3,110
Views
33
CrossRef citations to date
0
Altmetric
Theme: Alzheimer's Disease - Review

Rationale for combining glutamatergic and cholinergic approaches in the symptomatic treatment of Alzheimer’s disease

, &
Pages 1351-1365 | Published online: 09 Jan 2014

Abstract

A total of 40 years of biochemical, clinical and neuropathological research have revolutionized our understanding of the pathophysiology of Alzheimer’s disease, yet at the present moment the only drugs licensed for treatment are targeted essentially at symptoms. Some disease-modifying drugs remain in clinical trials, but many that have used similar approaches have failed. It is therefore of considerable interest to examine the optimal way of using existing medications for the benefit of patients. This article looks at the rationale behind the combined use of acetylcholinesterase inhibitors and the N-methyl-d-aspartate-receptor antagonist, memantine, from both preclinical and clinical perspectives.

Figure 1. Normal cortical cholinergic and glutamatergic neurotransmission, changes that occur in Alzheimer’s disease, and the proposed mechanism of action of acetylcholinesterase inhibitors and memantine with respect to these changes.

(A) Normal cortical cholinergic and glutamergic neurotransmission. Cholinergic innervation of glutamatergic cortical pyramidal neurons is provided by cells in the nbM in the basal forebrain. Acetylcholine (ACh) is released following depolarization to act on nicotinic and muscarinic receptors located on glutamatergic neurons, with neurotransmitter action being terminated by AChE. Glutamatergic neurons also receive innervation from other glutamatergic cortical pyramidal neurons – in this case, released glutamate acts on α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and NMDA receptors and action is terminated mostly by the reuptake of glutamate into associated glial cells. (B) Changes that occur in Alzheimer’s disease (AD). In AD atrophy and dysfunction of cholinergic and glutamatergic pyramidal neurons occurs such that remaining cortical neurons receive less innervation and consequently are less likely to be depolarized by synaptic signals. Signal-to-noise ratio at glutamatergic synapses would be reduced by a combination of a weaker ‘signal’ (from ACh and glutamate release in response to depolarization) and greater ‘noise’ (caused by an increased concentration of glutamate in the synaptic cleft between synaptic activation, due to partial failure of reuptake mechanisms). (C) The proposed mechanism of action of AChEIs and memantine with respect to the changes in AD. The use of AChEIs in AD is proposed to return the concentration of ACh in the synapse towards the normal level by reducing its breakdown, and thereby increasing the chance of the interaction of ACh with cholinergic receptors on glutamatergic pyramidal neurons. Memantine is proposed to reduce ‘noise’ at glutamatergic synapses by preventing the NMDA receptors from responding to the increased concentrations of glutamate present in the synaptic cleft between synaptic activation. In combination, it is proposed that the AChEIs and memantine would have their individual effects, but the potential for synergistic action occurs because an improved signal-to-noise ratio at glutamatergic synapses in the nbM would be expected to increase the firing rate of cholinergic neurons, and AChE inhibition would enhance this effect.

Glutamate receptor; α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor has been excluded for clarity.

ACheE: Acetylcholinesterase; AChEI: Acetylcholinesterase inhibitor; ACh-r: Acetylcholine receptor; nbM: Nucleus basalis of Meynert; NMDA-r: NMDA receptor.

Left part of each panel adapted with permission from [Danysz W, Unpublished Data].

Figure 1. Normal cortical cholinergic and glutamatergic neurotransmission, changes that occur in Alzheimer’s disease, and the proposed mechanism of action of acetylcholinesterase inhibitors and memantine with respect to these changes.(A) Normal cortical cholinergic and glutamergic neurotransmission. Cholinergic innervation of glutamatergic cortical pyramidal neurons is provided by cells in the nbM in the basal forebrain. Acetylcholine (ACh) is released following depolarization to act on nicotinic and muscarinic receptors located on glutamatergic neurons, with neurotransmitter action being terminated by AChE. Glutamatergic neurons also receive innervation from other glutamatergic cortical pyramidal neurons – in this case, released glutamate acts on α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and NMDA receptors and action is terminated mostly by the reuptake of glutamate into associated glial cells. (B) Changes that occur in Alzheimer’s disease (AD). In AD atrophy and dysfunction of cholinergic and glutamatergic pyramidal neurons occurs such that remaining cortical neurons receive less innervation and consequently are less likely to be depolarized by synaptic signals. Signal-to-noise ratio at glutamatergic synapses would be reduced by a combination of a weaker ‘signal’ (from ACh and glutamate release in response to depolarization) and greater ‘noise’ (caused by an increased concentration of glutamate in the synaptic cleft between synaptic activation, due to partial failure of reuptake mechanisms). (C) The proposed mechanism of action of AChEIs and memantine with respect to the changes in AD. The use of AChEIs in AD is proposed to return the concentration of ACh in the synapse towards the normal level by reducing its breakdown, and thereby increasing the chance of the interaction of ACh with cholinergic receptors on glutamatergic pyramidal neurons. Memantine is proposed to reduce ‘noise’ at glutamatergic synapses by preventing the NMDA receptors from responding to the increased concentrations of glutamate present in the synaptic cleft between synaptic activation. In combination, it is proposed that the AChEIs and memantine would have their individual effects, but the potential for synergistic action occurs because an improved signal-to-noise ratio at glutamatergic synapses in the nbM would be expected to increase the firing rate of cholinergic neurons, and AChE inhibition would enhance this effect.†Glutamate receptor; α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor has been excluded for clarity.ACheE: Acetylcholinesterase; AChEI: Acetylcholinesterase inhibitor; ACh-r: Acetylcholine receptor; nbM: Nucleus basalis of Meynert; NMDA-r: NMDA receptor.Left part of each panel adapted with permission from [Danysz W, Unpublished Data].

Biochemical rationale for cholinergic & glutamatergic approaches in the treatment of cognitive & noncognitive symptoms

Cholinergic anatomy & pharmacology

Anatomical studies have identified the presence of cholinergic markers (choline acetyltransferase [ChAT], and vesicular acetylcholine transporter) associated with the cell bodies and dendrites of cortical neurons and, in particular, glutamatergic cortical pyramidal neurons Citation[1,2]. Most cortical neurons, and many glia, appear to carry muscarinic and/or nicotinic receptors Citation[3–5] making them sensitive to the activity of cholinergic basal forebrain neurons Citation[3,6,7]. Cholinergic neurons that innervate the neocortex and hippocampus are located in the basal forebrain, with cell groups in the medial septum, diagonal band of Broca, and the nucleus basalis of Meynert (nbM) Citation[8]. Furthermore, the activity of these cholinergic neurons is likely to be regulated by glutamatergic inputs from regions such as the amygdala, the brainstem reticular formation, the hippocampus and the cerebral cortex Citation[9–12]. Consistent with these observations, the nbM expresses large numbers of N-methyl-d-aspartate (NMDA) glutamate receptors Citation[13]. Various studies using microdialysis have investigated this regulation of cholinergic function by glutamatergic systems – the pharmacology appears complex and depends on the site of administration of the compounds. For example, administration of a glutamatergic antagonist intracerebroventricularly stimulated cortical acetylcholine (ACh) release Citation[14,15], whereas the same compound given through the dialysis probe in the nbM inhibited ACh release Citation[14]. It is therefore proposed that there is both a direct and an indirect (via a γ-aminobutyric acid inhibitory interneuron) regulation of ACh release by glutamate Citation[14,16]. The importance of the direct pathway is supported by in vivo studies which showed that both NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), applied through a dialysis probe in the nbM, increased cortical ACh release in a concentration-dependent manner Citation[17]; and by in vitro investigations in which NMDA antagonists inhibited ACh release from cortical slices Citation[18,19], and NMDA-induced tonic firing of nbM neurons in basal forebrain slices Citation[20].

A range of compounds that have been investigated as cognition enhancers have been shown to increase glutamate release in vivo, including an acetylcholinesterase inhibitor (AChEI) Citation[6], nicotine and subtype-specific nicotinic agonists Citation[21,22], a muscarinic agonist Citation[6], serotonin (5-HT)1A antagonists Citation[23,24], and a 5-HT6 antagonist Citation[25]. In this context, it is a tenable hypothesis that a key feature of compounds that act as cognitive enhancers is likely to be their ability to act on receptors located on pyramidal neurons, influencing pyramidal neuron activity and glutamate release Citation[26]. In the case of cholinomimetics, such an effect would be direct for muscarinic and nicotinic agonists, or indirect (by increasing the concentration of ACh at cholinergic–glutamatergic synapses) for AChEIs.

Cholinergic mechanisms in Alzheimer’s disease

The activation of cholinergic receptors underlies improvements in attention and other aspects of cognition Citation[27], and cholinergic dysfunction is associated with the early cognitive impairments observed in Alzheimer’s disease (AD). Studies of patients in the early stages of AD have found a consistent reduction in presynaptic markers of the cholinergic system. One investigation found that the levels of ChAT activity, ACh synthesis and high-affinity choline uptake were reduced to approximately half of the control level in patients with a mean of 3.5 years since symptom onset Citation[26,28]. A similar finding was reported in patients who had displayed symptoms for less than 1 year Citation[28,29]. These changes in cholinergic marker enzymes also appear to correlate with the extent of cognitive decline in AD Citation[28,30]. However, there is also evidence that ChAT activity may show stabilization, or even upregulation, in patients with mild cognitive impairment and early AD Citation[30,31] – although ChAT is not the rate-limiting step of ACh synthesis, which may have an influence on the interpretation of these data.

Further to enzymatic findings, studies have shown that AD brains have a reduced number of some ACh receptor types – for example, fewer nicotinic Citation[32] and M2 muscarinic Citation[33] receptors, located on presynaptic cholinergic terminals. By contrast, postsynaptic M1 and M3 muscarinic receptors appear to be relatively preserved, although there may be disruptions in signaling from M1 receptors to their secondary messenger systems (via G proteins) Citation[34–36]. There is also evidence that the extent of uncoupling between M1 receptors and G proteins correlates with dementia severity Citation[37]. These reports of postsynaptic disruption indicate that cholinergic abnormalities in AD may not be restricted to presynaptic locations, as previously thought. With regard to nicotinic receptors, the presynaptic reduction observed in AD reflects a decrease in both the α7 and α4β2-receptor subtypes Citation[38–40]. The importance of the α4β2 receptor in AD pathology is further suggested by observations of a positive correlation between cognitive performance (Mini-Mental State Examination score) and α4β2-receptor binding [Francis PT, Unpublished Data]. Indeed, preliminary studies have shown that agents acting at these nicotinic sites may have therapeutic effects in AD Citation[41], and several nicotinic agonists are currently in clinical development.

The AChEIs were developed to target presynaptic cholinergic dysfunction. They help to address cognitive deficits by preventing the breakdown of the already diminished ACh supply at the synaptic cleft. In this way, cholinergic neurotransmission is facilitated as the neurotransmitter binds and activates cholinergic receptors on postsynaptic cells, including glutamatergic pyramidal neurons Citation[6,26]. The three main AChEIs marketed for the treatment of AD – donepezil, rivastigmine, and galantamine – are licensed for use in patients with mild to moderately severe AD (donepezil is now also licensed in some countries for severe AD). Tacrine, the first licensed AChEI, is also still marketed but is no longer recommended due to safety issues, including hepatotoxicity. A summary of the symptomatic mechanisms (as well as approval dates and indications) of marketed cholinergic drugs is given in . Details of other cholinergic compounds in development, and those that have been discontinued due to safety concerns or lack of efficacy, are also shown.

Donepezil, rivastigmine and galantamine have similar mechanisms of action, and similar efficacy profiles. However, they can be distinguished by some mechanistic differences. As reviewed by Wilkinson et al. Citation[42], while all three drugs inhibit acetylcholinesterase (AChE), rivastigmine shows equally potent inhibition of butyrylcholinesterase, and in certain studies galantamine has demonstrated allosteric activation of α4 and α7-nicotinic receptors half-life. In terms of administration, donepezil’s long half-life (70 h), means that it can be dosed once daily Citation[43], while oral rivastigmine (half-life = 1 h, with 9 h of AChE inhibition) and galantamine (half-life = 7–8 h) require twice-daily dosing Citation[42,44,45], although a once-daily extended release formulation of galantamine Citation[46] and a once-daily rivastigmine skin-patch Citation[47] are both now widely available.

Translating these mechanisms into clinically relevant effects, large controlled studies have shown that AChEI monotherapy can produce cognitive benefits in most patients with mild-to-moderate AD Citation[48]. In addition, the AChEIs have also been shown to improve noncognitive behavioral symptoms such as apathy, irritability and anxiety Citation[49]. This latter effect may be explained by the reduction in ChAT and other cholinergic markers (e.g., M2 receptors) that have been seen to correlate with behavioral changes in AD Citation[33,50]. Therefore, cholinergic dysfunction may underlie more than cognitive deficit in AD. However, the symptomatic effects of AChEIs are frequently modest, suggesting that the cholinergic system cannot be the only contributing factor to cognitive (and behavioral) decline. It is likely that other neurotransmitter systems are also involved in mediating cognitive function, with cholinergic processes influencing particular aspects, such as attention and arousal Citation[28,51]. In addition, the potential involvement of postsynaptic cholinergic dysfunction in AD raises the possibility that the limited function of AChEIs is related to their addressing only presynaptic deficits, while downstream signaling remains impaired.

Glutamatergic mechanisms in AD

Glutamate is the principal excitatory neurotransmitter of the CNS. It accounts for approximately two-thirds of synapses in the neocortex (ACh synapses account for ~5%) Citation[52], and is involved in all aspects of cognition and higher mental function. In normal, healthy individuals, glutamatergic neurotransmission acts via both AMPA and NMDA receptors to produce long-term potentiation (LTP). LTP refers to a strengthening of synapses through repeated use and is central to the processes of learning and memory and, in addition to glutamatergic mechanisms, ACh and its receptors play a significant role in both the induction and maintenance of LTP Citation[53,54]. This latter point again emphasizes the intimate relationship of cholinergic and glutamatergic systems in cognitive aspects of brain function.

In AD, the number of glutamatergic pyramidal neurons is significantly reduced Citation[26,55,56], as is reflected in a decrease in wet weight and total protein content in the cortex Citation[57,58]. In the cerebral cortex and hippocampus, there is concomitant loss of glutamatergic neurons Citation[55,59] and abnormalities in NMDA and AMPA-receptor expression Citation[60–63] in patients with AD. There is also evidence that NMDA glutamate receptors may be internalized on exposure to amyloid-b (Ab) Citation[64], perhaps in part as a protective mechanism Citation[65], which may also lead to an apparent reduction in receptor binding in AD as the study method used isolates of cell membranes. However, in severe pathological cases, or severely affected brain regions, these biochemical data may not follow typical patterns when expressed as wet weight or total protein. For example, markers of extrinsic neurons (e.g., ChAT activity for cholinergic neurons) are reduced in cortical regions in severe AD, as they form only a small proportion of the neuropil. By contrast, markers of pyramidal neurons contribute significantly to the total protein content and may, therefore, not always follow this pattern of reduction in severe disease Citation[66]. Certainly, studies of b-tubulin, synaptophysin and the glial marker, glial fibrillary acidic protein (GFAP) have shown that these protein levels were unaltered relative to total protein content in the temporal cortex of patients with AD Citation[66,67]. However, even at the end stage of the disease, GFAP levels were increased in the less severely affected parietal cortex.

With regard to understanding the disease process in AD, it is of considerable interest to consider the temporal relationship between the loss of cholinergic and glutamatergic neurons. Biochemical studies of biopsy samples indicate that cholinergic neurons of the basal forebrain are the first to be affected, as discussed above. However, as the histopathological definition of AD must include the presence of neurofibrillary tangles that occur in glutamatergic cortical pyramidal neurons and are found in preclinical cases of AD Citation[68], it can be argued that changes in the glutamatergic system must be the first to occur. Such a conclusion could have implications for when a particular treatment approach might be expected to be effective, although it must be remembered that only a small proportion of glutamatergic neurons in selected regions will be involved while the loss of a significant proportion of cholinergic innervation of the entire cortex may be more relevant. It is clearly the case that, by the time of current clinical diagnosis, both cholinergic and glutamatergic systems are affected by the disease process.

As well as the consequences of complete neuronal loss, there may also be dysfunction in remaining glutamatergic neurons in the AD brain. A study of human autopsy samples (AD vs control brains) found that the level of the major glial glutamate transporter (excitatory amino acid transporter [EAAT2]) was unaffected in all the studied brain regions Citation[69], whereas the ability of glial cells to remove glutamate from the synaptic cleft has been shown to be impaired in the temporal cortex, as well as other brain regions Citation[70]. This may be explained by the action of free radicals on the glial glutamate transporter, as seen in the rat system with GLT-1 (the rat equivalent of EAAT2) Citation[71]. In addition, although the level of the vesicular glutamate transporter, VGLUT1 (which packages neurotransmitter glutamate into vesicles for synaptic release), was unaffected in the AD temporal (but not parietal) cortex Citation[66,67], its activity was lowered in this region Citation[72]. The mechanism(s) by which these protein activities are altered are, as yet, unknown. However, regarding their consequences, it is proposed that in AD, inadequate removal of glutamate from the synaptic cleft leads to an abnormal build-up of the neurotransmitter between the pre- and post-synaptic neurons Citation[73]. This raised background concentration (‘noise’) of glutamate causes the postsynaptic membrane to become more frequently depolarized, displacing the magnesium ion (Mg2+) that blocks the NMDA ion channel under physiological resting conditions Citation[74]. The reduced efficiency of this voltage-dependent Mg2+ block impairs the detection of incoming physiological signals by the NMDA receptor and the capacity for generating LTP is reduced. This mechanism, involving dysfunction in glutamate neurotransmission, may contribute to cognitive impairment in AD Citation[73–75]. Furthermore, excessive pathological glutamate stimulation can lead to neuronal death via excitotoxicity. This is thought to be a major factor in neuronal loss in stroke Citation[59,74], and may also contribute to the cell death observed in AD, although the exact extent of this contribution remains unclear Citation[26]. However, despite the correlations observed between the loss of glutamatergic neurons and cognitive decline in AD, very few reports have, as yet, documented a direct link between presynaptic glutamatergic markers and cognitive deficits at the neurochemical level Citation[66,76].

Positive modulation of the glutamate NMDA and AMPA receptors has been tested as a strategy for normalizing glutamatergic function, with some agents undergoing clinical trials in AD, although no such treatment has received regulatory approval. An alternative approach, using an uncompetitive NMDA-receptor antagonist, memantine, has shown greater success in modulating the glutamatergic system in AD. Rather than simply providing a complete block of the NMDA receptor, memantine has moderate voltage dependency and fast blocking/unblocking kinetics. There is evidence that these characteristics allow it to block the NMDA-receptor channel during the constant background ‘noise’ of pathological glutamatergic activation at rest, while still being displaced upon the arrival of a physiological signal Citation[74,75]. In this way, the neuron is likely to be protected against the neurotoxic effects of pathological glutamate stimulation Citation[77], while also being able to manifest LTP Citation[73,74]. As described earlier, AChEIs are able to stimulate glutamatergic neurons. Therefore, in combined use, it may be hypothesized that while memantine would reduce the background ‘noise’ of glutamate in AD, the AChEIs would serve to increase the level of physiological signals Citation[73].

As listed in , memantine is currently the only marketed drug for AD that targets the glutamatergic system, although other agents acting at NMDA and AMPA receptors are in development. Memantine is indicated for the treatment of moderate-to-severe disease stages, with support coming from multiple large-scale, controlled clinical studies, including demonstration of efficacy against overall and individual items of cognitive function Citation[78,79]. In addition to cognitive benefits, a range of post hoc analyses (including pooled data) have demonstrated that memantine improves behavioral scores in general, and in particular, subscales associated with agitation and aggressive behavior Citation[80–83]. The potential mechanisms underlying such improvements have been reviewed elsewhere Citation[75].

Preclinical behavioral studies of individual cholinergic & glutamatergic approaches

The biochemical rationale for the use of individual cholinergic and glutamatergic therapies in AD is complemented by findings from cognitive behavioral studies in several animal model systems.

In a recent study using the novel object recognition test in rats, donepezil (and also memantine) was shown to have improved ‘natural forgetting’ when the animals were exposed to a second test after a 4-h intertrial interval Citation[84]. In similar earlier studies, donepezil and metrifonate (also an AChEI) improved discrimination in the same test, particularly in older animals Citation[85–87]. In addition, lesions of the fimbria-fornix, and local microinfusion of scopolamine (a muscarinic-receptor antagonist) into the hippocampus, both impair short-term object recognition in rats, whereas local microinfusion of physostigmine (a reversible AChEI) enhances object recognition Citation[88]. Donepezil was shown to reverse the effects of scopolamine in a wide range of cognitive behavioral tests to a varying degree Citation[89], and also ameliorated sustained attentional deficits in a five-choice serial reaction time test in the triple transgenic mouse model Citation[90].

The effects of memantine on cognitive performance in animals have been the subject of previous reviews Citation[91], with beneficial actions on aspects of memory seen in several models, including amyloid precursor protein APP23, double (APP/presenilin-1, PS1) and triple (APP/PS1/Tau) transgenic mice models. Memantine has also been shown to reverse the effects of intracerebral Ab injections on both object recognition memory and passive avoidance in rats Citation[92].

In the APP transgenic mouse model of cognitive decline, memantine, galantamine and donepezil have all been shown independently to reverse cognitive deficits observed in the water maze test, both immediately Citation[93] and after a 3-week washout, indicating a potentially disease-modifying action Citation[94,95]. In addition, chronic memantine treatment has been shown to fully reverse, and donepezil to partially reverse, the effects of fimbria-fornix lesions on a delayed nonmatch to sample task in rats Citation[16].

Preclinical pharmacological & behavioral studies of combined cholinergic & glutamatergic approaches

The efficacy of individual treatment approaches with contrasting cholinergic and glutamatergic mechanisms of action prompted investigation of the effects of targeting both systems for the treatment of AD.

To date, there have been few preclinical studies investigating the combined effects of memantine and an AChEI (mostly donepezil) on cognitive behavior in animals . One study examined the effect of combination therapy on cognitive outcomes in rats, alongside a microdialysis investigation of ACh levels Citation[16]. A strong limitation of the study was that all experiments were conducted in the presence of the AChEI, neostigmine, without which the levels of ACh would not be detected in this system Citation[16]. In anesthetized rats, memantine or donepezil, given systemically, dose-dependently increased extracellular fluid concentrations of ACh in the neocortex and hippocampus, and when the drugs were combined at submaximal concentrations, greater increases were observed when compared with either drug alone Citation[16]. Increases in ACh concentration were also observed following the administration of memantine, and memantine plus donepezil, in rats with partial fimbria-fornix lesions; however, the combination was no more effective than memantine alone Citation[16]. Taken together, these results indicate that memantine is likely to increase ACh release both by increasing the firing rate of cholinergic neurons of the basal forebrain and at the level of cholinergic terminals. This latter action is not likely to be as a result of a direct effect on AChE inhibition, as therapeutic concentrations of memantine do not alter the degree of inhibition of AChEIs Citation[96,97], and memantine alone does not act as an AChEI Citation[96]. Initial attempts in this study to find a direct correlation between treatment-induced changes in ACh levels and improvements in cognitive outcomes were not successful Citation[16].

Other studies in healthy, uncompromised animals, showed that while either approach alone could improve performance Citation[98,99], no additional benefit was obtained when the drugs were combined Citation[100,101]. Similarly, while individually memantine and donepezil ameliorated the effects of Ab injections in a delayed nonmatch to position task in rats, the combination was no more effective Citation[102]. It is important to consider, however, that with drug combinations ceiling effects may come into play in situations where one drug is being used at an effective dose.

In more recent studies, memantine treatment alone and the combination of memantine and donepezil, significantly improved spatial memory (acquisition and retention in the Morris water maze) in 6-month and 15-month triple-transgenic mice with cognitive impairment and severe AD pathology (Ab plaques and neurofibrillary tangles) Citation[103]. By contrast, donepezil treatment alone significantly improved the retention (but not the acquisition) of spatial memory in younger mice, but no significant effects were seen in older mice Citation[103]. In both age groups, the memantine–donepezil combination was the only treatment that significantly improved the latency aspect of retention Citation[103]. A further study in the APP23 transgenic mouse model has suggested that memantine and donepezil may have a synergistic effect on spatial memory, improving moving time (reflecting memory acquisition) and resting time (reflecting retrieval time), respectively, in a dry land maze test Citation[104].

Clinical experience of combination treatment

Due, in part, to the logical nature of the approach, combination therapy is already widely used in clinical practice. Although compounds with a combined cholinergic–glutamatergic action have been trialed, none has yet been successful in the later stages of development . Consequently, combination therapy currently involves the administration of one cholinergic drug (donepezil, galantamine or rivastigmine) and one glutamatergic drug (memantine).

There have been no clinical trials involving simultaneous initiation of therapy with memantine and an AChEI, rather all studies have involved memantine being added on in patients receiving stable AChEI therapy. There are also no studies looking at AChEI addition in patients already on stable therapy with memantine. This may partly be explained by their usual licensed indications being slightly different – mild to moderately severe AD for AChEIs, and moderate-to-severe AD for memantine.

In line with in vitro, in vivo and ex vivo preclinical findings Citation[96,97,105], no pharmacokinetic interactions have been observed in clinical studies of memantine combined with donepezil, galantamine or rivastigmine Citation[106–108]. These pharmacokinetic studies were performed in healthy volunteers, and in patients with mild to moderate AD, and their findings are consistent with the favorable safety profile observed in Phase III and postmarketing studies of combination treatment to date Citation[79,109,110].

Efficacy data from the clinical studies have been systematically reviewed elsewhere Citation[111], and are listed in . In summary, two large-scale randomized, double-blind, placebo-controlled Phase III studies have assessed the efficacy of memantine in patients already receiving stable doses of AChEIs. One of these studies included patients with moderate-to-severe AD Citation[79], and the second included patients with mild-to-moderate disease Citation[109]. In addition, pooled and meta-analyses and several post hoc assessments have been based on these study data . Overall, the available studies indicate that combination treatment is superior to treatment with AChEI monotherapy in both general and specific measures of cognition, function, behavior and global status in patients with moderate-to-severe AD Citation[79,80,82,83,112–114]. Furthermore, there is evidence that combination therapy may reduce the occurrence of clinical worsening (concurrent worsening across symptomatic domains) Citation[115], and prevent the emergence of behavioral symptoms including agitation/aggression Citation[83]. By contrast, combination therapy has shown no significant benefits over AChEI monotherapy in patients with mild disease Citation[109,116–118]. Another prospective, placebo-controlled study of the memantine–donepezil combination (vs monotherapy with either agent) in patients with moderate-to-severe AD has recently been published Citation[119]. Although the power of this study was limited by significant difficulties with patient recruitment, the efficacy of memantine–donepezil combination treatment was seen on the coprimary end point measures of cognition and function Citation[119]. The superiority of combination therapy over monotherapy did not reach statistical significance, despite appearing differentiated for a large part of the treatment period – a finding which has since been described as ‘clinically relevant’ in expert commentary, which also suggests that there is no actual evidence of lack of effect of adjunctive memantine in this study due to issues such as small sample sizes at the end of the study and differential dropout rates Citation[201].

Outside the clinical setting, observational studies have also provided interesting preliminary findings regarding the long-term and ‘real world’ application of combination therapy for AD. Although these studies are, by their nature, nonrandomized and open-label and there are potential confounds since the cohorts receiving the different treatments are potentially dissimilar and largely from different time periods, results indicate that combination therapy may retain its benefits over AChEI monotherapy for a period of years Citation[120] and have an impact on meaningful outcomes, such as time to nursing home admission Citation[121].

Expert commentary

The use of combination therapy with a cholinomimetic (AChEI) and a glutamatergic (memantine) agent for the treatment of AD is justified by findings from biochemical studies, and preclinical investigations in established animal models of AD. This is borne out by most clinical experience and, indeed, combination therapy is already considered standard treatment practice in many countries throughout the world. Despite this extensive use, the clinical evidence support for combination therapy is not complete, as it is based solely on the addition of memantine to existing AChEI treatment, with no data on the opposite order of initiation or the simultaneous treatment de novo with memantine and an AChEI. These appear to be gaps, but the available data actually reflect the real-world situation in which AChEIs (licensed for mild to moderately severe AD) are given early in the disease course, while memantine (licensed for moderate-to-severe AD) is initiated later. Therefore, the natural order of events is that patients who receive combination therapy will have started their treatment with AChEI monotherapy.

There is a great hope that disease-modifying treatments will succeed in the near future although the risk is that those based on Ab are being investigated through clinical trials too late in the disease course. New initiatives are being considered where people with autosomal dominant inherited AD and those with genetic risk factors such as apolipoprotein E4 (APOE4) are given anti-Ab compounds. This would arguably be the strongest possible test of the amyloid cascade hypothesis.

The direction taken by AD management over the coming years will very much depend on the effectiveness of these treatments; if effectiveness is less than clear cut, then the adoption of such novel therapies is likely to be limited. However, regardless of the outcome with disease-modifying medications, current symptomatic treatments will continue to be the foundation of therapy for many patients with AD, including those diagnosed too late to benefit from any disease-modifying option – with particular relevance in developing countries. Therefore, combination therapy with memantine and an AChEI is likely to remain a widely used approach. In addition, it is recommended that new treatments for cognitive symptoms, and especially noncognitive symptoms, ought to be a major priority for development. These treatments should be identified by a combination of pragmatism – assessing what is available and what might work – as well as an effort to pinpoint mechanism-based approaches. In this context, it is relevant to recognize an important initiative by the American Alzheimer’s Association in bringing together scientists, clinicians and pharmaceutical companies in a ‘round table’ on behavior in dementia. This has resulted in the formation of a Professional Interest Area (PIA) on neuropsychiatric symptoms and the establishment of several working groups to address individual symptoms Citation[122].

Five-year view

The future of the AD treatment landscape in 5 years’ time will depend almost entirely on two factors – whether disease-modifying treatments are proven effective and whether they represent value for money. The expectation is that the current symptomatic drugs will still be used extensively, with clinical trials of other drugs to target behavioral disturbance near to completion. One of the current disease-modifying approaches may show promise at this time, but may not be widely available due to the limitations of cost, complexity of administration or side effects.

Diagnosis would be expected to present an enormous challenge worldwide, with the need to facilitate diagnosis in primary care balanced against the development of more sophisticated and expensive techniques (e.g., positron-emission tomography scanning) to improve diagnostic accuracy. Furthermore, in line with the push towards disease modification, the characterization of biomarkers to aid in early diagnosis is likely to have advanced.

Together, the increased range of options for diagnosis and treatment will undoubtedly present hard choices, creating (one would hope) the potential for enhanced treatment effects, but also producing greater disparity in care availability within and between countries.

Table 1. Cholinergic drugs in clinical development/marketplace.

Table 2. Glutamatergic drugs in clinical development/marketplace.

Table 3. Preclinical investigations of memantine–acetylcholinesterase inhibitor combination treatment.

Table 4. Mixed cholinergic and glutamatergic drugs in clinical development/market place.

Table 5. Studies examining the efficacy of memantine–acetylcholinesterase inhibitor combination treatment in patients with Alzheimer’s disease/dementia.

Key issues

  • • Cholinergic deficit is a feature of Alzheimer’s disease (AD) that correlates with disease severity.

  • • Glutamatergic dysfunction is another major contributory factor to AD.

  • • Preclinical evidence supports the targeting of cholinomimetic and glutamatergic mechanisms for the treatment of AD, with improved cognition observed in pharmacological and behavioral studies in animal models.

  • • In the clinical setting, cholinomimetic therapy (with acetylcholinesterase inhibitors) and N-methyl-d-aspartate-receptor antagonism (with memantine), have shown significant symptomatic efficacy and are approved for the treatment of AD.

  • • The acetylcholinesterase inhibitors and memantine have been widely used as monotherapies for AD over the past decade but, increasingly, the application of both approaches in combination treatment is seen as the standard of care, although no studies looking at the simultaneous use of the two types of drug de novo have been conducted.

  • • Preclinical and clinical evidence indicates that combination therapy is the way forward for the treatment of AD.

Financial and competing interest disclosure

PT Francis has received speaker bureau honoraria and grant support from H. Lundbeck A/S; speaker bureau honoraria from Novartis and Eisai; and has provided expert testimony to the UK High Court in a patent challenge against Novartis. CG Parsons is an employee of Merz Pharmaceuticals GmbH. RW Jones has received speaker bureau honoraria and/or consultancy fees from H Lundbeck A/S, Merz, Pfizer, Eisai and Novartis, as well as other companies involved in the development of new compounds for the treatment of Alzheimer’s disease and other dementias. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Writing assistance was utilized in the production of this manuscript. The authors gratefully acknowledge the editorial support provided by Cambridge Medical Communication Limited, funded by H. Lundbeck A/S.

References

  • Mesulam MM, Hersh LB, Mash DC, Geula C. Differential cholinergic innervation within functional subdivisions of the human cerebral cortex: a choline acetyltransferase study. J. Comp. Neurol. 318(3), 316–328 (1992).
  • Turrini P, Casu MA, Wong TP, De Koninck Y, Ribeiro-da-Silva A, Cuello AC. Cholinergic nerve terminals establish classical synapses in the rat cerebral cortex: synaptic pattern and age-related atrophy. Neuroscience 105(2), 277–285 (2001).
  • Chessell IP, Francis PT, Bowen DM. Changes in cortical nicotinic acetylcholine receptor numbers following unilateral destruction of pyramidal neurones by intrastriatal volkensin injection. Neurodegeneration 4(4), 415–424 (1995).
  • Chessell IP, Francis PT, Pangalos MN, Pearson RC, Bowen DM. Localisation of muscarinic (m1) and other neurotransmitter receptors on corticofugal-projecting pyramidal neurones. Brain Res. 632(1–2), 86–94 (1993).
  • Chessell IP, Humphrey PP. Nicotinic and muscarinic receptor-evoked depolarizations recorded from a novel cortical brain slice preparation. Neuropharmacology 34(10), 1289–1296 (1995).
  • Dijk SN, Francis PT, Stratmann GC, Bowen DM. Cholinomimetics increase glutamate outflow via an action on the corticostriatal pathway: implications for Alzheimer’s disease. J. Neurochem. 65(5), 2165–2169 (1995).
  • Kozhemyakin M, Rajasekaran K, Kapur J. Central cholinesterase inhibition enhances glutamatergic synaptic transmission. J. Neurophysiol. 103(4), 1748–1757 (2010).
  • Bigl V, Woolf NJ, Butcher LL. Cholinergic projections from the basal forebrain to frontal, parietal, temporal, occipital, and cingulate cortices: a combined fluorescent tracer and acetylcholinesterase analysis. Brain Res. Bull. 8(6), 727–749 (1982).
  • Carnes KM, Fuller TA, Price JL. Sources of presumptive glutamatergic/aspartatergic afferents to the magnocellular basal forebrain in the rat. J. Comp. Neurol. 302(4), 824–852 (1990).
  • Fonnum F, Walaas I. The effect of intrahippocampal kainic acid injections and surgical lesions on neurotransmitters in hippocampus and septum. J. Neurochem. 31(5), 1173–1181 (1978).
  • Francis PT, Carl R, Pearson A et al. The dementia of Alzheimer’s disease: an update. J. Neurol. Neurosurg. Psychiatr. 50(2), 242–243 (1987).
  • Mesulam MM, Mufson EJ. Neural inputs into the nucleus basalis of the substantia innominata (Ch4) in the rhesus monkey. Brain 107(Pt 1), 253–274 (1984).
  • Zilles K, Werner L, Qü M, Schleicher A, Gross G. Quantitative autoradiography of 11 different transmitter binding sites in the basal forebrain region of the rat–evidence of heterogeneity in distribution patterns. Neuroscience 42(2), 473–481 (1991).
  • Giovannini MG, Giovannelli L, Bianchi L, Kalfin R, Pepeu G. Glutamatergic modulation of cortical acetylcholine release in the rat: a combined in vivo microdialysis, retrograde tracing and immunohistochemical study. Eur. J. Neurosci. 9(8), 1678–1689 (1997).
  • Hasegawa M, Kinoshita H, Amano M, Hasegawa T, Kameyama T, Nabeshima T. MK-801 increases endogenous acetylcholine release in the rat parietal cortex: a study using brain microdialysis. Neurosci. Lett. 150(1), 53–56 (1993).
  • Ihalainen J, Sarajärvi T, Rasmusson D et al. Effects of memantine and donepezil on cortical and hippocampal acetylcholine levels and object recognition memory in rats. Neuropharmacology 61(5–6), 891–899 (2011).
  • Fournier GN, Materi LM, Semba K, Rasmusson DD. Cortical acetylcholine release and electroencephalogram activation evoked by ionotropic glutamate receptor agonists in the rat basal forebrain. Neuroscience 123(3), 785–792 (2004).
  • Lodge D, Johnston GA. Effect of ketamine on amino acid-evoked release of acetylcholine from rat cerebral cortex in vitro. Neurosci. Lett. 56(3), 371–375 (1985).
  • Ulus IH, Buyukuysal RL, Wurtman RJ. N-methyl-d-aspartate increases acetylcholine release from rat striatum and cortex: its effect is augmented by choline. J. Pharmacol. Exp. Ther. 261(3), 1122–1128 (1992).
  • Khateb A, Fort P, Serafin M, Jones BE, Mühlethaler M. Rhythmical bursts induced by NMDA in guinea-pig cholinergic nucleus basalis neurones in vitro. J. Physiol. (Lond.) 487 (Pt 3), 623–638 (1995).
  • Toth E. Effect of nicotine on the level of extracellular amino acids in the hippocampus of rat. Neurochem. Res. 21(8), 903–907 (1996).
  • Zappettini S, Grilli M, Lagomarsino F, Cavallero A, Fedele E, Marchi M. Presynaptic nicotinic α7 and non-α7 receptors stimulate endogenous GABA release from rat hippocampal synaptosomes through two mechanisms of action. PLoS ONE 6(2), e16911 (2011).
  • Dijk SN, Francis PT, Stratmann GC, Bowen DM. NMDA-induced glutamate and aspartate release from rat cortical pyramidal neurones: evidence for modulation by a 5-HT1A antagonist. Br. J. Pharmacol. 115(7), 1169–1174 (1995).
  • Schechter LE, Smith DL, Rosenzweig-Lipson S et al. Lecozotan (SRA-333): a selective serotonin 1A receptor antagonist that enhances the stimulated release of glutamate and acetylcholine in the hippocampus and possesses cognitive-enhancing properties. J. Pharmacol. Exp. Ther. 314(3), 1274–1289 (2005).
  • Dawson LA, Nguyen HQ, Li P. The 5-HT(6) receptor antagonist SB-271046 selectively enhances excitatory neurotransmission in the rat frontal cortex and hippocampus. Neuropsychopharmacology 25(5), 662–668 (2001).
  • Francis PT, Sims NR, Procter AW, Bowen DM. Cortical pyramidal neurone loss may cause glutamatergic hypoactivity and cognitive impairment in Alzheimer’s disease: investigative and therapeutic perspectives. J. Neurochem. 60(5), 1589–1604 (1993).
  • Stanhope KJ, McLenachan AP, Dourish CT. Dissociation between cognitive and motor/motivational deficits in the delayed matching to position test: effects of scopolamine, 8-OH-DPAT and EAA antagonists. Psychopharmacology (Berl.) 122(3), 268–280 (1995).
  • Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic hypothesis of Alzheimer’s disease: a review of progress. J. Neurol. Neurosurg. Psychiatr. 66(2), 137–147 (1999).
  • Bowen DM, Benton JS, Spillane JA, Smith CC, Allen SJ. Choline acetyltransferase activity and histopathology of frontal neocortex from biopsies of demented patients. J. Neurol. Sci. 57(2–3), 191–202 (1982).
  • Davis KL, Mohs RC, Marin D et al. Cholinergic markers in elderly patients with early signs of Alzheimer disease. JAMA 281(15), 1401–1406 (1999).
  • DeKosky ST, Ikonomovic MD, Styren SD et al. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Ann. Neurol. 51(2), 145–155 (2002).
  • Court J, Martin-Ruiz C, Piggott M, Spurden D, Griffiths M, Perry E. Nicotinic receptor abnormalities in Alzheimer’s disease. Biol. Psychiatry 49(3), 175–184 (2001).
  • Lai MK, Lai OF, Keene J et al. Psychosis of Alzheimer’s disease is associated with elevated muscarinic M2 binding in the cortex. Neurology 57(5), 805–811 (2001).
  • Warpman U, Alafuzoff I, Nordberg A. Coupling of muscarinic receptors to GTP proteins in postmortem human brain–alterations in Alzheimer’s disease. Neurosci. Lett. 150(1), 39–43 (1993).
  • Flynn DD, Weinstein DA, Mash DC. Loss of high-affinity agonist binding to M1 muscarinic receptors in Alzheimer’s disease: implications for the failure of cholinergic replacement therapies. Ann. Neurol. 29(3), 256–262 (1991).
  • Tsang SW, Pomakian J, Marshall GA et al. Disrupted muscarinic M1 receptor signaling correlates with loss of protein kinase C activity and glutamatergic deficit in Alzheimer’s disease. Neurobiol. Aging 28(9), 1381–1387 (2007).
  • Tsang SW, Lai MK, Kirvell S et al. Impaired coupling of muscarinic M1 receptors to G-proteins in the neocortex is associated with severity of dementia in Alzheimer’s disease. Neurobiol. Aging 27(9), 1216–1223 (2006).
  • Perry EK, Morris CM, Court JA et al. Alteration in nicotine binding sites in Parkinson’s disease, Lewy body dementia and Alzheimer’s disease: possible index of early neuropathology. Neuroscience 64(2), 385–395 (1995).
  • Nordberg A. Nicotinic receptor abnormalities of Alzheimer’s disease: therapeutic implications. Biol. Psychiatry 49(3), 200–210 (2001).
  • Gotti C, Moretti M, Bohr I et al. Selective nicotinic acetylcholine receptor subunit deficits identified in Alzheimer’s disease, Parkinson’s disease and dementia with Lewy bodies by immunoprecipitation. Neurobiol. Dis. 23(2), 481–489 (2006).
  • Levin ED, McClernon FJ, Rezvani AH. Nicotinic effects on cognitive function: behavioral characterization, pharmacological specification, and anatomic localization. Psychopharmacology (Berl.) 184(3–4), 523–539 (2006).
  • Wilkinson DG, Francis PT, Schwam E, Payne-Parrish J. Cholinesterase inhibitors used in the treatment of Alzheimer’s disease: the relationship between pharmacological effects and clinical efficacy. Drugs Aging 21(7), 453–478 (2004).
  • Aricept® Summary of Product Characteristics (SPC). Eisai Ltd, NJ, USA, June 2011.
  • Exelon® Summary of Product Characteristics (SPC). Novartis Pharmaceuticals UK Ltd., April 2012.
  • Reminyl® Summary of Product Characteristics (SPC). Shire Pharmaceuticals Ltd., October 2010.
  • Reminyl® XL Summary of Product Characteristics (SPC). Shire Pharmaceuticals Ltd., August 2010.
  • Exelon® Transdermal patch Summary of Product Characteristics (SPC). Novartis Pharmaceuticals UK Ltd., April 2012.
  • Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst. Rev. 1, CD005593 (2006).
  • Gauthier S, Feldman H, Hecker J et al.; Donepezil MSAD Study Investigators Group. Efficacy of donepezil on behavioral symptoms in patients with moderate to severe Alzheimer’s disease. Int. Psychogeriatr. 14(4), 389–404 (2002).
  • Minger SL, Esiri MM, McDonald B et al. Cholinergic deficits contribute to behavioral disturbance in patients with dementia. Neurology 55(10), 1460–1467 (2000).
  • Francis PT, Ramírez MJ, Lai MK. Neurochemical basis for symptomatic treatment of Alzheimer’s disease. Neuropharmacology 59(4–5), 221–229 (2010).
  • Fonnum F. Glutamate: a neurotransmitter in mammalian brain. J. Neurochem. 42(1), 1–11 (1984).
  • Auerbach JM, Segal M. A novel cholinergic induction of long-term potentiation in rat hippocampus. J. Neurophysiol. 72(4), 2034–2040 (1994).
  • Tai SK, Leung LS. Vestibular stimulation enhances hippocampal long-term potentiation via activation of cholinergic septohippocampal cells. Behav. Brain Res. 232(1), 174–182 (2012).
  • Morrison JH, Hof PR. Life and death of neurons in the aging brain. Science 278(5337), 412–419 (1997).
  • Neary D, Snowden JS, Mann DM et al. Alzheimer’s disease: a correlative study. J. Neurol. Neurosurg. Psychiatr. 49(3), 229–237 (1986).
  • Najlerahim A, Bowen DM. Biochemical measurements in Alzheimer’s disease reveal a necessity for improved neuroimaging techniques to study metabolism. Biochem. J. 251(1), 305–308 (1988).
  • Najlerahim A, Bowen DM. Regional weight loss of the cerebral cortex and some subcortical nuclei in senile dementia of the Alzheimer type. Acta Neuropathol. 75(5), 509–512 (1988).
  • Greenamyre JT, Maragos WF, Albin RL, Penney JB, Young AB. Glutamate transmission and toxicity in Alzheimer’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 12(4), 421–430 (1988).
  • Greenamyre JT, Penney JB, Young AB, D’Amato CJ, Hicks SP, Shoulson I. Alterations in L-glutamate binding in Alzheimer’s and Huntington’s diseases. Science 227(4693), 1496–1499 (1985).
  • Greenamyre JT, Penney JB, D’Amato CJ, Young AB. Dementia of the Alzheimer’s type: changes in hippocampal L-[3H]glutamate binding. J. Neurochem. 48(2), 543–551 (1987).
  • Procter AW, Wong EH, Stratmann GC, Lowe SL, Bowen DM. Reduced glycine stimulation of [3H]MK-801 binding in Alzheimer’s disease. J. Neurochem. 53(3), 698–704 (1989).
  • Wakabayashi K, Narisawa-Saito M, Iwakura Y et al. Phenotypic down-regulation of glutamate receptor subunit GluR1 in Alzheimer’s disease. Neurobiol. Aging 20(3), 287–295 (1999).
  • Snyder EM, Nong Y, Almeida CG et al. Regulation of NMDA receptor trafficking by amyloid-b. Nat. Neurosci. 8(8), 1051–1058 (2005).
  • Danysz W, Parsons CG. Alzheimer’s disease, β-amyloid, glutamate, NMDA receptors and memantine – searching for the connections. Br. J. Pharmacol. 167(2), 324–352 (2012).
  • Kirvell SL, Esiri M, Francis PT. Down-regulation of vesicular glutamate transporters precedes cell loss and pathology in Alzheimer’s disease. J. Neurochem. 98(3), 939–950 (2006). Erratum in: J. Neurochem. 100(6), 1713 (2007).
  • Kirvell SL, Francis PT. Vesicular glutamate transporters and their possible role in Alzheimer’s disease. Poster presented at: Society for Neuroscience Annual Meeting. San Diego, California, USA, 23–27 October (2004).
  • Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82(4), 239–259 (1991).
  • Beckstrøm H, Julsrud L, Haugeto O et al. Interindividual differences in the levels of the glutamate transporters GLAST and GLT, but no clear correlation with Alzheimer’s disease. J. Neurosci. Res. 55(2), 218–229 (1999).
  • Procter AW, Palmer AM, Francis PT et al. Evidence of glutamatergic denervation and possible abnormal metabolism in Alzheimer’s disease. J. Neurochem. 50(3), 790–802 (1988).
  • Keller JN, Mark RJ, Bruce AJ et al. 4-Hydroxynonenal, an aldehydic product of membrane lipid peroxidation, impairs glutamate transport and mitochondrial function in synaptosomes. Neuroscience 80(3), 685–696 (1997).
  • Westphalen RI, Scott HL, Dodd PR. Synaptic vesicle transport and synaptic membrane transporter sites in excitatory amino acid nerve terminals in Alzheimer disease. J. Neural Transm. 110(9), 1013–1027 (2003).
  • Francis PT. Glutamatergic systems in Alzheimer’s disease. Int. J. Geriatr. Psychiatry 18(Suppl. 1), S15–S21 (2003).
  • Danysz W, Parsons CG, Mobius HJ, Stoffler A, Quack G. Neuroprotective and symptomatological action of memantine relevant for Alzheimer’s disease – a unified glutamatergic hypothesis on the mechanism of action. Neurotox. Res. 2(2–3), 85–97 (2000).
  • Francis PT. Altered glutamate neurotransmission and behaviour in dementia: evidence from studies of memantine. Curr. Mol. Pharmacol. 2(1), 77–82 (2009).
  • Kashani A, Lepicard E, Poirel O et al. Loss of VGLUT1 and VGLUT2 in the prefrontal cortex is correlated with cognitive decline in Alzheimer disease. Neurobiol. Aging 29(11), 1619–1630 (2008).
  • Chen HS, Pellegrini JW, Aggarwal SK et al. Open-channel block of N-methyl-d-aspartate (NMDA) responses by memantine: therapeutic advantage against NMDA receptor-mediated neurotoxicity. J. Neurosci. 12(11), 4427–4436 (1992).
  • Reisberg B, Doody R, Stöffler A, Schmitt F, Ferris S, Möbius HJ; Memantine Study Group. Memantine in moderate-to-severe Alzheimer’s disease. N. Engl. J. Med. 348(14), 1333–1341 (2003).
  • 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 291(3), 317–324 (2004).
  • Winblad B, Jones RW, Wirth Y, Stöffler A, Möbius HJ. Memantine in moderate to severe Alzheimer’s disease: a meta-analysis of randomised clinical trials. Dement. Geriatr. Cogn. Disord. 24(1), 20–27 (2007).
  • Gauthier S, Loft H, Cummings J. Improvement in behavioural symptoms in patients with moderate to severe Alzheimer’s disease by memantine: a pooled data analysis. Int. J. Geriatr. Psychiatry 23(5), 537–545 (2008).
  • Gauthier S, Wirth Y, Möbius HJ. Effects of memantine on behavioural symptoms in Alzheimer’s disease patients: an analysis of the Neuropsychiatric Inventory (NPI) data of two randomised, controlled studies. Int. J. Geriatr. Psychiatry 20(5), 459–464 (2005).
  • Cummings JL, Schneider E, Tariot PN, Graham SM; Memantine MEM-MD-02 Study Group. Behavioral effects of memantine in Alzheimer disease patients receiving donepezil treatment. Neurology 67(1), 57–63 (2006).
  • Kendall I, Slotten HA, Codony X et al. E-6801, a 5-HT6 receptor agonist, improves recognition memory by combined modulation of cholinergic and glutamatergic neurotransmission in the rat. Psychopharmacology (Berl.) 213(2–3), 413–430 (2011).
  • Lieben CK, Blokland A, Sik A, Sung E, van Nieuwenhuizen P, Schreiber R. The selective 5-HT6 receptor antagonist Ro4368554 restores memory performance in cholinergic and serotonergic models of memory deficiency in the rat. Neuropsychopharmacology 30(12), 2169–2179 (2005).
  • Prickaerts J, Sik A, van der Staay FJ, de Vente J, Blokland A. Dissociable effects of acetylcholinesterase inhibitors and phosphodiesterase type 5 inhibitors on object recognition memory: acquisition versus consolidation. Psychopharmacology (Berl.) 177(4), 381–390 (2005).
  • Scali C, Giovannini MG, Bartolini L et al. Effect of metrifonate on extracellular brain acetylcholine and object recognition in aged rats. Eur. J. Pharmacol. 325(2–3), 173–180 (1997).
  • Hunsaker MR, Rogers JL, Kesner RP. Behavioral characterization of a transection of dorsal CA3 subcortical efferents: comparison with scopolamine and physostigmine infusions into dorsal CA3. Neurobiol. Learn. Mem. 88(1), 127–136 (2007).
  • Lindner MD, Hogan JB, Hodges DB Jr et al. Donepezil primarily attenuates scopolamine-induced deficits in psychomotor function, with moderate effects on simple conditioning and attention, and small effects on working memory and spatial mapping. Psychopharmacology (Berl.) 188(4), 629–640 (2006).
  • Romberg C, Mattson MP, Mughal MR, Bussey TJ, Saksida LM. Impaired attention in the 3xTgAD mouse model of Alzheimer’s disease: rescue by donepezil (Aricept). J. Neurosci. 31(9), 3500–3507 (2011).
  • Parsons CG, Stöffler A, Danysz W. Memantine: a NMDA receptor antagonist that improves memory by restoration of homeostasis in the glutamatergic system – too little activation is bad, too much is even worse. Neuropharmacology 53(6), 699–723 (2007).
  • Nyakas C, Granic I, Halmy LG, Banerjee P, Luiten PG. The basal forebrain cholinergic system in aging and dementia. Rescuing cholinergic neurons from neurotoxic amyloid-β42 with memantine. Behav. Brain Res. 221(2), 594–603 (2011).
  • Van Dam D, Abramowski D, Staufenbiel M, De Deyn PP. Symptomatic effect of donepezil, rivastigmine, galantamine and memantine on cognitive deficits in the APP23 model. Psychopharmacology (Berl.) 180(1), 177–190 (2005).
  • Van Dam D, Coen K, De Deyn PP. Cognitive evaluation of disease-modifying efficacy of donepezil in the APP23 mouse model for Alzheimer’s disease. Psychopharmacology (Berl.) 197(1), 37–43 (2008).
  • Van Dam D, De Deyn PP. Cognitive evaluation of disease-modifying efficacy of galantamine and memantine in the APP23 model. Eur. Neuropsychopharmacol. 16(1), 59–69 (2006).
  • Enz A, Gentsch C. Co-administration of memantine has no effect on the in vitro or ex vivo determined acetylcholinesterase inhibition of rivastigmine in the rat brain. Neuropharmacology 47(3), 408–413 (2004).
  • Wenk GL, Quack G, Moebius HJ, Danysz W. No interaction of memantine with acetylcholinesterase inhibitors approved for clinical use. Life Sci. 66(12), 1079–1083 (2000).
  • Zoladz PR, Campbell AM, Park CR, Schaefer D, Danysz W, Diamond DM. Enhancement of long-term spatial memory in adult rats by the noncompetitive NMDA receptor antagonists, memantine and neramexane. Pharmacol. Biochem. Behav. 85(2), 298–306 (2006).
  • Wise LE, Iredale PA, Stokes RJ, Lichtman AH. Combination of rimonabant and donepezil prolongs spatial memory duration. Neuropsychopharmacology 32(8), 1805–1812 (2007).
  • Wise LE, Lichtman AH. The uncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist memantine prolongs spatial memory in a rat delayed radial-arm maze memory task. Eur. J. Pharmacol. 575(1–3), 98–102 (2007).
  • Woodruff-Pak DS, Tobia MJ, Jiao X, Beck KD, Servatius RJ. Preclinical investigation of the functional effects of memantine and memantine combined with galantamine or donepezil. Neuropsychopharmacology 32(6), 1284–1294 (2007).
  • Yamada K, Takayanagi M, Kamei H et al. Effects of memantine and donepezil on amyloid b-induced memory impairment in a delayed-matching to position task in rats. Behav. Brain Res. 162(2), 191–199 (2005).
  • Martinez-Coria H, Green K, Banerjee PK, LaFerla FM. Combination of memantine and donepezil reverses cognitive deficits in transgenic mice with both amyloid-b plaques and neurofibrillary tangles. Poster presented at: 22nd Congress of the European College of Neuropsychopharmacology (ECNP). Istanbul, Turkey, 12–16 September 2009.
  • Neumeister KL, Riepe MW. Synergistic effects of antidementia drugs on spatial learning and recall in the APP23 transgenic mouse model of Alzheimer’s disease. J. Alzheimers Dis. 30(2), 245–251 (2012).
  • Gupta RC, Dekundy A. Memantine does not influence AChE inhibition in rat brain by donepezil or rivastigmine but does with DFP and metrifonate in in vivo studies. Drug Dev. Res. 64, 71–81 (2005).
  • Periclou AP, Ventura D, Sherman T, Rao N, Abramowitz WT. Lack of pharmacokinetic or pharmacodynamic interaction between memantine and donepezil. Ann. Pharmacother. 38(9), 1389–1394 (2004).
  • Yao C, Raoufinia A, Gold M et al. Steady-state pharmacokinetics of galantamine are not affected by addition of memantine in healthy subjects. J. Clin. Pharmacol. 45(5), 519–528 (2005).
  • Shua-Haim J, Smith J, Picard F et al. Steady-state pharmacokinetics of rivastigmine in patients with mild to moderate Alzheimer’s disease not affected by co-administration of memantine: an open-label, crossover, single-centre study. Clin. Drug Investig. 28(6), 361–374 (2008).
  • Porsteinsson AP, Grossberg GT, Mintzer J, Olin JT; Memantine MEM-MD-12 Study Group. Memantine treatment in patients with mild to moderate Alzheimer’s disease already receiving a cholinesterase inhibitor: a randomized, double-blind, placebo-controlled trial. Curr. Alzheimer Res. 5(1), 83–89 (2008).
  • Hartmann S, Möbius HJ. Tolerability of memantine in combination with cholinesterase inhibitors in dementia therapy. Int. Clin. Psychopharmacol. 18(2), 81–85 (2003).
  • Gauthier S, Molinuevo JL. Benefits of combined cholinesterase inhibitor and memantine treatment in moderate–severe Alzheimer’s disease. Alzheimers Dement. (2012) (In Press).
  • Feldman HH, Schmitt FA, Olin JT; Memantine MEM-MD-02 Study Group. Activities of daily living in moderate-to-severe Alzheimer disease: an analysis of the treatment effects of memantine in patients receiving stable donepezil treatment. Alzheimer Dis. Assoc. Disord. 20(4), 263–268 (2006).
  • Schmitt FA, van Dyck CH, Wichems CH, Olin JT; for the Memantine MEM-MD-02 Study Group. Cognitive response to memantine in moderate to severe Alzheimer disease patients already receiving donepezil: an exploratory reanalysis. Alzheimer Dis. Assoc. Disord. 20(4), 255–262 (2006).
  • van Dyck CH, Schmitt FA, Olin JT; Memantine MEM-MD-02 Study Group. A responder analysis of memantine treatment in patients with Alzheimer disease maintained on donepezil. Am. J. Geriatr. Psychiatry 14(5), 428–437 (2006).
  • Wilkinson D, Andersen HF. Analysis of the effect of memantine in reducing the worsening of clinical symptoms in patients with moderate to severe Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 24(2), 138–145 (2007).
  • Choi SH, Park KW, Na DL et al.; Expect Study Group. Tolerability and efficacy of memantine add-on therapy to rivastigmine transdermal patches in mild to moderate Alzheimer’s disease: a multicenter, randomized, open-label, parallel-group study. Curr. Med. Res. Opin. 27(7), 1375–1383 (2011).
  • Farlow MR, Alva G, Meng X, Olin JT. A 25-week, open-label trial investigating rivastigmine transdermal patches with concomitant memantine in mild-to-moderate Alzheimer’s disease: a post hoc analysis. Curr. Med. Res. Opin. 26(2), 263–269 (2010).
  • Schneider LS, Insel PS, Weiner MW; Alzheimer’s Disease Neuroimaging Initiative. Treatment with cholinesterase inhibitors and memantine of patients in the Alzheimer’s Disease Neuroimaging Initiative. Arch. Neurol. 68(1), 58–66 (2011).
  • Howard R, McShane R, Lindesay J et al. Donepezil and memantine for moderate-to-severe Alzheimer’s disease. N. Engl. J. Med. 366(10), 893–903 (2012).
  • Atri A, Shaughnessy LW, Locascio JJ, Growdon JH. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis. Assoc. Disord. 22(3), 209–221 (2008).
  • Lopez OL, Becker JT, Wahed AS et al. Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J. Neurol. Neurosurg. Psychiatry 80(6), 600–607 (2009).
  • Lyketsos CG, Miller DS; for the Neuropsychiatric Syndromes Professional Interest Area of the International Society to Advance Alzheimer’s Research and Treatment. Addressing the Alzheimer’s disease crisis through better understanding, treatment, and eventual prevention of associated neuropsychiatric syndromes. Alzheimers Dement. 8(1), 60–64 (2012).
  • Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol. 10(6), 501 (2011).
  • Birks J, Harvey RJ. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst. Rev. (1), CD001190 (2006).
  • Aricept® Product Information (PI). Eisai Ltd, NJ, USA, February 2012.
  • Birks J, Grimley Evans J, Iakovidou V, Tsolaki M, Holt FE. Rivastigmine for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD001191 (2009).
  • Loy C, Schneider L. Galantamine for Alzheimer’s disease and mild cognitive impairment. Cochrane Database Syst. Rev. (1), CD001747 (2006).
  • Brown FW. Cognitive enhancers (Chapter 41). In: The American Psychiatric Publishing Textbook of Psychopharmacology (4th Edition). Schatzberg AF, Nemeroff CB (Eds.). American Psychiatric Publishing, Arlington, VA, USA (2009).
  • Li J, Wu HM, Zhou RL, Liu GJ, Dong BR. Huperzine A for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD005592 (2008).
  • Ha GT, Wong RK, Zhang Y. Huperzine A as potential treatment of Alzheimer’s disease: an assessment on chemistry, pharmacology, and clinical studies. Chem. Biodivers. 8(7), 1189–1204 (2011).
  • Rafii MS, Walsh S, Little JT et al.; Alzheimer’s Disease Cooperative Study. A Phase II trial of huperzine A in mild to moderate Alzheimer disease. Neurology 76(16), 1389–1394 (2011).
  • Mancuso C, Siciliano R, Barone E, Butterfield DA, Preziosi P. Pharmacologists and Alzheimer disease therapy: to boldly go where no scientist has gone before. Expert Opin. Investig. Drugs 20(9), 1243–1261 (2011).
  • Hilt D, Gawryl M, Koenig G; EVP-6124 Study Group. EVP-6124: Safety, tolerability and cognitive effects of a novel α7 nicotinic receptor agonist in Alzheimer’s disease patients on stable donepezil or rivastigmine therapy. Alzheimers Dement. 5( Suppl. 4), e32 (2009).
  • Weinstock M, Luques L, Bejar C, Shoham S. Ladostigil, a novel multifunctional drug for the treatment of dementia co-morbid with depression. J. Neural Transm. Suppl. 70, 443–446 (2006).
  • Shioda N, Yamamoto Y, Han F, Moriguchi S, Fukunaga K. (Neurochemical mechanisms of a novel Alzheimer’s disease therapeutics on improvement of cognition and depressive behavior). Yakugaku Zasshi 131(4), 505–511 (2011).
  • Yamaguchi Y, Miyashita H, Tsunekawa H et al. Effects of a novel cognitive enhancer, spiro[imidazo-[1,2-alpha]pyridine-3,2-indan]-2(3H)-one (ZSET1446), on learning impairments induced by amyloid-β1-40 in the rat. J. Pharmacol. Exp. Ther. 317(3), 1079–1087 (2006).
  • Champix® Summary of Product Characteristics (SPC). Pfizer Ltd., April 2012.
  • Togashi H, Matsumoto M, Yoshioka M, Saito Y, Saito H. Effects of a novel cholinergic M1 agonist, AF102B, on ambulation and water drinking behavior in rats. Hokkaido Igaku Zasshi. 66(1), 59–66 (1991).
  • Terry AV Jr, Buccafusco JJ, Borsini F, Leusch A. Memory-related task performance by aged rhesus monkeys administered the muscarinic M(1)-preferring agonist, talsaclidine. Psychopharmacology (Berl.) 162(3), 292–300 (2002).
  • Adamus WS, Leonard JP, Tröger W. Phase I clinical trials with WAL 2014, a new muscarinic agonist for the treatment of Alzheimer’s disease. Life Sci. 56(11–12), 883–890 (1995).
  • Higgins JP, Flicker L. Lecithin for dementia and cognitive impairment. Cochrane Database Syst. Rev. (4), CD001015 (2000).
  • López-Arrieta JM, Rodríguez JL, Sanz F. Nicotine for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD001749 (2000).
  • Coelho F, Birks J. Physostigmine for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD001499 (2001).
  • López-Arrieta JM, Schneider L. Metrifonate for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD003155 (2006).
  • Birks J, Wilcock GG. Velnacrine for Alzheimer’s disease. Cochrane Database Syst. Rev. (2), CD004748 (2004).
  • McShane R, Areosa Sastre A, Minakaran N. Memantine for dementia. Cochrane Database Syst. Rev. (2), CD003154 (2006).
  • Ebixa® Summary of Product Characteristics (SPC). Lundbeck Ltd., October 2011.
  • Johnson SA, Simmon VF. Randomized, double-blind, placebo-controlled international clinical trial of the Ampakine CX516 in elderly participants with mild cognitive impairment: a progress report. J. Mol. Neurosci. 19(1–2), 197–200 (2002).
  • Jones R, Laake K, Øksengård AR. D-cycloserine for Alzheimer’s disease. Cochrane Database Syst. Rev. 2, CD003153 (2002).
  • Jones RW, Wesnes KA, Kirby J. Effects of NMDA modulation in scopolamine dementia. Ann. NY Acad. Sci. 640, 241–244 (1991).
  • Chappell AS, Gonzales C, Williams J, Witte MM, Mohs RC, Sperling R. AMPA potentiator treatment of cognitive deficits in Alzheimer disease. Neurology 68(13), 1008–1012 (2007).
  • Jones RW. Dimebon disappointment. Alzheimers. Res. Ther. 2(5), 25 (2010).
  • Narahashi T, Moriguchi S, Zhao X, Marszalec W, Yeh JZ. Mechanisms of action of cognitive enhancers on neuroreceptors. Biol. Pharm. Bull. 27(11), 1701–1706 (2004).
  • Ahmed AH, Oswald RE. Piracetam defines a new binding site for allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors. J. Med. Chem. 53(5), 2197–2203 (2010).
  • Flicker L, Grimley Evans J. Piracetam for dementia or cognitive impairment. Cochrane Database Syst. Rev. 1, CD001011 (2004).
  • Dantoine T, Auriacombe S, Sarazin M, Becker H, Pere JJ, Bourdeix I. Rivastigmine monotherapy and combination therapy with memantine in patients with moderately severe Alzheimer’s disease who failed to benefit from previous cholinesterase inhibitor treatment. Int. J. Clin. Pract. 60(1), 110–118 (2006).
  • Olin JT, Bhatnagar V, Reyes P, Koumaras B, Meng X, Brannan S. Safety and tolerability of rivastigmine capsule with memantine in patients with probable Alzheimer’s disease: a 26-week, open-label, prospective trial (Study ENA713B US32). Int. J. Geriatr. Psychiatry 25(4), 419–426 (2010).
  • Riepe MW, Adler G, Ibach B, Weinkauf B, Tracik F, Gunay I. Domain-specific improvement of cognition on memantine in patients with Alzheimer’s disease treated with rivastigmine. Dement. Geriatr. Cogn. Disord. 23(5), 301–306 (2007).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.