for long-term memory formation? Trends Neurosci 1999;22:38–44. [85] Rammes G, Rupprecht R, Ferrari U, Zieglgansberger W, Parsons CG. The N-methyl-d-aspartate receptor channel blockers memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes antagonise 5-HT(3) receptor currents in cultured HEK-293 and N1E-115 cell systems in a non-competitive manner. Neurosci Lett 2001;306:81–4. [86] Masuo K, Enomoto K, Maeno T. Effects of memantine on the frog neuromuscular junction. Eur J Pharmacol 1986;130:187–95. [87] Buisson B, Bertrand D. Open-channel blockers at the human alpha4beta2 neuronal nicotinic acetylcholine receptor. Mol Pharmacol 1998;53:555–63. [88] Kim WK, Choi YB, Rayudu PV, Das P, Asaad W, Arnelle DR, et al. Attenuation of NMDA receptor activity and neurotoxicity by nitroxyl anion, NO. Neuron 1999;24:461–9. [89] Choi YB, Tenneti L, Le DA, Ortiz J, Bai G, Chen HS, et al. Molecular basis of NMDA receptor-coupled ion channel modulation by S-nitrosylation. Nat Neurosci 2000;3:15–21. [90] Lipton SA, Gendelman HE. Seminars in medicine of the Beth Israel Hospital, Boston. Dementia associated with the acquired immunodeficiency syndrome. N Engl J Med 1995;332:934–40. [91] Le DA, Lipton SA. Potential and current use of N-methyl-daspartate (NMDA) receptor antagonists in diseases of aging. Drugs Aging 2001;18:717–24. [92] El Idrissi A, Trenkner E. Growth factors and taurine protect against excitotoxicity by stabilizing calcium homeostasis and energy metabolism. J Neurosci 1999;19:9459–68. [93] Dreyer EB, Grosskreutz CL. Excitatory mechanisms in retinal ganglion cell death in primary open angle glaucoma (POAG). Clin Neurosci 1997;4:270–3. [94] Naskar R, Vorwerk CK, Dreyer EB. Saving the nerve from glaucoma: memantine to caspaces. Semin Ophthalmol 1999;14: 152–8.
15
[95] Lipton SA. Possible role for memantine in protecting retinal ganglion cells from glaucomatous damage. Surv Ophthalmol 2003;48(Suppl 1):S38–46. [96] Hare W, WoldeMussie E, Lai R, Ton H, Ruiz G, Feldmann B, et al. Efficacy and safety of memantine, an NMDA-type open-channel blocker, for reduction of retinal injury associated with experimental glaucoma in rat and monkey. Surv Ophthalmol 2001;45(Suppl 3):S284–9 [discussion S286–95]. [97] Danysz W, Parsons CG, Kornhuber J, Schmidt WJ, Quack G. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents—preclinical studies. Neurosci Biobehav Rev 1997;21:455–68. [98] Lange KW, Kornhuber J, Riederer P. Dopamine/glutamate interactions in Parkinson’s disease. Neurosci Biobehav Rev 1997;21:393– 400. [99] Stieg PE, Sathi S, Warach S, Le DA, Lipton SA. Neuroprotection by the NMDA receptor-associated open-channel blocker memantine in a photothrombotic model of cerebral focal ischemia in neonatal rat. Eur J Pharmacol 1999;375:115–20. [100] Kohl BK, Dannhardt G. The NMDA receptor complex: a promising target for novel antiepileptic strategies. Curr Med Chem 2001;8:1275–89. [101] Paul C, Bolton C. Modulation of blood–brain barrier dysfunction and neurological deficits during acute experimental allergic encephalomyelitis by the N-methyl-d-aspartate receptor antagonist memantine. J Pharmacol Exp Ther 2002;302:50–7. [102] Maldonado C, Cauli O, Rodriguez-Arias M, Aguilar MA, Minarro J. Memantine presents different effects from MK-801 in motivational and physical signs of morphine withdrawal. Behav Brain Res 2003;144:25–35. [103] Levi MS, Borne RF. A review of chemical agents in the pharmacotherapy of addiction. Curr Med Chem 2002;9:1807–18. [104] Parsons CG, Danysz W, Quack G. Memantine is a clinically well tolerated N-methyl-d-aspartate (NMDA) receptor antagonist—a review of preclinical data. Neuropharmacology 1999;38:735–67. [105] Berrino L, Oliva P, Massimo F, Aurilio C, Maione S, Grella A, et al. Antinociceptive effect in mice of intraperitoneal N-methyl-d-aspartate receptor antagonists in the formalin test. Eur J Pain 2003;7:131–7. [106] Smith PF. Therapeutic N-methyl-d-aspartate receptor antagonists: will reality meet expectation? Curr Opin Investig Drugs 2003;4:826– 32. [107] Villetti G, Bergamaschi M, Bassani F, Bolzoni PT, Maiorino M, Pietra C, et al. Antinociceptive activity of the N-methyl-daspartate receptor antagonist N-(2-indanyl)-glycinamide hydrochloride (CHF3381) in experimental models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 2003;306:804–14. [108] Parsons CG, Quack G, Bresink I, Baran L, Przegalinski E, Kostowski W, et al. Comparison of the potency, kinetics and voltage-dependency of a series of uncompetitive NMDA receptor antagonists in vitro with anticonvulsive and motor impairment activity in vivo. Neuropharmacology 1995;34:1239–58. [109] Barnes CA, Danysz W, Parsons CG. Effects of the uncompetitive NMDA receptor antagonist memantine on hippocampal long-term potentiation, short-term exploratory modulation and spatial memory in awake, freely moving rats. Eur J Neurosci 1996;8:565–71. [110] Danysz W, Parsons CG. The NMDA receptor antagonist memantine as a symptomatological and neuroprotective treatment for Alzheimer’s disease: preclinical evidence. Int J Geriatr Psychiatry 2003;18:S23–32. [111] Zajaczkowski W, Frankiewicz T, Parsons CG, Danysz W. Uncompetitive NMDA receptor antagonists attenuate NMDA-induced impairment of passive avoidance learning and LTP. Neuropharmacology 1997;36:961–71. [112] Zajaczkowski W, Quack G, Danysz W. Infusion of (+)-MK-801 and memantine—contrasting effects on radial maze learning in rats with entorhinal cortex lesion. Eur J Pharmacol 1996;296:239–46.
16
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17
[113] Bachurin S, Tkachenko S, Baskin I, Lermontova N, Mukhina T, Petrova L, et al. Neuroprotective and cognition-enhancing properties of MK-801 flexible analogs. Structure-activity relationships. Ann NY Acad Sci 2001;939:219–36. [114] Miguel-Hidalgo JJ, Alvarez XA, Cacabelos R, Quack G. Neuroprotection by memantine against neurodegeneration induced by beta-amyloid(1-40). Brain Res 2002;958:210–21. [115] Muller WE, Schroder HC, Ushijima H, Dapper J, Bormann J. gp120 of HIV-1 induces apoptosis in rat cortical cell cultures: prevention by memantine. Eur J Pharmacol 1992;226:209–14. [116] Winblad B, Poritis N. Memantine in severe dementia: results of the 9M-Best Study (benefit and efficacy in severely demented patients during treatment with memantine). Int J Geriatr Psychiatry 1999;14:135–46. [117] Ruther E, Glaser A, Bleich S, Degner D, Wiltfang J. A prospective PMS study to validate the sensitivity for change of the D-scale in advanced stages of dementia using the NMDA-antagonist memantine. Pharmacopsychiatry 2000;33:103–8. [118] Hartmann S, Mobius HJ. Tolerability of memantine in combination with cholinesterase inhibitors in dementia therapy. Int Clin Psychopharmacol 2003;18:81–5. [119] Wenk GL, Quack G, Moebius HJ, Danysz W. No interaction of memantine with acetylcholinesterase inhibitors approved for clinical use. Life Sci 2000;66:1079–83. [120] Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA 2004;291:317–24. [121] Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med 2003;348:1333–41. [122] Jarvis B, Figgitt DP. Memantine. Drugs Aging 2003;20:465–76 [discussion 468–77]. [123] Wilcock G, Mobius HJ, Stoffler A. A double-blind, placebocontrolled multicentre study of memantine in mild to moderate vascular dementia (MMM500). Int Clin Psychopharmacol 2002;17:297–305. [124] Porter RH, Greenamyre JT. Regional variations in the pharmacology of NMDA receptor channel blockers: implications for therapeutic potential. J Neurochem 1995;64:614–23. [125] Zajaczkowski W, Hetman M, Nikolaev E, Quack G, Danysz W, Kaczmarek L. Behavioural evaluation of long-term neurotoxic effects of NMDA receptor antagonists. Neurotox Res 2000;1:299– 310. [126] Hesselink MB, De Boer BG, Breimer DD, Danysz W. Brain penetration and in vivo recovery of NMDA receptor antagonists amantadine and memantine: a quantitative microdialysis study. Pharm Res 1999;16:637–42. [127] Kornhuber J, Bormann J, Hubers M, Rusche K, Riederer P. Effects of the 1-amino-adamantanes at the MK-801-binding site of the NMDA-receptor-gated ion channel: a human postmortem brain study. Eur J Pharmacol 1991;206:297–300. [128] Kornhuber J, Quack G. Cerebrospinal fluid and serum concentrations of the N-methyl-d-aspartate (NMDA) receptor antagonist memantine in man. Neurosci Lett 1995;195:137–9. [129] Ametamey SM, Samnick S, Leenders KL, Vontobel P, Quack G, Parsons CG, et al. Fluorine-18 radiolabelling, biodistribution studies and preliminary PET evaluation of a new memantine derivative for imaging the NMDA receptor. J Recept Signal Transduct Res 1999;19:129–41. [130] Freudenthaler S, Meineke I, Schreeb KH, Boakye E, Gundert-Remy U, Gleiter CH. Influence of urine pH and urinary flow on the renal excretion of memantine. Br J Clin Pharmacol 1998;46:541–6. [131] Frankiewicz T, Potier B, Bashir ZI, Collingridge GL, Parsons CG. Effects of memantine and MK-801 on NMDA-induced currents in cultured neurones and on synaptic transmission and LTP in area CA1 of rat hippocampal slices. Br J Pharmacol 1996;117:689–97.
[132] Wenk GL, Danysz W, Mobley SL. MK-801, memantine and amantadine show neuroprotective activity in the nucleus basalis magnocellularis. Eur J Pharmacol 1995;293:267–70. [133] Spanagel R, Eilbacher B, Wilke R. Memantine-induced dopamine release in the prefrontal cortex and striatum of the rat—a pharmacokinetic microdialysis study. Eur J Pharmacol 1994;262:21–6. [134] Hesselink MB, De Boer AG, Breimer DD, Danysz W. Dopamine release in the prefrontal cortex in response to memantine following sub-chronic NMDA receptor blockade with memantine: a microdialysis study in rats. J Neural Transm 1999;106:803–18. [135] Fredriksson A, Danysz W, Quack G, Archer T. Co-administration of memantine and amantadine with sub/suprathreshold doses of l-Dopa restores motor behaviour of MPTP-treated mice. J Neural Transm 2001;108:167–87. [136] Fisher A, Starr MS. Opposite effects of glutamate antagonists and antiparkinsonian drugs on the activities of DOPA decarboxylase and 5-HTP decarboxylase in the rat brain. Brain Res 2000;868:268–74. [137] Fredriksson A, Archer T. Functional alteration by NMDA antagonist: effects of l-Dopa, neuroleptics drug and postnatal administration. Amino Acids 2002;23:111–32. [138] Kozela E, Danysz W, Popik P. Uncompetitive NMDA receptor antagonists potentiate morphine antinociception recorded from the tail but not from the hind paw in rats. Eur J Pharmacol 2001;423:17– 26. [139] Belozertseva IV, Dravolina OA, Neznanova ON, Danysz W, Bespalov AY. Antinociceptive activity of combination of morphine and NMDA receptor antagonists depends on the inter-injection interval. Eur J Pharmacol 2000;396:77–83. [140] Peeters M, Maloteaux JM, Hermans E. Distinct effects of amantadine and memantine on dopaminergic transmission in the rat striatum. Neurosci Lett 2003;343:205–9. [141] Hesselink MB, Smolders H, Eilbacher B, De Boer AG, Breimer DD, Danysz W. The role of probenecid-sensitive organic acid transport in the pharmacokinetics of N-methyl-d-aspartate receptor antagonists acting at the glycine(B)-site: microdialysis and maximum electroshock seizures studies. J Pharmacol Exp Ther 1999;290:543– 50. [142] Olmos G, DeGregorio-Rocasolano N, Paz Regalado M, Gasull T, Assumpcio Boronat M, Trullas R, et al. Protection by imidazol(ine) drugs and agmatine of glutamate-induced neurotoxicity in cultured cerebellar granule cells through blockade of NMDA receptor. Br J Pharmacol 1999;127:1317–26. [143] Pabel J, Hofner G, Wanner KT. Synthesis and resolution of racemic eliprodil and evaluation of the enantiomers of eliprodil as NMDA receptor antagonists. Bioorg Med Chem Lett 2000;10:1377–80. [144] Schoemaker H, Claustre Y, Fage D, Rouquier L, Chergui K, Curet O, et al. Neurochemical characteristics of amisulpride, an atypical dopamine D2/D3 receptor antagonist with both presynaptic and limbic selectivity. J Pharmacol Exp Ther 1997;280:83–97. [145] Hu LY, Guo J, Magar SS, Fischer JB, Burke-Howie KJ, Durant GJ. Synthesis and pharmacological evaluation of N-(2,5-disubstituted phenyl)-N -(3-substituted phenyl)-N -methylguanidines as N-methyl-d-aspartate receptor ion-channel blockers. J Med Chem 1997; 40:4281–9. [146] Thompson WJ, Anderson PS, Britcher SF, Lyle TA, Thies JE, Magill CA, et al. Synthesis and pharmacological evaluation of a series of dibenzo[a,d]cycloalkenimines as N-methyl-d-aspartate antagonists. J Med Chem 1990;33:789–808. [147] Turski L, Huth A, Sheardown M, McDonald F, Neuhaus R, Schneider HH, et al. ZK200775: a phosphonate quinoxalinedione AMPA antagonist for neuroprotection in stroke and trauma. Proc Natl Acad Sci USA 1998;95:10960–5. [148] Karbon EW, Abreu ME, Erickson RH, Kaiser C, Natalie Jr KJ, Clissold DB, et al. NPC 16377, a potent and selective sigma-ligand. I. Receptor binding, neurochemical and neuroendocrine profile. J Pharmacol Exp Ther 1993;265:866–75.
S.K. Sonkusare et al. / Pharmacological Research 51 (2005) 1–17 [149] Salvati P, Maj R, Caccia C, Cervini MA, Fornaretto MG, Lamberti E, et al. Biochemical and electrophysiological studies on the mechanism of action of PNU-151774E, a novel antiepileptic compound. J Pharmacol Exp Ther 1999;288:1151–9. [150] Baudy RB, Fletcher III H, Yardley JP, Zaleska MM, Bramlett DR, Tasse RP, et al. Design, synthesis, SAR, and biological evaluation
17
of highly potent benzimidazole-spaced phosphono-alpha-amino acid competitive NMDA antagonists of the AP-6 type. J Med Chem 2001;44:1516–29. [151] Gortelmeyer R, Erbler H. Memantine in the treatment of mild to moderate dementia syndrome. A double-blind placebo-controlled study. Arzneimittelforschung 1992;42:904–13.