Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Feb 18.
Published in final edited form as: Curr Alzheimer Res. 2021;18(13):1010–1022. doi: 10.2174/1567205018666211215150547

Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer’s Disease: Nexus of Cholinergic and Nerve Growth Factor Dysfunction

Donald E Moss 1,*, Ruth G Perez 2,#
PMCID: PMC8855657  NIHMSID: NIHMS1773269  PMID: 34911424

Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is increasingly viewed as a complex multi-dimensional disease without effective treatments. Recent randomized, placebo-controlled studies have shown volume losses of ~0.7% and ~3.5% per year, respectively, in the cholinergic basal forebrain (CBF) and hippocampus in untreated suspected prodromal AD. One year of donepezil treatment reduced these annualized rates of atrophy to about half of untreated rates. Similar positive although variable results, have also been found in volumetric measurements of cortex and whole brain in patients with mild cognitive impairment as well as more advanced AD stages after treatments with all three currently available acetylcholinesterase (AChE) inhibitors (donepezil, rivastigmine, and galantamine). Here we review the anti-neurodegenerative benefits of AChE inhibitors and the expected parallel disease-accelerating impairments caused by anticholinergics, within a framework of the cholinergic hypothesis of AD and AD-associated loss of nerve growth factor (NGF). Consistent with the “loss of trophic factor hypothesis of AD”, we propose that AChE inhibitors enhance acetylcholine-dependent release and uptake of NGF, thereby sustaining cholinergic neuronal viability and thus slowing AD-associated degeneration of the CBF, to ultimately delay dementia progression. We propose that improved cholinergic therapies for AD started early in asymptomatic persons, especially those with risk factors, will delay the onset, progression, or emergence of dementia. The currently available competitive and pseudo-irreversible AChE inhibitors are not CNS-selective and thus induce gastrointestinal toxicity that limits cortical AChE inhibition to ~30% (ranges from 19% to 41%) as measured by in vivo PET studies in patients undergoing therapy. These levels of inhibition are marginal relative to what is required for effective symptomatic treatment of dementia or slowing AD-associated neurodegeneration. In contrast, because of the inherently slow de novo synthesis of AChE in the CNS (about one-tenth the rate of synthesis in peripheral tissues), irreversible AChE inhibitors safely produce significantly higher levels of inhibition in the CNS than in peripheral tissues. For example, methanesulfonyl fluoride, an irreversible inhibitor reduces CNS AChE activity by ~68% in patients undergoing therapy and ~80% in cortical biopsies of non-human primates. The full therapeutic benefits of AChE inhibitors, whether for symptomatic treatment of dementia or disease-slowing, thus would benefit by producing high levels of CNS inhibition. One way to obtain such higher levels of CNS AChE inhibition would be by using irreversible inhibitors.

Keywords: Alzheimer’s, acetylcholinesterase, EC 3.1.1.7, butyrylcholinesterase, EC 3.1.1.8, nerve growth factor, anticholinergic, acetylcholinesterase inhibitor, tauopathy

1. INTRODUCTION

AD is a devastating progressive degenerative brain disorder that affects 6.2 million in the U.S. and many millions more worldwide [1]. Since the first decade of the last century [2], AD has been primarily defined by pathology that includes senile plaques (now known to be accumulations of extracellular aggregated β-amyloid) and neurofibrillary tangles (now known to be intracellular aggregates of hyperphosphorylated tau) in postmortem brain. Despite decades of effort and hundreds of failed clinical trials mainly targeting β-amyloid, no effective prevention, disease modification, or treatment for AD has emerged [35]. The problem of finding an effective disease-modifying treatment may be due to increasing evidence that there are different subtypes of AD [6,7] that begin many years before the onset of dementia as shown by varying biomarkers including amyloidosis [8,9], tauopathy [7,10,11], inflammation [12,13], and AD-associated loss of NGF [14,15]. Regardless of the particular biomarkers or subtype, however, all AD is associated with a loss of CBF cholinergic function necessary for cognition [16].

In view of the complexity of AD, there is growing interest in developing treatments that target key downstream pathophysiological steps that will slow neurodegeneration, delay the onset of mild cognitive impairment (MCI), or prevent the transition from MCI to clinically diagnosed AD dementia [17,18]. One such treatment that targets a downstream step and is able to slow advancement through clinical stages of AD is long-term use of AChE (EC 3.1.1.7) inhibitors [1925]. Besides delaying clinical deterioration, AChE inhibitors also slow AD-associated atrophy in the CBF, hippocampus, cortex, and whole brain in prodromal (presymptomatic) AD [2628] and even in mild/moderate stages of dementia [2931]. Taken together, these findings strongly suggest that AChE inhibitors warrant further consideration as disease-modifying secondary prevention therapies [32,33]. The purpose of this review is to examine the anti-neurodegenerative benefits of AChE inhibition within a framework of cholinergic and NGF dysfunction during the early prodromal stages of AD [3437]. The hypothesis being explored is that AChE inhibitors stimulate acetylcholine-dependent release and uptake of NGF and thereby produce anti-neurodegenerative benefits in the CBF in AD.

2. THE CHOLINERGIC SYSTEM IN AD AND OTHER DEMENTING DISEASES

It has been long known that the hippocampus, neocortex, and amygdala accumulate high densities of neurofibrillary tangles and show a loss of the enzymes responsible for both the synthesis and metabolism of acetylcholine in AD [38]. There is specifically a severe loss of CBF neuronal cell body immunostaining, especially in the nucleus basalis of Meynert (nbM) [39], the main source of acetylcholine for limbic and neocortical areas [40,41]. The absence of acetylcholine in the neocortex, hippocampus, and related terminal field networks is the proximate cause of the severe cognitive loss in AD dementia [16,42]. Loss of CBF basal forebrain volume, at least in some cases, precedes and predicts the cortical spread of AD [43,44] and is associated with the development of MCI and AD [4549]. Magnetic resonance imaging (MRI) studies show that AD-associated neurodegeneration occurs in the CBF, hippocampus and neocortex in prodromal as well as in more advanced stages of the disease [9,2628,43,44,50,51]. There is also severe damage to the nbM in the related disorders of Lewy body dementia [52], Creutzfeldt-Jakob disease [53,54], and frontotemporal dementia [55], identifying other conditions in which improving acetylcholine levels would likely be beneficial.

3. CHOLINERGIC TONE AFFECTS THE RATE OF CLINICAL PROGRESSION IN AD

AChE inhibitors were originally introduced as symptomatic treatments for AD dementia to amplify the impact of the endogenous acetylcholine remaining in the neocortex and hippocampus to improve cognition, but with no expectation that they could affect the rate of AD progression [33,56,57]. However, multiple retrospective studies have confirmed that patients on long-term AChE inhibitor therapy experience slower advancement through increasingly more severe clinical stages of AD [1924]. For example, a matched cohort of ~17,000 AD patients followed for an average of 5 years showed that donepezil, rivastigmine, or galantamine use was associated with dose-dependent higher cognitive scores and slower cognitive decline as compared to nonusers. However, only galantamine, an AChE inhibitor that is also an allosteric potentiating ligand for nicotinic acetylcholine receptors (nAChR) [58], reduced the risk of advancing to severe dementia [25]. Also, as expected from their opposing pharmacological mode of action, long-term use of CNS-penetrating anticholinergics are associated with increased AD incidence, accelerated progression from normal cognition to MCI, and a higher risk of advancing from MCI to outright dementia [5967]. Taken together, these data indicate that increasing normal cholinergic tone (via AChE inhibition) or deceasing cholinergic tone (via anticholinergics) slow or accelerate, respectively, the clinical progression of AD [33,68], demonstrating the key role of the cholinergic system in AD.

4. AChE AND BChE (EC 3.1.1.8) INHIBITORS SLOW CNS GRAY AND WHITE MATTER ATROPHY IN AD

CNS atrophy is a major biomarker of AD [69], with whole brain atrophy progressing at 2.3%/year in AD patients as compared to 0.4%/year in healthy age matched controls [70]. Hippocampal atrophy also occurs in normal aging but is also an early biomarker of AD [70,71], with hippocampal volumes in AD patients being consistently less than those in healthy controls [70,7274]. The rate of hippocampal atrophy is greater and more variable in AD than in healthy controls, with means ranging from up to 15%/year vs 1.5%/year, respectively [74]. The rate of AD-associated atrophy of the hippocampus and cortex is also higher in APOE ε4 carriers [31,75] and MCI patients who often subsequently convert to AD [76].

4.1. Anti-neurodegenerative Benefits of Selective AChE Inhibition on Gray Matter

AChE inhibitors slow CNS atrophy in prodromal as well as in more advanced stages of disease. In suspected prodromal AD patients, randomized placebo-controlled trials (RCT) of short-term (1 year) treatment with donepezil, an AChE-selective inhibitor, produced ~60% reduction in the rate of basal forebrain CBF atrophy as compared to placebo-treated controls [27]. Donepezil also produced a similar ~55% reduction in the rate of hippocampal atrophy [28] and a nonsignificant trend toward reducing the rate of neocortex thinning [26] (Table 1). In MCI patients, RCT’s of donepezil (48 weeks) [33] or galantamine (two years), both of which are AChE selective inhibitors, failed to show benefit in the hippocampus [30], although both treatments reduced whole brain atrophy (Table 1). In patients in the galantamine trial, slowing of whole brain atrophy was limited to those who were APOE ε4 positive [30]. However, in patients with mild to moderate AD, short-term donepezil (24 weeks) produced a slowing of hippocampal atrophy [29] (Table 1). These results show that AChE inhibitor therapy has anti-neurodegenerative benefits in the CBF that sometimes extend to its gray matter postsynaptic projection areas in both prodromal and more advanced stages of AD.

Table 1.

Anti-Neurodegenerative Effects of AChE Inhibitors in Prodromal AD by Structure as Compared to MCI and Mild/moderate AD

Dementia Stage Brain Structure Duration Drug1 Annualized % Change - Control Annualized % Change - Drug Drug Benefit (Annualized %) Author/Year
Prodromal Basal Forebrain 1 yr DNZ
10 mg/d
0.74%
N=88
0.30%
N=75
+0.44% p=0.008 Cavedo et al. (2017) [27]
Prodromal Hippocampus 1 yr DNZ
10 mg/d
3.47%
N=92
1.89%
N=82
+1.58 % p<0.001 Dubois et al. (2015) [28]
Prodromal Cortex (mean) 1 yr DNZ
10 mg/d
−1.01% (SEM .25%) N=92 +0.075% (SEM 0.23%) N=82 +1.08%
(see note below2)
Cavedo et al. (2016) [26]
MCI Hippocampus 48 wk DNZ
10 mg/d
−2.23% N=125 −1.86% N=105 +0.37%
p=0.446 ns
Schuff et al. (2011) [31]
Entorhinal cortex −3.00% N=112 −0.85% N=100 +2.15%
p=0.33 ns
Whole brain 0.54%
N=90
+0.01%
N=74
+0.55% p<0.001
MCI Hippocampus 2 yr GAL
16–24 mg/d
−1.70% N=160 −1.94% N=142 −0.24%
ns
Prins et al. (2014) [30]
Whole brain 0.63%
N=130
0.46%
N=112
+0.17% p<0.05
Mild/moderate Hippocampus 24 wk DNZ
10 mg/d
17.7%
N=31
0.87%
N=32
+16.8% p=0.01 Krishnan et. al. (2003) [29]
1

DNZ = donepezil, GAL = galantamine.

2

Note: Significant drug effects shown in bold. Cavedo et al. [26] shows mean of seven neocortical areas, drug vs. placebo comparisons in all individual areas p<0.05 (same patients studied for hippocampal atrophy by Dubois et al. [28]).

4.2. Anti-neurodegenerative Benefits of Dual AChE and BChE Inhibition on White Matter

In contrast to gray matter (Table 1), the anti-neurodegenerative benefits seen in white matter may not be due entirely to AChE inhibition but may also involve BChE inhibition. For example, a 3 to 4-year retrospective analysis of rivastigmine, a mixed inhibitor of both AChE and BChE, showed reduced white matter loss and less ventricle enlargement in women, but not men, although there was no effect noted on hippocampal volumes [77]. A 20-week experiment comparing the anti-neurodegenerative effects of rivastigmine, donepezil, and galantamine in 30 patients with mild-to-moderate AD showed that only rivastigmine, but not the AChE-selective inhibitors donepezil or galantamine, preserved both neocortical white matter and gray matter [78]. The anti-neurodegenerative effect of rivastigmine in white matter is thus attributed to both AChE and BChE inhibition [79], both of which are involved in cholinergic signaling in the myelination of neurons [80]. AChE and BChE also impact muscarinic and nicotinic acetylcholine receptors that are expressed on myelinating oligodendroglia, and acetylcholine-dependent intracellular signaling that occurs in these myelinating glia [80]. BChE inhibition may thus be a useful therapeutic target in the treatment of AD [81].

5. CEREBROSPINAL FLUID (CSF) BIOMARKERS SUGGEST THAT ACHE INHIBITION MAY PRODUCE ANTI-NEURODEGENERATIVE BENEFITS BY STIMULATING CNS REGENERATION

Sustained, although sometimes enigmatic, changes in CSF cholinergic markers include increases and decreases in both AChE and BChE enzyme protein levels and enzymatic activities after long-term AChE inhibitor therapy. For example, patients treated with the readily reversible (competitive) AChE inhibitors donepezil and galantamine show paradoxical increases in CSF AChE activity [8284]. On the other hand, treatment with the pseudo-irreversible inhibitor rivastigmine (dual AChE and BChE inhibitor) results in decreases in CSF AChE activity [8385], but no change [84] or sometimes decreased levels of [83,85] BChE activity.

An increase or no change in CSF enzyme activities [8285] is contrary to the a priori expectation that cholinesterase inhibitors would decrease AChE and/or BChE activity. However, the 20% to 35% increase in CSF AChE activity that followed long-term donepezil treatment was concurrently accompanied by 120% to 160% increases in CSF AChE protein content. The increase in AChE protein was initially interpreted as a compensatory upregulation of enzyme synthesis as a response to AChE inhibition [86]. However, in vivo PET scans of AD patients treated with galantamine show 30% to 36% inhibition of CNS AChE activity that is accompanied by an increase in the activity of CSF choline acetyltransferase (ChAT), the intracellular enzyme responsible for acetylcholine synthesis that is a biomarker of cholinergic neurons [87]. It is interesting to note that the galantamine treatment also resulted in a parallel increase in synaptic nAChR binding sites in most regions of the AD brain [87], a finding that is also reported after rivastigmine treatment [88] and after chronic donepezil or galantamine treatment in the brains of old rats [89]. Thus, changes in cholinergic neuronal and synaptic markers induced by AChE inhibitors suggest that they likely cause generalized synaptic regeneration, especially of nicotinic cholinoceptive neurons, as well as improved CNS cholinergic signaling [87].

6. THE INTERSECTION OF THE CHOLINERGIC AND NGF TROPHIC FACTOR HYPOTHESES IN THE BASAL CHOLINERGIC FOREBRAIN

The AChE inhibitor-induced increases in CNS nAChR binding [8789] and CSF ChAT activity [87] reviewed above suggest that AChE inhibitor-induced anti-neurodegenerative benefits might be due, at least in part, to CBF nicotinic cholinergic stimulation that has been shown to upregulate NGF-related genes and prolong NGF release after direct administration of nicotine [90]. NGF levels in the hippocampus are also selectively upregulated in response to nicotinic cholinergic stimulation [91]. Therefore the purpose of the following section is to examine the reciprocal relationship between cholinergic modulation of NGF and the essential role of NGF the maintenance of CBF neurons, especially as related to AD-associated loss of NGF, cholinergic dysfunction in AD, and AChE inhibitor-induced anti-neurodegenerative benefits in AD.

6.1. Trophic Factors in the Normal Maintenance of CNS Neurons and Dysfunction in Disease

As early as 1981 it was proposed that maintenance of certain neuronal cell bodies depends on stimulating the tissues that receive their projections (target tissues) to release neurotrophic hormones (factors) that then undergo retrograde transport to maintain the viability of presynaptic neuronal cell bodies and it is the loss of these factors that result in neurodegeneration [92]. In accordance with this hypothesis, basal forebrain atrophy in rats, monkeys, and humans following cortical damage [93,94], disease [53,54], normal aging, and in various neurodegenerative disorders [9597] is attributed to a loss of NGF retrograde transport. An example showing that neurotrophic support is essential for the survival of specific neuronal cell bodies in neurodegenerative disease is also found in Huntington’s disease, where the aberrant microtubule-associated protein, huntingtin, appears to be solely responsible for interfering with microtubule-dependent retrograde transport of brain derived neurotrophic factor (BDNF) and the specific pattern of striatal atrophy characteristic of Huntington’s disease [98]. In a parallel example relevant to AD, phospho-tau is associated with impaired microtubule-dependent NGF/TrkA retrograde signaling, reduced NGF trophic support, and reduced axonal trafficking-mediated support of CBF neurons in aged rats [99]. It is tempting to speculate that AD-associated phospho-tau may be responsible for impaired microtubule-dependent NGF/TrkA retrograde transport [34] and the AD-specific pattern of CBF basal forebrain atrophy [100]. Such a possibility is consistent with the early appearance of AD-associated patterns of aberrant tau [101] and the resulting progression of AD neurodegeneration. In any event, NGF metabolic pathways are known to be seriously dysregulated in both AD and Down’s syndrome [96].

6.2. NGF and Basal Forebrain Cholinergic Neuron Survival

As shown in (Fig. 1), acetylcholine (ACh) from cholinergic basal forebrain (CBF) projections stimulates activity-dependent synthesis and release of the NGF precursor protein (proNGF) in the neocortex and hippocampus, where it then undergoes extracellular conversion to mature NGF (mNGF) in the synapse [14,37,90,91,102104]. Mature mNGF is then taken up into presynaptic terminals after binding to TrkA followed by its uptake into endosomes for microtubule-dependent retrograde transport of mNGF to CBF cell bodies [14,37,102,104107]. Survival of CBF neuronal cells depends on this retrograde transport of NGF and its subsequent actions on the abundant NGF receptors localized to cholinergic neurons in the basal forebrain in both nonhuman primates [108] and humans [109,110]. NGF receptors in the neocortex are mainly limited to CBF projection areas [108,110]. The specific distribution of NGF and its receptors demonstrate a precise interrelationship between CBF neurons and their NGF-producing neocortical and hippocampal targets, areas that are both severely damaged in AD [110113].

Fig. (1).

Fig. (1).

An illustration of a non-exhaustive list of AD-associated changes to CBF/NGF functions that support direct enhancement of NGF as a therapeutic strategy in AD [37]. As also reviewed elsewhere [15,37] these pathophysiological changes include impaired conversion of proNGF to mNGF (I in Fig. 1C) [114], a failure that causes: proNGF accumulation, reduced mNGF levels, [115], an imbalance in TrkA/p75 receptor ratio (II in Fig. 1C), impaired retrograde transport of NGF (III in Fig. 1C), and an increase in inflammation due to the loss of acetylcholine-mediated-stimulation of glial cell anti-inflammatory effects (IV in Fig. 1C) [37,116]. (Created using Biorender.com online software).

6.3. Anti-Neurodegenerative Benefits of AChE Inhibitors May Occur in Response To Acetylcholine-dependent Stimulation of NGF, Likely Mediated at Least in Part, by Nicotinic Cholinergic Receptors

It has long been appreciated that the loss of the CBF nbM is attributed to an AD-associated failure of NGF retrograde transport [117] that occurs during AD progression [15,34], including NGF-related changes that begin during prodromal AD stages [35,36,118]. The loss of NGF support is known to cause CBF neurons to lose their cholinergic phenotype yet still persist, but just in an atrophied state [119,120], which provides an opportunity to re-establish their normal functions if NGF levels can be restored. Therefore, we hypothesize that the anti- neurodegenerative benefits of long-term AChE inhibitor treatments [2630] likely also result from direct acetylcholine-dependent-stimulation of NGF-induced regeneration as well as AChE inhibitor-induced increases in CNS nAChR upregulation [8891].

6.4. The NGF/Cholinergic Hypothesis is not Exclusive of Other Pathophysiological Processes

AChE inhibition may exert anti-neurodegenerative effects through cholinergic modulation of AD- associated pathophysiological mechanisms other than NGF. For example, there are other complex interactions that occur between CBF cholinergic neurons, microglia and astrocytes that stimulate anti- inflammatory pathways (Figure 1C) [37,114,116,] that are mediated, at least in part, by α7-nAChRs [37,121]. In addition, AChE inhibition has more general effects, such as increasing regional cerebral blood flow and functional connectivity in cholinergic pathways in AD [122], which may directly or indirectly affect other pathophysiological processes like glucose hypometabolism and dysmyelination that occur in AD [123]. These various alternative mechanisms support the possibility that AChE inhibition affects AD through more than one pathway.

7. LIMITATIONS OF CURRENT ACHE INHIBITOR THERAPY

The main barrier to obtaining the full clinical benefit of AChE inhibitors in AD, whether for symptomatic treatment or anti-neurodegenerative benefit, is the inadequate level of CNS inhibition produced by the currently approved inhibitors [32]. Donepezil, galantamine, and rivastigmine produce a range of only 19% to 41% inhibition of neocortical AChE in AD patients undergoing therapy [124128] (Table 2), levels that are marginal compared to the need for much greater than ~40% AChE inhibition that is required for optimal efficacy [129,130]. This is because they are not CNS-selective and, thus induce peripheral AChE inhibitor toxicity, especially in the gastrointestinal tract, which prevents the higher dosing essential for full cognitive or anti-neurodegenerative benefits [129,130]. Another inhibitor, metrifonate, a short-acting pseudo-irreversible AChE inhibitor [131,132], was also dose-limited by gastrointestinal toxicity, but its development was discontinued because it also caused respiratory depression and organophosphate-induced delayed neuropathy [133]. In summary, a need for higher levels of CNS AChE inhibition for treating AD [32,129,130] and the finding of a positive relationship between dose and cognitive benefit [25] strongly suggest that the real benefits of AChE inhibitors for symptomatic and anti-neurodegenerative success in AD has only begun to be explored because of the dose-limiting gastrointestinal toxicity caused by the currently available short-acting drugs [32] (Table 2). This has led to a call to identify more robust CNS cholinergic therapies [32,33] as will be described below.

Table 2.

CNS AChE Inhibition in AD patients with current therapies.

Author/Year Drug1/Dose Duration/Number of Subjects (N) Structure Mean Inhibition
+/− SD2
Kuhl et al. (2000) [126] DNZ
10 mg/d
5 wk (N=6) Cerebral cortex 27% +/− 11% **
Kaasinen et al. (2002) [125] DNZ
10 mg/d
3 mo (N=6) Frontal Temporal Parietal 39% +/− 5.9% **
29% +/− 11% *
28% +/− 17% *
RIV
9 mg/d
3–5 mo (N=5) Frontal Temporal Parietal 37% +/− 7% **
28% +/− 16% *
28%+/− 18% ns
Bohnen et al. (2005) [124] DNZ
10 mg/d
12 wk (N=14) Mean cortical 19% +/− 9% ***
Kadir et al. (2008) [127] GAL
16–24 mg/d
3 mo (N=10)
12 mo (N=11)
Mean cortical 36% +/− 5% ***
41% +/− 6% ***
Ota et al. (2010) [128] DNZ
5 mg/d
5 mo (N=16) Mean cortical 36% +/− 7% ***
1

DNZ = donepezil, RIV = rivastigmine, GAL = galantamine.

2

*p<0.5, **p<0.01, ***p<0.001 inhibition compared to no inhibitor condition.

In contrast to the competitive and pseudo-irreversible AChE inhibitors described above, very high levels of CNS inhibition can be produced by irreversible AChE inhibition [32]. This type of inhibition dissipates only as new AChE enzyme is synthesized in each affected tissue [134]. The major advantage of irreversible inhibition, as illustrated below (Figure 2), is that de novo replacement of AChE in the CNS occurs at only about one-tenth the rate at which it occurs in peripheral tissues [132,134]. The naturally slow rate of CNS enzyme replacement thus results in the accumulation of very high levels of CNS AChE inhibition after repeated dosing (Figure 2) like that which would be used in long-term treatments for AD [32,134,135]. The stark difference in CNS AChE inhibition expected from a short-acting AChE inhibitor versus an irreversible inhibitor is demonstrated in Figure 2.

Fig. (2).

Fig. (2).

A side-by-side computational comparison of expected competitive versus irreversible AChE inhibition through 21 days of treatment. This figure shows the expected accumulated AChE inhibition in the brain (upper red line) vs the intestine (lower blue line) after three weeks of daily doses of a competitive inhibitor. Panel A: competitive inhibitor computations based on donepezil. The upward saw-tooth appearance of the lines show 25% inhibition in the CNS versus 20% inhibition in the intestine produced by each daily dose, a ratio of 1.25 selectivity toward the CNS. The downward slope between doses shows spontaneous recovery from donepezil-induced competitive inhibition with a half-time of 76 hours. Panel B: irreversible inhibitor computations based on methanesulfonyl fluoride. The upward saw-tooth appearance of the lines shows an equal increment of 10% inhibition of remaining active AChE in both intestine and CNS with each daily dose. The downward slope between doses is the reduction of AChE inhibition that is produced by new enzyme synthesis that occurs with a half-time of ~12 days in the CNS and a half-time of ~1 day in the intestine [134]. The separation between the accumulated AChE inhibition in CNS and intestines shown in Panel B is due entirely to the slow rate of de novo synthesis of enzyme protein in the CNS versus the peripheral tissues (from Moss et al. [32], with permission, see acknowledgements).

The hypothetical advantage of using an irreversible inhibitor to produce high CNS versus limited peripheral tissue AChE inhibition (Figure 2) has been tested in rats treated three times per week with methanesulfonyl fluoride for 3 weeks, a schedule that simulates actual clinical use of the same compound [135,136]. At 3 weeks, the animals were sacrificed and AChE activity was assayed in the CNS and peripheral tissues [135]. The finding of ~75% CNS AChE inhibition in this rat study (Figure 3) exceeded the expected ~65% shown in Figure 2B. Also in general agreement with the expected results shown in Figure 2B, AChE inhibition in the peripheral tissues remained at or below ~20% and did not induce toxicity. Our results shown in Figure 3 further confirm the high inherent CNS selectivity of irreversible AChE inhibitors is due to the inherent slow de novo protein synthesis of that enzyme in the CNS, which is only about one-tenth the rate of enzyme replacement found in peripheral tissues [134,135].

Fig. (3).

Fig. (3).

AChE Inhibition accumulated in rat gastrointestinal smooth muscle (ileum), skeletal muscle (pectoral muscle), and heart (cardiac muscle) versus CNS (whole brain) after three weeks of treatment with methanesulfonyl fluoride. ** Brain AChE inhibition (shown in red) is significantly more inhibited than peripheral tissues (shown in blue), while peripheral tissues are not different from each other. The error bars show the standard error of the mean. (from Moss et al. [135] with permission, see acknowledgements).

Comparable experiments in humans and nonhuman primates are consistent with the CNS AChE inhibition shown in Figure 3. In AD patients, methanesulfonyl fluoride produced ~70% CNS AChE inhibition as estimated by measuring erythrocyte AChE activity [136], an estimate that was later confirmed in cortical biopsies of nonhuman primates that showed ~80% AChE inhibition [32]. As expected from the high selectivity toward CNS AChE inhibition and minimal inhibition in peripheral tissues, neither humans nor nonhuman primates showed troublesome gastrointestinal toxicity or any other noticeable adverse effects [32,135,136]. Taken together, these experiments show that when freed from the barrier of dose-limiting gastrointestinal toxicity, high-level AChE inhibition (>50%) can be obtained and be well tolerated. The high CNS selectivity and low peripheral toxicity shown for methanesulfonyl fluoride would also be expected if using any safe irreversible AChE inhibitor [32]. Thus, irreversible inhibition is one strategy for producing the robust cholinergic enhancement needed to test the limits of AChE inhibitor-induced anti-neurodegenerative benefits [32,33,137].

8. DISCUSSION AND FUTURE DIRECTIONS

Successful disease-modifying interventions that slow AD-associated CNS atrophy can significantly mitigate the economic impact and suffering in AD [17,18,138]. For example, a hypothetical intervention initiated in 2025 that could delay dementia onset by only 5 years could reduce the estimated 13.5 million with AD in 2050 by about half to 7.8 million or even fewer [139]. Similarly, a hypothetical treatment that does not affect AD onset, but slows the progression to more advanced stages of dementia, would reduce the numbers of patients with severe dementia by ~80%, from 6.5 million to 1.2 million by 2050 [139].

Even in the face of the limited CNS inhibition produced by the currently approved AChE inhibitors (Table 2), they do have modest anti-neurodegenerative benefits in the basal forebrain and hippocampus (Table 1). It thus seems likely that if they were started up to two decades before onset of cognitive decline when the first signs of aberrant tau [10], β-amyloid or hippocampal atrophy [9] begin, they could significantly delay the onset of dementia or slow cognitive decline to more advanced stages of dementia. Such prophylactic use of the currently approved AChE inhibitors thus might be worthwhile for treating cognitively normal elderly, especially those who are at risk of AD like those with familial history, APOE ε4 carriers [18], or with abnormal CSF biomarkers linked to accelerated AD progression associated with β-amyloid or tauopathy [50].

Potential prophylactic use of AChE inhibitors in persons without an existing need for symptomatic treatment may, however, introduce ethical and other questions. Short-term tests of AChE inhibitors in cognitively normal healthy older subjects have been shown to produce no significant cognitive disturbance and only minor, if any, other effects [135,140,141]. The potential for AChE inhibitor use in cognitively normal persons thus requires further risk/benefit consideration [142].

Other cholinergic drugs, in addition to AChE inhibitors, are potential preventative agents against AD. For example, both nAChR and muscarinic acetylcholine (mAChR) receptor agonists, much like AChE inhibitors, affect a wide variety of AD-associated pathophysiological processes [80,116,143,144]. Both muscarinic and nicotinic receptor agonists also provide neuroprotection for the cholinergic system and thus are proposed to be rational therapeutic targets for AD [90,91,145147].

SUMMARY AND CONCLUSION

AChE inhibitors produce neuroprotective/regenerative benefits as shown by slowing AD-associate atrophy of the basal forebrain and hippocampus (Table 1) and stimulate signs of CNS regeneration as shown by CSF biomarkers [87]. It is hypothesized that AChE-induced anti-neurodegenerative benefits are likely to be due to acetylcholine-dependent stimulation of NGF-induced protection of CBF cholinergic neurons [33,36,37], a possibility that deserves further study. Regardless of the mechanism of action, the consistent benefits of AChE inhibitor therapy reviewed here offer unparalleled hope as compared to the prior failed clinical trials of other agents [4,5]. Moreover, the prolonged presymptomatic period that exists in AD [9,10] offers ample opportunity for interventions with benefits compounded over several years, or decades. AChE inhibitors, especially those that yield high level CNS AChE inhibition [32,137], thus offer a new frontier for delaying the onset or slowing the relentless progression of AD.

ACKNOWLEDGEMENTS

Figure 2 is reprinted from Journal of Alzheimer’s Disease, 55, Moss et al., Cholinesterase Inhibitor Therapy in Alzheimer’s Disease: The Limits and Tolerability of Irreversible CNS-Selective Acetylcholinesterase Inhibition in Primates, pages 1285-1294, copyright (2017) with permission from IOS Press. The publication is available at IOS Press through http://dx.doi.org/10.3233/JAD-160733.

Figure 3 is reprinted from the British Journal of Clinical Pharmacology, Moss et al., 2013 [135], with permission of Wiley Publications.

Suggestions from Louis N. Irwin, Ph.D., and the bibliographic assistance from Mr. Eduardo Oropeza-Sánchez, University of Texas at El Paso Library, are gratefully acknowledged.

FUNDING

Preparation of this work was supported, in part by National Institute on Minority Health and Health Disparities Grant # 5U54MD007592 (NIMHD), a component of the National Institutes of Health (NIH). The sponsors had no role in the design and conduct of the study; in the collection, analysis, and interpretation of data; in the preparation of the manuscript; or in the review or approval of the manuscript.

Footnotes

CONFLICT OF INTEREST

Dr. Moss and Dr. Perez declare no conflict of interest related to the content of this manuscript.

REFERENCES

  • [1].Alzheimer’s Association. Alzheimer’s disease facts and figures 2021. Alzheimer’s Dement 2021;17:327–406. 10.1002/alz.12328. [DOI] [PubMed] [Google Scholar]
  • [2].Stelzmann RA, Norman Schnitzlein H, Reed Murtagh F. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige erkankung der hirnrinde.” Clin Anat 1995;8:429–31. 10.1002/ca.980080612. [DOI] [PubMed] [Google Scholar]
  • [3].Bachurin SO, Bovina EV., Ustyugov AA. Drugs in clinical trials for Alzheimer’s disease: The major trends. Med Res Rev 2017;37:1186–225. 10.1002/med.21434. [DOI] [PubMed] [Google Scholar]
  • [4].Cummings JL, Morstorf T, Zhong K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimer’s Res Ther 2014;6:37. 10.1186/alzrt269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Mehta D, Jackson R, Paul G, Shi J, Sabbagh M, Network VH, et al. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin Investig Drugs 2018;26:735–9. 10.1080/13543784.2017.1323868.Why. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Ferreira D, Nordberg A, Westman E. Biological subtypes of Alzheimer disease: A systematic review and meta-analysis. Neurology 2020;94:436–48. 10.1212/WNL.0000000000009058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Vogel JW, Young AL, Oxtoby NP, Smith R, Ossenkoppele R, Strandberg OT, et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat Med 2021;27(5):871–81. 10.1038/s41591-021-01309-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Nixon RA. Amyloid precursor protein and endosomal-lysosomal dysfunction in Alzheimer’s disease: Inseparable partners in a multifactorial disease. FASEB J 2017;31:2729–43. 10.1096/fj.201700359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Villemagne VL, Burnham S, Bourgeat P, Brown B, Ellis KA, Salvado O, et al. Amyloid β deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: A prospective cohort study. Lancet Neurol 2013;12:357–67. 10.1016/S1474-4422(13)70044-9. [DOI] [PubMed] [Google Scholar]
  • [10].Sanchez JS, Becker JA, Jacobs HIL, Hanseeuw BJ, Jiang S, Schultz AP, et al. The cortical origin and initial spread of medial temporal tauopathy in Alzheimer’s disease assessed with positron emission tomography. Sci Transl Med 2021;13(577):eabc0655. 10.1126/scitranslmed.abc0655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Franzmeier N, Neitzel J, Rubinski A, Smith R, Strandberg O, Ossenkoppele R, et al. Functional brain architecture is associated with the rate of tau accumulation in Alzheimer’s disease. Nat Commun 2020;11(1):347. 10.1038/s41467-019-14159-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].McGeer PL, Rogers J, McGeer EG. Inflammation, Antiinflammatory agents, and Alzheimer’s disease: The last 22 Years. J Alzheimer’s Dis 2016;54:853–7. 10.3233/JAD-160488. [DOI] [PubMed] [Google Scholar]
  • [13].Potter H, Woodcock JH, Boyd TD, Coughlan CM, O’Shaughnessy JR, Borges MT, et al. Safety and efficacy of sargramostim (GM-CSF) in the treatment of Alzheimer’s disease. Alzheimer’s Dement Transl Res Clin Interv 2021;7(1):e12158. 10.1002/trc2.12158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Cuello AC, Bruno MA, Allard S, Leon W, Iulita MF. Cholinergic involvement in Alzheimer’s disease. A link with NGF maturation and degradation. J Mol Neurosci 2010;40:230–5. 10.1007/s12031-009-9238-z. [DOI] [PubMed] [Google Scholar]
  • [15].Mufson EJ, Counts SE, Ginsberg SD, Mahady L, Perez SE, Massa SM, et al. Nerve growth factor pathobiology during the progression of Alzheimer’s disease. Front Neurosci 2019;13:533. 10.3389/fnins.2019.00533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018;141:1917–33. 10.1093/brain/awy132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Carrillo MC, Brashear HR, Logovinsky V, Ryan JM, Feldman HH, Siemers ER, et al. Can we prevent Alzheimer’s disease? Secondary “prevention” trials in Alzheimer’s disease. Alzheimer’s Dement 2013;9:123–131.e1. 10.1016/j.jalz.2012.12.004. [DOI] [PubMed] [Google Scholar]
  • [18].Sperling R, Mormino E, Johnson K. The evolution of preclinical Alzheimer’s disease: Implications for prevention trials. Neuron 2014;84:608–22. 10.1016/j.neuron.2014.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Atri A, Shaughnessy LW, Locascio JJ, Growdon JH. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord 2008;22:209–21. 10.1097/WAD.0b013e31816653bc. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Frankfort SV, Appels BA, De Boer A, Tulner LR, Campen JPCM, Koks CHW, et al. Treatment effects of rivastigmine on cognition, performance of daily living activities and behaviour in Alzheimer’s disease in an outpatient geriatric setting. Int J Clin Pract 2006;60:646–82. 10.1111/j.1368-5031.2006.00970.x. [DOI] [PubMed] [Google Scholar]
  • [21].Gauthier S, Molinuevo JL. Benefits of combined cholinesterase inhibitor and memantine treatment in moderate-severe Alzheimer’s disease. Alzheimer’s Dement 2013;9:326–31. 10.1016/j.jalz.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • [22].Lopez OL, Becker JT, Wahed AS, Saxton J, Sweet RA, Wolk DA, et al. Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J Neurol Neurosurg Psychiatry 2009;80:600–7. 10.1136/jnnp.2008.158964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Rountree SD, Atri A, Lopez OL, Doody RS. Effectiveness of antidementia drugs in delaying Alzheimer’s disease progression. Alzheimer’s Dement 2013;9:338–45. 10.1016/j.jalz.2012.01.002. [DOI] [PubMed] [Google Scholar]
  • [24].Scarpini E, Bruno G, Zappalà G, Adami M, Richarz U, Gaudig M, et al. Cessation versus continuation of galantamine treatment after 12 months of therapy in patients with alzheimer’s disease: A randomized, double blind, placebo controlled withdrawal trial. J Alzheimer’s Dis 2011;26:211–20. 10.3233/JAD-2011-110134. [DOI] [PubMed] [Google Scholar]
  • [25].Xu H, Garcia-Ptacek S, Jönsson L, Wimo A, Nordström P, Eriksdotter M. Long-term Effects of Cholinesterase Inhibitors on Cognitive Decline and Mortality. Neurology 2021;96:e2220–30. 10.1212/WNL.0000000000011832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Cavedo E, Dubois B, Colliot O, Lista S, Croisile B, Tisserand GL, et al. Reduced regional cortical thickness rate of change in donepezil-treated subjects with suspected prodromal Alzheimer’s disease. J Clin Psychiatry 2016;77:e1631–8. [DOI] [PubMed] [Google Scholar]
  • [27].Cavedo E, Grothe MJ, Colliot O, Lista S, Chupin M, Dormont Di, et al. Reduced basal forebrain atrophy progression in a randomized donepezil trial in prodromal Alzheimer’s disease. Sci Rep 2017;7:1–10. 10.1038/s41598-017-09780-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Dubois B, Chupin M, Hampel H, Lista S, Cavedo E, Croisile B, et al. Donepezil decreases annual rate of hippocampal atrophy in suspected prodromal Alzheimer’s disease. Alzheimer’s Dement 2015;11:1041–9. 10.1016/j.jalz.2014.10.003. [DOI] [PubMed] [Google Scholar]
  • [29].Krishnan KR., Cecil Charles MH, Murali Doraiswamy P, Jacobo Mintzer M, Weisler R, Yu X, et al. Randomized, placebo-controlled trial of the effects of donepezil on neuronal markers and hippocampal volumes in Alzheimer’s disease. Am J Psychiatry 2003;160:2003–11. [DOI] [PubMed] [Google Scholar]
  • [30].Prins ND, Van Der Flier WA, Knol DL, Fox NC, Brashear HR, Nye JS, et al. The effect of galantamine on brain atrophy rate in subjects with mild cognitive impairment is modified by apolipoprotein E genotype: Post-hoc analysis of data from a randomized controlled trial. Alzheimer’s Res Ther 2014;6(4):47. 10.1186/alzrt275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Schuff N, Suhy J, Goldman R, Xu Y, Sun Y, Truran-Sacrey D, et al. An MRI substudy of a donepezil clinical trial in mild cognitive impairment. Neurobiol Aging 2011;32(12):2318.e31–41. 10.1016/j.neurobiolaging.2010.04.005. [DOI] [PubMed] [Google Scholar]
  • [32].Moss DE, Perez RG, Kobayashi H. Cholinesterase inhibitor therapy in Alzheimer’s disease: The limits and tolerability of irreversible CNS-selective acetylcholinesterase inhibition in primates. J Alzheimer’s Dis 2017;55:1285–94. 10.3233/JAD-160733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Hampel H, Mesulam MM, Cuello AC, Khachaturian AS, Vergallo A, Farlow MR, et al. Revisiting the cholinergic hypothesis in Alzheimer’s disease: Emerging evidence from translational and clinical research. J Prev Alzheimer’s Dis 2019;6:2–15. 10.14283/jpad.2018.43. [DOI] [PubMed] [Google Scholar]
  • [34].Latina V, Caioli S, Zona C, Ciotti MT, Amadoro G, Calissano P. Impaired NGF/TrkA signaling causes early AD-linked presynaptic dysfunction in cholinergic primary neurons. Front Cell Neurosci 2017;11:1–23. 10.3389/fncel.2017.00068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Canu N, Amadoro G, Triaca V, Latina V, Sposato V, Corsetti V, et al. The intersection of NGF/TrkA signaling and amyloid precursor protein processing in Alzheimer’s disease neuropathology. Int J Mol Sci 2017;18:1–17. 10.3390/ijms18061319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Counts SE, Mufson EJ. The role of nerve growth factor receptors in cholinergic basal forebrain degeneration in prodromal Alzheimer disease. J Neuropathol Exp Neurol 2005;64:263–72. [DOI] [PubMed] [Google Scholar]
  • [37].Mitra S, Behbahani H, Eriksdotter M. Innovative therapy for Alzheimer’s disease-with focus on biodelivery of NGF. Front Neurosci 2019;13(38). 10.3389/fnins.2019.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Davies P, Maloney AJF. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 1976;2:1403. [DOI] [PubMed] [Google Scholar]
  • [39].Whitehouse PJ, Price DL, Clark AW, Coyle JT, DeLong MR. Alzheimer disease: Evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol 1981;10:122–6. [DOI] [PubMed] [Google Scholar]
  • [40].Gielow MR, Zaborszky L. The input-output relationship of the cholinergic basal forebrain. Cell Rep 2017;18:1817–30. 10.1016/j.celrep.2017.01.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Mesulam MM, Mufson EJ, Wainer BH, Levey AI. Central cholinergic pathways in the rat: An overview based on an alternative nomenclature (Ch1-Ch6). Neuroscience 1983;10:1185–201. 10.1016/0306-4522(83)90108-2. [DOI] [PubMed] [Google Scholar]
  • [42].Pepeu G, Giovannini G. The fate of the brain cholinergic neurons in neurodegenerative diseases. Brain Res 2017;1670:173–84. 10.1016/j.brainres.2017.06.023. [DOI] [PubMed] [Google Scholar]
  • [43].Schmitz TW, Nathan Spreng R, Weiner MMW, Aisen P, Petersen R, Jack CR, et al. Basal forebrain degeneration precedes and predicts the cortical spread of Alzheimer’s pathology. Nat Commun 2016;7(1):13249. 10.1038/ncomms13249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Fernández-Cabello S, Kronbichler M, van Dijk KRA, Goodman JA, Nathan Spreng R, Schmitz TW. Basal forebrain volume reliably predicts the cortical spread of Alzheimer’s degeneration. Brain 2020;143:993–1009. 10.1093/brain/awaa012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Cantero JL, Zaborszky L, Atienza M. Volume loss of the nucleus basalis of Meynert is associated with atrophy of innervated regions in mild cognitive impairment. Cereb Cortex 2017;27:3881–9. 10.1093/cercor/bhw195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Grothe M, Heinsen H, Teipel SJ. Atrophy of the cholinergic basal forebrain over the adult age range and in early stages of Alzheimer’s disease. Biol Psychiatry 2012;71:805–13. 10.1016/j.biopsych.2011.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Kilimann I, Hausner L, Fellgiebel A, Filippi M, Würdemann TJ, Heinsen H, et al. Parallel atrophy of cortex and basal forebrain cholinergic system in mild cognitive impairment. Cereb Cortex 2017;27:1841–8. 10.1093/cercor/bhw019. [DOI] [PubMed] [Google Scholar]
  • [48].Muth K, Schönmeyer R, Matura S, Haenschel C, Schröder J, Pantel J. Mild cognitive impairment in the elderly is associated with volume loss of the cholinergic basal forebrain region. Biol Psychiatry 2010;67(6):588–91. 10.1016/j.biopsych.2009.02.026. [DOI] [PubMed] [Google Scholar]
  • [49].Teipel SJ, Cavedo E, Hampel H, Grothe MJ, Aguilar LF, Babiloni C, et al. Basal forebrain volume, but not hippocampal volume, is a predictor of global cognitive decline in patients with Alzheimer’s disease treated with cholinesterase inhibitors. Front Neurol 2018;9:642. 10.3389/fneur.2018.00642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Alexopoulos P, Werle L, Roesler J, Thierjung N, Gleixner LS, Yakushev I, et al. Conflicting cerebrospinal fluid biomarkers and progression to dementia due to Alzheimer’s disease. Alzheimer’s Res Ther 2016;8(1):51. 10.1186/s13195-016-0220-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Dickerson BC, Bakkour A, Salat DH, Feczko E, Pacheco J, Greve DN, et al. The cortical signature of Alzheimer’s disease: Regionally specific cortical thinning relates to symptom severity in very mild to mild AD dementia and is detectable in asymptomatic amyloid-positive individuals. Cereb Cortex 2009;19:497–510. 10.1093/cercor/bhn113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Grothe MJ, Schuster C, Bauer F, Heinsen H, Prudlo J, Teipel SJ. Atrophy of the cholinergic basal forebrain in dementia with Lewy bodies and Alzheimer’s disease dementia. J Neurol 2014;261:1939–48. 10.1007/s00415-014-7439-z. [DOI] [PubMed] [Google Scholar]
  • [53].Arendt T, Bigl V, Arendt A. Neurone loss in the nucleus basalis of Meynert in Creutzfeldt-Jakob disease. Acta Neuropathol 1984;65:85–8. 10.1007/BF00689832. [DOI] [PubMed] [Google Scholar]
  • [54].Cartier L, Verdugo R, Vergara C, Galvez S. The nucleus basalis of Meynert in 20 definite cases of Creutzfeldt-Jakob disease. J Neurol Neurosurg Psychiatry 1989;52:304–9. 10.1136/jnnp.52.3.304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Convery RS, Neason MR, Cash DM, Cardoso MJ, Modat M, Ourselin S, et al. Basal forebrain atrophy in frontotemporal dementia. NeuroImage Clin 2020;26:102210. 10.1016/j.nicl.2020.102210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Summers W, Viesselman J, Marsh G, Candelora K. Use of THA in treatment of Alzheimer-like dementia: Pilot study in twelve patients. Biol Psychiatry 1981;16(2):145–53. [PubMed] [Google Scholar]
  • [57].Bartus R, Dean R, Beer B, Lippa A. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982;217:408–14. 10.1126/science.7046051. [DOI] [PubMed] [Google Scholar]
  • [58].Santos MD, Alkondon M, Pereira EFR, Aracava Y, Eisenberg HM, Maelicke A, et al. The nicotinic allosteric potentiating ligand galantamine facilitates synaptic transmission in the mammalian central nervous system. Mol Pharmacol 2002;61:1222–34. 10.1124/mol.61.5.1222. [DOI] [PubMed] [Google Scholar]
  • [59].Carrière I, Fourrier-Reglat A, Dartigues JF, Rouaud O, Pasquier F, Ritchie K, et al. Drugs with anticholinergic properties, cognitive decline, and dementia in an elderly general population: The 3-city study. Arch Intern Med 2009;169:1317–24. 10.1001/archinternmed.2009.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Chuang Y-F, Elango P, Gonzalez CE, Thambisetty M. Midlife anticholinergic drug use, risk of Alzheimer’s disease, and brain atrophy in community-dwelling older adults. Alzheimer’s Dement Transl Res Clin Interv 2017;3:471–9. 10.1016/j.trci.2017.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Dmochowski RR, Thai S, Iglay K, Enemchukwu E, Tee S, Varano S, et al. Increased risk of incident dementia following use of anticholinergic agents: A systematic literature review and meta-analysis. Neurourol Urodyn 2021;40:28–37. 10.1002/nau.24536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Gray SL, Anderson ML, Dublin S, Hanlon JT, Hubbard R, Walker R, et al. Cumulative use of strong anticholinergics and incident dementia: A prospective cohort study. JAMA Intern Med 2015;75:401–7. 10.1001/jamainternmed.2014.7663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Grossi CM, Richardson K, Fox C, Maidment I, Steel N, Loke YK, et al. Anticholinergic and benzodiazepine medication use and risk of incident dementia: A UK cohort study. BMC Geriatr 2019;19(1):276. 10.1186/s12877-019-1280-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Pieper NT, Grossi CM, Chan WY, Loke YK, Savva GM, Haroulis C, et al. Anticholinergic drugs and incident dementia, mild cognitive impairment and cognitive decline: A meta-analysis. Age Ageing 2020;49:939–47. 10.1093/ageing/afaa090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Risacher SL, McDonald B, Tallman E, West J, Farlow MR, Unverzagt FW, et al. Association between anticholinergic medication use and cognition, brain metabolism, and brain atrophy in cognitively normal older adults. JAMA Neurol 2016;73:721–32. 10.1001/jamaneurol.2016.0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Campbell NL, Lane KA, Gao S, Boustani MA. Anticholinergics influence transition from normal cognition to mild cognitive impairment in older adults in primary care. Pharmacotherapy 2018;38:511–9. 10.1002/phar.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Cai X, Campbell N, Khan B, Callahan C, Boustani M. Long-term anticholinergic use and the aging brain. Alzheimer’s Dement 2013;9(4):377–85. 10.1016/j.jalz.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Moss DE. Is combining an anticholinergic with a cholinesterase inhibitor a good strategy for high-level CNS cholinesterase inhibition? J Alzheimer’s Dis 2019;74:1099–103. 10.3233/JAD-190626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Jack CJ, Bennett D, Blennow K, Carrillo M, Dunne B. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement 2018;14:535–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Fox NC, Ma S, Cousens R, Scahill MA, Harvey RJ, Rossor MN. Using serial registered brain magnetic resonance imaging to measure disease progression in Alzheimer disease power calculations and estimates of sample size to detect treatment effects. Arch Neurol 2000;57(3):339–44. 10.10001/archneur.57.3.339. [DOI] [PubMed] [Google Scholar]
  • [71].Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991;82:239–59. 10.1007/BF00308809. [DOI] [PubMed] [Google Scholar]
  • [72].Killiany RJ, Moss MB, Albert MS, Sandor T, Tieman J, Jolesz F. Temporal lobe regions on magnetic resonance imaging identify patients with early Alzheimer’s disease. Arch Neurol 1993;50(9):949–54. 10.1001/archneur.1993.00540090052010. [DOI] [PubMed] [Google Scholar]
  • [73].Lehericy S, Baulac M, Chiras J, Pierot L, Martin N, Pillon B, et al. Amygdalohippocampal MR volume measurements in the early stages of Alzheimer disease. AJNR Am J Neuroradio 1994;15:927–37. [PMC free article] [PubMed] [Google Scholar]
  • [74].Jobst KA, Smith AD, Szatmari M, Esiri MM, Jaskowski A, Hindley N, et al. Rapidly progressing atrophy of medial temporal lobe in Alzheimer’s disease. Lancet 1994;343:829–30. 10.1016/S0140-6736(94)92028-1. [DOI] [PubMed] [Google Scholar]
  • [75].Hashimoto M, Kazui H, Matsumoto K, Nakano Y, Yasuda M, Mori E. Does donepezil treatment slow the progression of hippocampal atrophy in patients with Alzheimer’s disease? Am J Psychiatry 2005;162:676–82. 10.1176/appi.ajp.162.4.676. [DOI] [PubMed] [Google Scholar]
  • [76].Jack CR, Petersen RC, Grundman M, Jin S, Gamst A, Ward CP, et al. Longitudinal MRI findings from the vitamin E and donepezil treatment study for MCI. Neurobiol Aging 2008;29:1285–95. 10.1016/j.neurobiolaging.2007.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Ferris S, Nordberg A, Soininen H, Darreh-Shori T, Lane R. Progression from mild cognitive impairment to Alzheimer’s disease: Effects of gender, butyrylcholinesterase genotype and rivastigmine treatment 2009;19:635–46. 10.1097/FPC.0b013e32832f8c17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Venneri A, McGeown WJ, Shanks MF. Empirical evidence of neuroprotection by dual cholinesterase inhibition in Alzheimer’s disease. Neuroreport 2005;16:107–10. 10.1097/00001756-200502080-00006. [DOI] [PubMed] [Google Scholar]
  • [79].Venneri A, Lane R. Effects of cholinesterase inhibition on brain white matter volume in Alzheimerʼs disease. Neuroreport 2009;20:285–8. 10.1097/WNR.0b013e3283207d21. [DOI] [PubMed] [Google Scholar]
  • [80].Fields RD, Dutta DJ, Belgrad J, Robnett M. Cholinergic signaling in myelination. Glia 2017;65:687–98. 10.1002/glia.23101. [DOI] [PubMed] [Google Scholar]
  • [81].Darvesh S Butyrylcholinesterase as a diagnostic and therapeutic target for Alzheimer’s disease. Curr Alzheimer Res 2016;13:1173–7. 10.2174/1567205013666160404120542. [DOI] [PubMed] [Google Scholar]
  • [82].Davidsson P, Blennow K, Andreasen N, Eriksson B, Minthon L, Hesse C. Differential increase in cerebrospinal fluid-acetylcholinesterase after treatment with acetylcholinesterase inhibitors in patients with Alzheimer’s disease. Neurosci Lett 2001;300:157–60. 10.1016/S0304-3940(01)01586-5. [DOI] [PubMed] [Google Scholar]
  • [83].Nordberg A, Darreh-Shori T, Peskind E, Soininen H, Mousavi M, Eagle G, et al. Different cholinesterase inhibitor effects on CSF cholinesterases in Alzheimer patients. Curr Alzheimer Res 2009;6:4–14. 10.2174/156720509787313961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Parnetti L, Amici S, Lanari A, Romani C, Antognelli C, Andreasen N, et al. Cerebrospinal fluid levels of biomarkers and activity of acetylcholinesterase (AChE) and butyrylcholinesterase in AD patients before and after treatment with different AChE inhibitors. Neurol Sci 2002;23:s95–6. 10.1007/s100720200086. [DOI] [PubMed] [Google Scholar]
  • [85].Parnetti L, Chiasserini D, Andreasson U, Ohlson M, Hüls C, Zetterberg H, et al. Changes in CSF acetyl- and butyrylcholinesterase activity after long-term treatment with AChE inhibitors in Alzheimer’s disease. Acta Neurol Scand 2011;124:122–9. 10.1111/j.1600-0404.2010.01435.x. [DOI] [PubMed] [Google Scholar]
  • [86].Darreh-Shori T, Meurling L, Pettersson T, Hugosson K, Hellström-Lindahl E, Andreasen N, et al. Changes in the activity and protein levels of CSF acetylcholinesterases in relation to cognitive function of patients with mild Alzheimer’s disease following chronic donepezil treatment. J Neural Transm 2006;113:1791–801. 10.1007/s00702-006-0526-2. [DOI] [PubMed] [Google Scholar]
  • [87].Karami A, Eriksdotter M, Kadir A, Almkvist O, Nordberg A, Darreh-Shori T. CSF cholinergic index, a new biomeasure of treatment effect in patients with Alzheimer’s disease. Front Mol Neurosci 2019;12:239. 10.3389/fnmol.2019.00239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Kadir A, Darreh-Shori T, Almkvist O, Wall A, Långström B, Nordberg A. Changes in brain 11C–nicotine binding sites in patients with mild Alzheimer’s disease following rivastigmine treatment as assessed by PET. Psychopharmacology (Berl) 2007;191:1005–14. 10.1007/s00213-007-0725-z. [DOI] [PubMed] [Google Scholar]
  • [89].Barnes C, Meltzer J, Houston F, Orr G, McGann K, Wenk G. Chronic treatment of old rats with donepezil or galantamine: Effects on memory, hippocampal plasticity and nicotinic receptors. Neuroscience 2000;99(1):17–23. 10.1016/S0306-4522(00)00180-9. [DOI] [PubMed] [Google Scholar]
  • [90].Rattray M Is there nicotinic modulation of nerve growth factor? Implications for cholinergic therapies in Alzheimer’s disease. Biol Psychiatry 2001;49:185–93. 10.1016/S0006-3223(00)01047-7. [DOI] [PubMed] [Google Scholar]
  • [91].French SJ, Humby T, Horner CH, Sofroniew MV., Rattray M. Hippocampal neurotrophin and Trk receptor mRNA levels are altered by local administration of nicotine, carbachol and pilocarpine. Mol Brain Res 1999;67:124–36. 10.1016/S0169-328X(99)00048-0. [DOI] [PubMed] [Google Scholar]
  • [92].Appel SH. A unifying hypothesis for the cause of amyotrophc lateral sclerosis, parkinsonism, and Alzheimer disease. Ann Neurol 1981;10:499–505. [DOI] [PubMed] [Google Scholar]
  • [93].Sofroniew MV, Pearson RCA, Eckenstein F, Cuello AC, Powell TPS. Retrograde changes in cholinergic neurons in the basal forebrain of the rat following cortical damage. Brain Res 1983;289:370–4. 10.1016/0006-8993(83)90045-8. [DOI] [PubMed] [Google Scholar]
  • [94].Stephens PH, Tagari PC, Cuello AC. Retrograde degeneration of basal forebrain cholinergic neurons after neurotoxin lesions of the neocortex: Application of ganglioside GM1. Neurochem Int 1988;12:475–81. 10.1016/0197-0186(88)90031-9. [DOI] [PubMed] [Google Scholar]
  • [95].Allard S, Jacobs ML, Do Carmo S, Cuello AC. Compromise of cortical proNGF maturation causes selective retrograde atrophy in cholinergic nucleus basalis neurons. Neurobiol Aging 2018;67:10–20. 10.1016/j.neurobiolaging.2018.03.002. [DOI] [PubMed] [Google Scholar]
  • [96].Iulita MF, Cuello AC. Nerve growth factor metabolic dysfunction in Alzheimer’s disease and Down syndrome. Trends Pharmacol Sci 2014;35:338–48. 10.1016/j.tips.2014.04.010. [DOI] [PubMed] [Google Scholar]
  • [97].Kerwin JM, Morris CM, Perry RH, Perry EK. Hippocampal nerve growth factor receptor immunoreactivity in patients with Alzheimer’s and Parkinson’s disease. Neurosci Lett Lett 1992;143(1–2):101–4. 10.1016/0304-3040(92)90242-Y. [DOI] [PubMed] [Google Scholar]
  • [98].Gauthier LLR, Charrin BCB, Borrell-Pages M, Dompierre JPJ, Rangone H, Cordelieres F, et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 2004;118:127–38. 10.1016/j.cell.2004.06.018. [DOI] [PubMed] [Google Scholar]
  • [99].Niewiadomska G, Baksalerska-pazera M, Riedel G. Altered cellular distribution of phospho-tau proteins coincides with impaired retrograde axonal transport in neurons of aged rats. Ann N Y Acad Sci 2005;1048:287–95. 10.1196/annals.1342.026. [DOI] [PubMed] [Google Scholar]
  • [100].Mufson EJ, Counts S, Perez S, Ginsberg S. Cholinergic system during the progression of Alzheimer’s disease: Therapeutic implications. Expert Rev Neurother 2008;8:1703–1718. 10.1586/14737175.8.11.1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Tiernan CT, Mufson EJ, Kanaan NM, Counts SE. Tau oligomer pathology in nucleus basalis neurons during the progression of Alzheimer Disease. J Neuropathol Exp Neurol 2018;77:246–59. 10.1093/jnen/nlx120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Bruno MA, Cuello AC. Activity-dependent release of precursor nerve growth factor, conversion to mature nerve growth factor, and its degradation by a protease cascade. Proc Natl Acad Sci 2006;103:6735–40. 10.1073/pnas.0510645103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Sofroniew M, Howe C, Mobley W. Nerve growth factor signaling, neuroprotection, and neural repair. Annu Rev Neurosci 2001;24:1217–81. [DOI] [PubMed] [Google Scholar]
  • [104].Thoenen H, Zafra F, Hengerer B, Lindholm D. The synthesis of nerve growth factor and brain-derived neurotrophic factor in hippocampal and cortical neurons is regulated by specific transmitter systems. Ann N Y Acad Sci 1991;640:86–90. 10.1111/j.1749-6632.1991.tb00196.x. [DOI] [PubMed] [Google Scholar]
  • [105].Biane J, Conner JM, Tuszynski MH. Nerve growth factor is primarily produced by GABAergic neurons of the adult rat cortex. Front Cell Neurosci 2014;8:1–7. 10.3389/fncel.2014.00220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Cattaneo A, Calissano P. Nerve growth factor and Alzheimer’s disease: New facts for an old hypothesis. Mol Neurobiol 2012;46:588–604. 10.1007/s12035-012-8310-9. [DOI] [PubMed] [Google Scholar]
  • [107].Isaev NK, Stelmashook EV., Genrikhs EE. Role of nerve growth factor in plasticity of forebrain cholinergic neurons. Biochem 2017;82:291–300. 10.1134/S0006297917030075. [DOI] [PubMed] [Google Scholar]
  • [108].Kordower J, Bartus R, Bothwell M, Schmatteman G, Gash D. Nerve growth factor receptor immunoreactivity in nonhuman primate (Cebus apella): Distribution, morphology, colocalization with cholinergic enzymes. J Comp Neurol 1988;277:465–86. [DOI] [PubMed] [Google Scholar]
  • [109].Hefti F, Hartikka JI, Salvatierra A, Weiner WJ, Mash DC. Localization of nerve growth factor receptors in the cholinergic neurons of the human basal forebrain. Neurosci Lett 1986;69:37–41. [DOI] [PubMed] [Google Scholar]
  • [110].Mufson E, Bothwell M, Hersh L, Kordower J. Nerve growth factor receptor immunoreactivity profiles in the normal aged human basal forebrain: Colocalization with cholinergic neurons. J Comp Neurol 1989;285:196–217. [DOI] [PubMed] [Google Scholar]
  • [111].Cuello AC, Bruno MA. The failure in NGF maturation and its increased degradation as the probable cause for the vulnerability of cholinergic neurons in Alzheimer’s disease. Neurochem Res 2007;32:1041–5. 10.1007/s11064-006-9270-0. [DOI] [PubMed] [Google Scholar]
  • [112].Kotagal V, Müller MLTM, Kaufer DI, Koeppe RA, Bohnen NI. Thalamic cholinergic innervation is spared in Alzheimer disease compared to parkinsonian disorders. Neurosci Lett 2012;514:169–172. 10.1016/j.neulet.2012.02.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Strada O, Hirsch EC, Javoy-Agid F, Lehericy S, Ruberg M, Hauw JJ, et al. Does loss of nerve growth factor receptors precede loss of cholinergic neurons in Alzheimer’s disease? An autoradiographic study in the human striatum and basal forebrain. J Neurosci 1992;12:4766–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Iulita MF, Bistué Millón MB, Pentz R, Aguilar LF, Do Carmo S, Allard S, et al. Differential deregulation of NGF and BDNF neurotrophins in a transgenic rat model of Alzheimer’s disease. Neurobiol Dis 2017;108:307–23. 10.1016/j.nbd.2017.08.019. [DOI] [PubMed] [Google Scholar]
  • [115].Allard S, Leon WC, Pakavathkumar P, Bruno MA, Ribeiro-da-Silva A, Cuello AC. Impact of the NGF Maturation and Degradation Pathway on the Cortical Cholinergic System Phenotype. J Neurosci 2012;32:2002–12. 10.1523/JNEUROSCI.1144-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, et al. Cholinergic modulation of glial function during aging and chronic neuroinflammation. Front Cell Neurosci 2020;14:577912. 10.3389/fncel.2020.577912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Vogels OJM, Broere CAJ, Ter Laak HJ, Ten Donkelaar HJ, Nieuwenhuys R, Schulte BPM. Cell loss and shrinkage in the nucleus basalis Meynert complex in Alzheimer’s disease. Neurobiol Aging 1990;11(1):3–13. 10.1016/0197-4580(90)90056-6. [DOI] [PubMed] [Google Scholar]
  • [118].Sampaio TB, Savall AS, Gutierrez MEZ, Pinton S. Neurotrophic factors in Alzheimer’s and Parkinson’s diseases: Implications for pathogenesis and therapy. Neural Regen Res 2017;12(4):549–57. 10.4103/1673-5374.205084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Rinne JO, Paljärvi L, Rinne UK. Neuronal size and density in the nucleus basalis of Meynert in Alzheimer’s disease. J Neurol Sci 1987;79(1–2):67–76. 10.1016/0022-510X(87)90260-7. [DOI] [PubMed] [Google Scholar]
  • [120].Pearson RCA, Sofroniew MV, Cuello AC, Powell TPS, Eckenstein F, Esiri MM, et al. Persistence of cholinergic neurons in the basal nucleus in a brain with senile dementia of the Alzheimer’s type demonstrated by immunohistochemical staining for choline acetyltransferase. Brain Res 1983;289(1–2):375–9. 10.1016/0006-8993(83)90046-X. [DOI] [PubMed] [Google Scholar]
  • [121].Wu YJ, Wang L, Ji C fan, Gu S fei, Yin Q, Zuo J. The role of α7nAChR-mediated cholinergic anti- inflammatory pathway in immune cells. Inflammation 2021;44(3):821–34. 10.1007/s10753-020-01396-6. [DOI] [PubMed] [Google Scholar]
  • [122].Li W, Antuono PG, Xie C, Chen G, Jones JL, Ward BD, et al. Changes in regional cerebral blood flow and functional connectivity in the cholinergic pathway associated with cognitive performance in subjects with mild Alzheimer’s disease after 12-week donepezil treatment. Neuroimage 2012;60:1083–91. 10.1016/j.neuroimage.2011.12.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Saito ER, Miller JB, Harari O, Cruchaga C, Mihindukulasuriya KA, Kauwe JSK, et al. Alzheimer’s disease alters oligodendrocytic glycolytic and ketolytic gene expression. Alzheimer’s Dement 2021:1–13. 10.1002/alz.12310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Bohnen NI, Kaufer DI, Hendrickson R, Ivanco LS, Lopresti BJ, Koeppe RA, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzeimer’s disease. J Neurol Neurosurg Psychiatry 2005;76:315–9. 10.1136/jnnp.2004.038729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Kaasinen V, Någren K, Järvenpää T, Roivainen A, Yu M, Oikonen V, et al. Regional effects of donepezil and rivastigmine on cortical acetylcholinesterase activity in Alzheimer’s disease. J Clin Psychopharmacol 2002;22:615–20. 10.1097/00004714-200212000-00012. [DOI] [PubMed] [Google Scholar]
  • [126].Kuhl DE, Minoshima S, Frey KA, Foster NL, Kilbourn MR, Koeppe RA. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol 2000;48:391–5. . [DOI] [PubMed] [Google Scholar]
  • [127].Kadir A, Darreh-Shori T, Almkvist O, Wall A, Grut M, Strandberg B, et al. PET imaging of the in vivo brain acetylcholinesterase activity and nicotine binding in galantamine-treated patients with AD. Neurobiol Aging 2008;29:1204–17. 10.1016/j.neurobiolaging.2007.02.020. [DOI] [PubMed] [Google Scholar]
  • [128].Ota T, Shinotoh H, Fukushi K, Kikuchi T, Sato K, Tanaka N, et al. Estimation of plasma IC50 of donepezil for cerebral acetylcholinesterase inhibition in patients with Alzheimer disease using positron emission tomography. Clin Neuropharmacol 2010;33:74–8. 10.1097/WNF.0b013e3181c71be9. [DOI] [PubMed] [Google Scholar]
  • [129].Imbimbo BP. Pharmacodynamic-tolerability relationships of cholinesterase inhibitors for Alzheimer’s disease. CNS Drugs 2001;15:375–90. [DOI] [PubMed] [Google Scholar]
  • [130].Jann MW, Shirley KL, Small GW. Clinical pharmacokinetics and pharmacodynamics of cholinesterase inhibitors. Clin Pharmacokinet 2002;41:719–39. [DOI] [PubMed] [Google Scholar]
  • [131].Hallak M, Giacobini E. A comparison of the effects of two inhibitors on brain cholinesterase. Neuropharmacology 1987;26:521–30. 10.1016/0028-3908(87)90143-2. [DOI] [PubMed] [Google Scholar]
  • [132].Kobayashi H, Nakano T, Moss DE, Suzuki T. Effects of a central anticholinesterase, methanesulfonyl fluoride on the cerebral cholinergic system and behavior in mice: Comparison with an organophosphate DDVP. J Heal Sci 1999;45(4):191–202. 10.1248/jhs.45.191. [DOI] [Google Scholar]
  • [133].López-Arrieta J, Schneider L. Metrifonate for Alzheimer’s disease. Cochrane Database Syst Rev 2006;2. 10.1002/14651858.CD003155.pub3. [DOI] [PubMed] [Google Scholar]
  • [134].Moss D, Kobayashi H, Pacheco G, Palacios R, Perez R. Methanesulfonyl fluoride: A CNS selective cholinesterase inhibitor. In: Giacobini E, Becker R, editors. Curr. Res. Alzheimer Ther. Cholinesterase Inhib, New York: Taylor and Francis; 1988, p. 305–14. [Google Scholar]
  • [135].Moss DE, Fariello RG, Sahlmann J, Sumaya I, Pericle F, Braglia E. A randomized phase I study of methanesulfonyl fluoride, an irreversible cholinesterase inhibitor, for the treatment of Alzheimer’s disease. Br J Clin Pharmacol 2013;75:1231–9. 10.1111/bcp.12018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Moss DE, Berlanga P, Hagan MM, Sandoval H, Ishida C. Methanesulfonyl fluoride (MSF): A double-blind, placebo-controlled study of safety and efficacy in the treatment of senile dementia of the Alzheimer type. Alzheimer Dis Assoc Disord 1999;13:20–5. 10.1097/00002093-199903000-00003. [DOI] [PubMed] [Google Scholar]
  • [137].Moss DE. Improving anti-neurodegenerative benefits of acetylcholinesterase inhibitors in Alzheimer’s disease: Are irreversible inhibitors the future? Int J Mol Sci 2020;21(10):3438. 10.3390/ijms21103438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].McDade E, Bednar MM, Brashear HR, Miller DS, Maruff P, Randolph C, et al. The pathway to secondary prevention of Alzheimer’s disease. Alzheimer’s Dement Transl Res Clin Interv 2020;6:e12069. 10.1002/trc2.12069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Alzheimer’s Association. Trajectory Report. Https://WwwAlzOrg/Help-Support/Resources/Publications/TrajectoryReport 2015. https://www.alz.org/help-support/resources/publications/trajectoryreport (accessed April 6, 2021).
  • [140].Repantis D, Laisney O, Heuser I. Acetylcholinesterase inhibitors and memantine for neuroenhancement in healthy individuals: A systematic review. Pharmacol Res 2010;61(6):473–81. 10.1016/j.phrs.2010.02.009. [DOI] [PubMed] [Google Scholar]
  • [141].Balsters JH, O’Connell RG, Martin MP, Galli A, Cassidy SM, Kilcullen SM, et al. Donepezil impairs memory in healthy Older subjects: Behavioural, EEG and simultaneous EEG/fMRI biomarkers. PLoS One 2011;6(9):e24126. 10.1371/journal.pone.0024126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Daubner J, Arshaad MI, Henseler C, Hescheler J, Ehninger D, Broich K, et al. Pharmacological neuroenhancement: Current aspects of categorization, epidemiology, pharmacology, drug development, ethics, and future perspectives. Neural Plast 2021;2021:8823383. 10.1155/2021/8823383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Nizri E, Brenner T. Modulation of inflammatory pathways by the immune cholinergic system. Amino Acids 2013;45(1):73–85. 10.1007/s00726-011-1192-8. [DOI] [PubMed] [Google Scholar]
  • [144].Hall H, Iulita MF, Gubert P, Flores Aguilar L, Ducatenzeiler A, Fisher A, et al. AF710B, an M1/sigma-1 receptor agonist with long-lasting disease-modifying properties in a transgenic rat model of Alzheimer’s disease. Alzheimer’s Dement 2018;14:811–23. 10.1016/j.jalz.2017.11.009. [DOI] [PubMed] [Google Scholar]
  • [145].Hernandez CM, Dineley KT. 7 Nicotinic acetylcholine receptors in Alzheimer’s disease: Neuroprotective, neurotrophic or both? Curr Drug Targets 2012;13:613–22. [DOI] [PubMed] [Google Scholar]
  • [146].Li L, Liu Z, Jiang Y-Y, Peng Y-P, Qiu Y-H. Acetylcholine suppresses microglial inflammatory response via αnAChR to protect hippocampal neurons. J Integr Neurosci 2019;18:51–6. 10.31083/j.jin.2019.01.114. [DOI] [PubMed] [Google Scholar]
  • [147].Fisher A Cholinergic treatments with emphasis on M1 muscarinic agonists as potential disease-modifying agents for Alzheimer’s disease. Neurotherapeutics 2008;5:433–42. 10.1016/j.nurt.2008.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES