Skip to main content
VA Author Manuscripts logoLink to VA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 10.
Published in final edited form as: Neurosci Lett. 2019 Feb 11;701:162–169. doi: 10.1016/j.neulet.2019.02.011

γ-Secretase and its modulators: Twenty years and beyond

Weiming Xia a,b,*
PMCID: PMC7008970  NIHMSID: NIHMS1057990  PMID: 30763650

Abstract

Twenty years ago, Wolfe, Xia, and Selkoe identified two aspartate residues in Alzheimer’s presenilin protein that constitute the active site of the γ-secretase complex. Mutations in the genes encoding amyloid precursor protein (APP) or presenilin (PS) cause early onset familial Alzheimer’s disease (AD), and sequential cleavages of the APP by β-secretase and γ-secretase/presenilin generate amyloid β protein (Aβ), the major component of pathological hallmark, neuritic plaques, in brains of AD patients. Therapeutic strategies centered on targeting γ-secretase/presenilin to reduce amyloid were implemented and led to several high profile clinical trials. This review article focuses on the studies of γ-secretase and its inhibitors/modulators since the discovery of presenilin as the γ-secretase. While a lack of complete understanding of presenilin biology renders failure of clinical trials, the lessons learned from some γ-secretase modulators, while premature for human testing, provide new directions to develop potential therapeutics. Imbalanced Aβ homeostasis is an upstream event of neurodegenerative processes. Exploration of γ-secretase modulators for their roles in these processes is highly significant, e.g., decreasing neuroinflammation and levels of phosphorylated tau, the component of the other AD pathological hallmark, neurofibrillary tangles. Agents with excellent human pharmacology hold great promise in suppressing neurodegeneration in pre-symptomatic or early stage AD patients.

Keywords: Alzheimer, Secretase, Modulator, Neuroinflammation

1. Introduction

Alzheimer’s disease (AD) is by far the most prevalent cause of dementia in the elderly, and the disease may evolve over the course of decades. Pre-symptomatic AD subjects usually do not exhibit any phenotype before converting to the earliest clinically detectable stage, known as “mild cognitive impairment (MCI)”. A certain percentage of MCI maintain their cognitive function throughout the remaining of their life, but a majority gradually converts from MCI to mild AD, moderate AD, and finally advanced stage AD. Overt cognitive decline from initial clinical manifestation lasts for 10–15 years toward the later stages of a more devastating neurodegenerative process. During the disease progression, AD patients exhibit many clinical features, memory impairment being the most prevalent symptom at all stages.

2. Alzheimer pathology: Aβ and Tau

Amyloid β protein (Aβ) plays an important role in AD pathogenesis. At the molecular level, senile amyloid plaques and neurofibrillary tangles (NFT) are two neuropathological hallmarks of AD [1]. Mutations in genes encoding amyloid precursor protein (APP) or presenilin (PS) cause early onset familial AD (FAD), and one or two copies of the apolipoprotein E (apoE) ε4 allele is a major risk factor for lateonset sporadic AD. Aβ is generated by sequential cleavages of the APP by β- and γ-secretases. First, APP is proteolytically processed by β-secretase (BACE1) and generates a 12 kDa C-terminal stub of APP (C99); second, C99 is cleaved by γ-secretase to yield two major species of Aβ ending at residue 40 (Aβ40) or 42 (Aβ42) [2,3]. Genetic studies show that detrimental familial AD-linked missense mutations in APP or PS increase the ratio of 42 residue of Aβ (Aβ42) to a more common 40-residue of Aβ (Aβ40) and cause early onset AD, while a beneficial mutation in APP leads to decreased Aβ production and those carriers maintain intact cognitive function at advanced ages [4]. ApoE has three major isoforms, ApoEε2, ε3 and ε4. ApoEε4 allele is the strongest known risk factor for AD. Brains of sporadic AD patients carrying ApoEε4 allele were found to have increased density of Aβ deposits, limited capability to clear Aβ, and enhanced neuroinflammation [5].

The Aβ42 peptide has been the center of investigation and the target for therapeutic exploration. In AD, the protein subunit of the amyloid plaques, Aβ, does not occur as a single molecular species; many different Aβ-containing peptides have been detected in human cerebrospinal fluid (CSF) and/or brain [6,7]. The most common Aβ isoform in vivo is Aβ40, i.e., a peptide that begins at Asp1 and terminates at Val40 of the Aβ region of APP. Increased accumulation of Aβ42, a peptide that differs from Aβ40 by the inclusion of Ile41 and Ala42, is particularly associated with development of AD. The extra two hydrophobic amino acids of Aβ42 greatly enhance its aggregation propensity [8], leading to accelerated formation of small (low-n) Aβ oligomers (oAβ), larger intermediate assemblies like protofibrils, and eventually the typical ~ 8 nm amyloid fibrils found abundantly in neuritic plaques and amyloid-bearing micro vessels. Small, soluble oligomers of Aβ have been linked to neuronal toxicity and synaptic failure (for review, see [9]). The ratio of Aβ42/Aβ40, rather than the total amount of Aβ, has been shown to correlate with the age of onset of FAD [10] and with the amount of plaques in mouse models [11,12]. Aβ42 constitutes approximately 10% of total Aβ species [13] and is more prone to aggregation than Aβ 40 [8,14], Furthermore, Aβ40 may play an antagonistic role in preventing Aβ42 aggregation in vivo [11,12] and in vitro [15-17]. Expression of Aβ42, rather than Aβ40, in Drosophila and mice led to the formation of Aβ plaques [18,19]. Therefore, specific inhibition of γ-secretase activity for Aβ42 generation would be an appealing strategy for the treatment of AD [20,21].

The Aβ peptide is closely linked to a second AD pathological protein, tau. This intracellular hallmark of AD is the paired helical filament (PHF) in NFT containing hyperphosphorylated tau. Mutation in the tau gene causes frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) [22]. Transgenic mice expressing mutant tau show close association of mutation to NFT formation and neurodegeneration [23,24]. Furthermore, Aβ has been shown to drive tau pathology in vivo [25]. The normal microtubule-associated tau gradually loses association with microtubules and hyperphosphorylated tau forms PHF which accumulate in neuronal cytoplasm as the major component of the NFT. Glycogen synthase kinase-3β (GSK3β) is one of several microtubule-associated kinases responsible for tau phosphorylation [26]. GSK3β has been found to phosphorylate a number of sites on tau that were identified by nanoelectrospray mass spectrometry, including the residue Thr181 to form pTau181 [27].

The significance of Aβ and tau as therapeutic targets is not only derived from pathological evidence of postmortem brain but also from biochemical analysis of cultured human neurons. This is largely based on the induced pluripotent stem cell (iPSC) technology that has been established to study Aβ, tau, and GSK3β in AD [28]. When iPSC lines from two normal subjects, two SAD (sAD1 and sAD2), and two FAD patients carrying a duplicated copy of the APP gene were established for human neuronal differentiation, those from two duplicate APP gene carriers and patient sAD2 showed very high levels of Aβ40, phosphortau(Thr 231) and active GSK3β [28]. Importantly, levels of Aβ, pTau and active GSK3β can be reduced by β-secretase inhibitors, indicating a direct relationship between γ-secretase substrate C99 and GSK3β activation/Tau phosphorylation. Other studies have demonstrated that iPSC-derived neuronal cells exhibit reduced Aβ levels in the presence of BACE1 inhibitor or γ-secretase inhibitor/modulators [29,30]. Thus, involvement of Aβ and tau in AD pathogenesis can be modeled in human neuronal cells amendable for testing of therapies targeting either Aβ or phosphorylated tau [31].

3. Targeting γ-secretase with inhibitors: from the end to the beginning?

Aβ targeted therapies are being actively pursued in preclinical and clinical studies for treatment of AD. These therapies are based on the amyloid cascade hypothesis, which postulates that Aβ peptides form neurotoxic species, trigger a pathological cascade and ultimately lead to neurodegeneration and dementia [32,33]. γ-Secretase, along with the β-secretase, have become the prime target for this purpose. γ-Secretase is composed of PS1, Presenilin Enhancer-2 (Pen-2), anterior pharynx defective-1 (Aph-1), and Nicastrin (Nct). PS1 carries two aspartate residues constituting the active site of γ-secretase [34], and Pen-2 is a small protein of 101 amino acids with two TM domains [35,36]. Many studies have demonstrated that overexpression of all four components results in increased γ-secretase activity, both in mammalian cells [37-42] and in yeast [43]. Purified PS1 and Pen-2 are sufficient to carry out γ-secretase cleavage of its substrates in vitro; PS1 itself has proteolytic activity [44,45], while Pen-2 promotes the conversion of PS1 from zymogen to the active protease [44,46].

A large number of potent γ-secretase inhibitors (GSIs) have been developed. Two GSIs, LY-450139 (Semagacestat) and BMS-708163 (Avagacestat), are among those tested in clinical trials. LY-450139 is known to block the cleavage of APP and Notch, another γ-secretase substrate [47], and subjects receiving LY-450139 presented with worsening of clinical measures of cognition and the ability to perform activities of daily living [48,49]. Because perturbed Notch signaling has been implicated in cancer formation, inhibition of Notch signaling by LY-450139 could be one of the culprits causing the undesired clinical outcomes [50].

More selective GSIs targeting APP over Notch were developed, like allosteric GSIs and BMS-708163. Allosteric GSIs (AGSI) displays specificity against Aβ42 production over Aβ40, Aβ38 and Notch1 cleavage [51]. These AGSIs bind to an allosteric site within γ-secretase rather than the APP substrate. Furthermore, AGSIs affect γ-secretase activity for both Aβ40 and Aβ38 production similarly and therefore lack the interconnected effect of the γ-secretase modulators (GSM) (see below) in which decreased Aβ42 resulted in increased Aβ38 generation [52]. Clearly, these AGSIs represent a class of inhibitors that are distinct from the Notch-sparing GSIs that have no selectivity for Aβ40 and Aβ42 [53-55].

BMS-708163 is a potent GSI that showed impressive γ-secretase inhibition with 50% inhibition concentrations (IC50) of 0.27 and 0.30 nM for Aβ42 and Aβ40, and 58 nM for Notch, respectively, thus representing a 193-fold selectivity for APP over Notch [56]. Pharmacokinetic (PK)-pharmacodynamic (PD) analysis of BMS-708163 in male dogs revealed plasma concentration of BMS-708163 of ~0.5 μM at 3h post-dosing (hpd) and brain concentration of~0.75 μM at 5 hpd, with a sustained decrease of brain Aβ40 by 50%. However, a separate study indicates that only 3–7 fold selectivity exists for APP over Notch [57], and clinical outcomes [58,59] seem to be discrepant with reported 193-fold selectivity for APP over Notch cleavage [56].

Clinical trials of all GSIs have ended prematurely, raising the question on our understanding of their target, the γ-secretase/presenilin. Development of compounds to target γ-secretase and reduce Aβ production is complicated by the fact that γ-secretase/presenilin has critical biological function, and γ-secretase mediates the final proteolytic cleavage of Notch [60,61] and many substrates. There are over 100 γ-secretase substrates that are type I membrane proteins and have diverse functions. Notch is one of the most interesting and important substrates with diverse functions. Notch has a large extracellular domain, a single transmembrane (TM) domain and an intracellular domain. Notch is proteolyzed in the trans-Golgi as part of its maturation process into a heterodimeric cell surface receptor, then undergoes a second proteolysis upon ligand activation, leading to shedding of the extracellular domain of the receptor. The remaining membrane-bound C-terminal stub, like APP C-terminal fragment, is cleaved by γ-secretase at two sites (in the middle of its TM domain and at a residue close to the interface of the membrane and cytoplasm) to release the Notch-1-β peptide (Nβ, similar to Aβ of APP) and Notch intracellular domain (NICD), which translocates to the nucleus where it regulates gene expression [61-63]. Notch ICD signaling is critical to a wide variety of cell fate determinations during embryonic development and adulthood.

The cytosolic ICDs from known γ-secretase substrates represent a unique library of signaling molecules. Like Notch, they are generated by γ-secretase/PS1 cleavage of substrates [64,65]. These ICDs have different physiological functions linked to regulation of transcription of downstream genes, such as ICDs of alcadeins, CD44, DCC, Notch, Delta, Jagged, E- and N-cadherin, receptor-like protein tyrosine phosphatases, and leukocyte-common antigen related protein. They are involved in a variety of cellular pathways including regulation of cell fate and death, neurite outgrowth, transcriptional regulation, cell-cell adhesion, regulation of ion conductance, and neurotrophin signaling [64,65]. A key concern with GSI is their lack of selectivity among these γ-secretase substrates, e.g., GSIs have shown Notch-related toxicity in rats, including interference with maturation of B- and T-lymphocytes and gastrointestinal tract toxicity [66,67]. A similar requirement of γ-secretase for neuronal survival was found in zebrafish [68,69]. Treating zebrafish with a potent γ-secretase inhibitor, DAPT, causes Notch phenotypes with defects in somitogenesis and neurogenesis [70-73]. The DAPT treated embryos exhibit suppression of Notch phenotypes after injection of Notch intracellular domains (NICD) mRNA [70].

In humans, a PS1 mutation that causes almost complete loss of γ-secretase activity was found in familial AD cases [74]. In mice, double conditional knockout (KO) of PS1 and its homolog PS2 showed progressive loss of synapses, dendrites and neurons, accompanying reduction of NMDA receptor mediated responses and synaptic levels of NMDA receptors [75,76]. Conditional KO of another γ-secretase component, nicastrin, in adult mice displays similar age-dependent cortical neuronal loss likely occurring through apoptosis [77]. With new knowledge of presenilin biology and γ-secretase substrates, it was apparent that development of GSIs for AD needed to be replaced with alternative approaches, such as γ-secretase modulators (GSM).

4. Modulating γ-secretase and neuroinflammation: one stone two birds?

GSMs theoretically have a “regulated” inhibition of γ-secretase activity that could reduce Aβ42 production without obliterating Notch signaling [78], unlike non-selective inhibition of γ-secretase and unwanted side effects for AD therapy caused by GSIs. GSMs only modulate the γ-secretase cleavage site of APP instead of the downstream ε-cleavage site [79]. Competition studies indicate that GSMs have distinct binding sites [80-82]. Cross-linking probes have independently identified PS1-NTF as a specific target of some potent GSMs, while APP is targeted by R-flurbiprofen [83]. These studies have demonstrated that the enzyme γ-secretase itself can be modulated, rather than inhibited, and in such a way that can lower the ratio of Aβ42/40 [80-82]. Therefore, GSMs that have less of an effect on Notch or other substrates will cause fewer adverse side effects.

The first GSM was identified from the discovery of non-steroidal anti-inflammatory drugs (NSAIDs). An amyloid reducing GSM that also suppresses inflammation is desirable. Inflammatory response is an invariable characteristic of AD pathogenesis, in part triggered by Aβ. During AD onset and progression, microglial cells and astrocytes are activated, and cytokines like TNFα are secreted by microglia [84]. In 3X Tg AD model, TNFα is upregulated as Aβ pathology appears, at about 3 months of age [85]. Most cytokines are expressed at very low levels in the healthy brain, and neuroinflammation can be detected years before neurons die. Previous studies have shown that Aβ can bind to scavenger receptors expressed on microglia like CD36 [86] and Scara1 [6], which enter microglia and activate inflammation. When microglia cells engulf extracellular aggregates like Aβ, they trigger inflammasomes (such as NOD-like receptor family pyrin domain-containing 3 (NLRP3)) and activate caspases, and promote IL-1β release [87]. This pathway was validated in AD transgenic mice where NLRP3 was shown to contribute to AD like pathology in mouse brains [88]. Aβ generation and inflammatory response are thus concurrent events associated with Aβ clearance. Accordingly, genetic mutations found in the microglial receptor TREM2 (triggering receptor expressed on myeloid cells 2) triple a person’s risk for AD [89,90] and increased expression of CD33, which functions to suppress Aβ uptake and clearance, modifies AD risk [91,92]. Systems analysis of hundreds AD brain reveals changes in network related to immunologic molecules and microglial cells, including microglial protein TYROBP that binds TREM2 and may regulate CD33 [93].

Several classes of GSMs have been developed [94,95]. As discussed above, one class of GSMs includes a subset of NSAID-like carboxylic acids that specifically block cleavage of the γ-secretase substrates in the middle of their TM domains without affecting generation of the ICD of several type I transmembrane proteins, including APP, ErbB-4, and Notch [96]. These GSMs inhibit Aβ42 production with a concurrent increase of Aβ38 and no effect on Aβ40 production or Notch processing. Dosing cultured cells and transgenic mice revealed that NSAIDs directly modulate the γ-secretase complex [78,97-101], independent of their inhibitory effects on cyclooxygenase (COX) and Rho activity in the Rho-Rock pathway [102].

Another GSM that suppress neuroinflammation is CHF5074. In cultured cells, CHF5074 exhibits IC50s of 18.4 and 3.6 μM for Aβ40 and Aβ42, respectively (6-fold selectivity for Aβ42). In HEK293 cells expressing Notch substrate, Notch cleavage by γ-secretase was not inhibited by CHF5074 at 5 μM [103,104]. In 10-month old Tg2576 mice (expressing the Swedish mutant of APP), steady state brain and plasma concentrations of CHF5074 reached 6.4 μM and 228 μM, respectively, with a ~50% reduction in both the number of plaques and the area occupied by plaques in brain was observed. This corresponded to a ~50% reduction of total brain Aβ and Aβ42 (49% and 42%, respectively). A third reduction of Aβ was found in a second transgenic mouse line expressing both Swedish and London mutant APP after chronic exposure with CHF5074, with brain and plasma drug exposure at 3 and 281 μM respectively [104]. CHF5074 showed reversal of contextual memory deficit and restoration of hippocampal neurogenesis potential [105].

CHF5074 is not only a GSM but also a neuroinflammation modulator [106,107]. Ross et al. reported that subjects on CHF5074 showed linear decreases in levels of inflammatory marker TNFα and soluble CD40 ligand in CSF [106]. Apparently, CHF5074 is the first GSM that shows positive cognitive outcomes in humans with clear decrease in CSF TNFα. TNFα is a major marker of neuroinflammation in AD. It is produced in glial cells and neurons during normal aging and in patients suffering brain trauma, neurodegenerative disorder or excitotoxic insults, and TNFα has detrimental effects on synaptic transmission and plasticity [108]. It is not specific for AD, as most neurodegenerative diseases are accompanied by a cytokine-mediated inflammatory response. Binding of TNFα to Tumor necrosis factor receptor 1 (TNFR1) leads to activation of NFƙB and MAPK pathways, and induction of death signaling [109]. CHF5074 is devoid of anticyclooxygenase (COX) and Notch-interfering activities in vitro [110]. It is likely that CHF5074 has a direct anti-inflammatory effect that is mediated by its interaction with the γ-secretase complex.

Although these compounds were no longer pursued in clinical trials, it is an attractive approach to develop a class of GSMs that may reduce Aβ42 and neuroinflammation markers. Apparently, Aβ deposition and neuroinflammation play critical roles in AD onset and progression, and the challenge is to identify highly potent compounds targeting both pathways.

5. γ-Secretase modulators: a loss of pharmacology?

A lack of translation from in vitro to in vivo pharmacology is an unresolved issue among most GSMs. R-flurbiprofen was tested in clinical trials, however, it did not achieve statistical significance on either of its primary endpoints – cognition or activities of daily living. R-flurbiprofen is a weak GSM with an IC50 for Aβ reduction at approximately 300 μM [100]. Due to its poor brain penetration, it was unlikely to have lowered brain Aβ42 levels in the clinical studies. Treatment of monkeys at 100 mg/kg of ibuprofen or humans with 800 mg single dose did not reveal any changes of Aβ in plasma; CSF Aβ from ibuprofen dosed monkeys did not show any changes [111].

Among 100 CHF5074 treated subjects with MCI, apoEε4 carrier improved on several cognitive measures over the initial three months of treatment [106,107]. During the open label extension period of more than one and half year, apoEε4 carriers maintained their improved cognition and even score better on verbal memory and tests of attention and executive function, compared to baseline performance. Non-carriers’ cognitive abilities have remained stable for almost two years [106,107]. The clinical outcome from apoEε4 carriers versus non-carriers could be explained by findings that apoEε4 carriers usually have more neuroinflammation compared to non-carriers [5], thus it was easier to achieve any anti-neuroinflammatory benefit of CHF5074. However, the IC50 for Aβ42 was at 3.6 μM in cultured cells, indicating that CHF5074 is a weak GSM [103,104].

Another class of non-NSAID derivative GSMs include Eisai’s E-2012 [112,113] and NeuroGenetics’ Compound 4 (Cpd 4) [114]. While Cpd4 directly interacts with PS and Pen2, the binding protein for E-2012 is not clear. When brain exposure was over 100-fold of IC50, Cpd 4 inhibited all three Aβ peptides (Aβ38, 40 and 42) in animals under chronic treatment [114], which is similar to Eisai’s E-2012 that inhibits both Aβ40 and Aβ42 [112,113]. In addition, a number of GSMs were reported by Merck [115-117], and among them, one GSM showed 70% Aβ42 reduction when brain GSM exposure reached 7.8 μM (~400-fold of IC50) [117].

More potent GSMs with IC50 at sub-μM have been reported [118-120]. GSM-10h is a NSAID-derived GSM with an in vitro IC50 of 0.8 μM. In a transgenic mouse line expressing mutant APP and PS1, GSM-10h brain and plasma levels at 6 hpd reached 54.7 μM and 32.9 μM, respectively, which were 40–70 fold of IC50, but brain Aβ42 was reduced by about 20% [121]. There was a concomitant>30% increase in Aβ38, with no effect on Aβ40 [122]. In rats, GSM-10h caused a dose-dependent decrease in the level of Aβ42, but not Aβ40, in brain, CSF, and plasma [123]. An analogue of GSM-10h, GSM-1, carries an in vitro IC50 at 0.35 μM, causes a dose dependent decrease of Aβ42 and an increase of Aβ38 in mice expressing Swedish mutant APP [80,124].

Among the GSMs with IC50 at sub-μM, EVP-A, EVP-B and JNJ-40418677 show similar potency. EVP-A and EVP-B (EnVivo, later Forum Pharmaceuticals) showed in vitro IC50 for reduction of Aβ40 and Aβ42 at 0.24 μM and 0.14 μM, respectively. In rats, a brain concentration of 2.7 μM EVP-A produced no reduction of Aβ, while a brain concentration of 10 μM (40–70 fold above the IC50) of EVP-B produced a 20–30% reduction of brain Aβ [125]. A better compound EVP-0015962 showed a similar IC50 of 0.12 μM in stable human cells, a 4-fold higher IC50 of 0.49 μM in neuronal cells, and no effect on Notch processing. Dose dependent reduction of Aβ42 was observed in rat models. Brain exposures at 2.8 μM and 8.3 μM (5- and 17-fold of IC50) led to a 22% and 38% reduction in brain Aβ42 respectively. Chronic dosing at 20 and 60 mg/kg/day in APP transgenic mice for 6 months led to a lowering of brain plaque load of 81% and 95% respectively [126,127].

A GSM with a similar potency to the EnVivo compounds, JNJ-40418677, selectively inhibited Aβ42 production with IC50s in neuroblastoma cells and primary rat cortical neuronal cultures of 0.20 μM and 0.18 μM, respectively [128]. A lack of effect of JNJ-40418677 on α-and β-secretase was confirmed by visualizing unchanged APP CTFα and CTFβ. In cell-free APP and Notch assays in vitro, JNJ-40418677 did not affect the AICD generation at 100 μM and NICD at 10 μM. Although the difference in its effect on AICD and NICD generation is not clear, a 50-fold selection for Aβ42 inhibition over NICD inhibition was achieved [128]. In mice, four hours after a single oral dose, both brain and plasma GSM exposures achieved 17 μM (85-fold of IC50), with a brain/plasma ratio of 1. Between 2 and 24 h, Aβ42 levels were significantly reduced, and total Aβ levels were not changed in the brain. Chronic dosing of JNJ-40418677 in Tg2576 mice for 7-months at doses of 20,60 and 120 mg/kg/day led to corresponding dose dependent effect. When brain exposure was at 2-fold of IC50 (0.42 μM), no effect on Aβ levels was found. When the brain exposure reached 12-fold of IC50 (2.4 μM) or higher, a significant reduction of Aβ42 was observed, and all three Aβ peptides, Aβ38, 40, and 42, were reduced. The Aβ reduction correlated with a significant reduction in the numbers of plaques that contained Aβ38, Aβ40 and Aβ42 [128]. This is similar to “Notch sparing” GSI, BMS-708163, that reduces levels of CSF Aβ38, 40 and 42 [129]. Therefore, chronic dosing of JNJ-40418677 in animals led to a complete inhibition of all Aβ peptides, a feature like that of GSI.

Two excellent GSMs with clear pharmacological effect across rats, dogs, monkeys, and human subjects are BMS-932481 and BMS-986133 with IC50 at 6.6 and 3.5 nM to reduce Aβ42, respectively. Both GSMs exhibit dose- and time-dependent activity in vivo by decreasing Aβl-42 and Aβ1-40 levels while increasing Aβ1-38 and Aβ1-37 [130,131]. Although the mechanism and central activity of these GSMs translate across preclinical species and humans, insufficient margin for human safety prevents further testing for efficacy of Aβ lowering in AD patients [131].

A unique GSM, BPN-15606, exhibited an IC50 of 7 nM and 17 nM to reduce Aβ42 and Aβ40 from cultured cells, with a concomitant increase of Aβ38 and no change in total Aβ [132]. BPN-15606 binds to an allosteric site within the γ-secretase complex and does not affect Notch cleavage at 25 μM. Dose dependent decreases of plasma, brain and CSF Aβ were found in both mice and rats. Chronic dosing of transgenic mice with BPN-15606 significantly reduced accumulation of Aβ plaques in both the hippocampus and cortex. Like previous reported BACE inhibitors [133], BPN-15606 treatment of 3-dimensional neuronal culture decreased total tau and phosphorylated pThr181 tau [132]. Based on in vivo pharmacokinetic profile of BPN-15606, sub micromolar plasma exposures of BPN-15606 expect to achieve a significant lowering of Aβ42 in human brain, thus requiring much lower doses than those reported for BMS-932481 and BMS-986133.

6. Detecting efficacy of γ-secretase modulators: in 20 years?

It is widely accepted that future AD therapies need to start at an earlier stage, as the onset of disease may occur 15–20 years before the appearance of clinical symptoms [134]. One of major factors contributing to the failure of GSIs and GSMs could be the timing of treatment, i.e., patients at mild to moderate stage might be too late for Aβ reducing therapies as neuronal damage is extensive and irreversible. To test asymptomatic patients at very early stages of disease, biomarkers are needed to identify those subjects for clinical trials. Alternative approaches have been pursued in AD patients carrying FAD mutations (e.g., Dominantly Inherited Alzheimer’s Network). The Alzheimer’s Prevention Initiative (API) was created for clinical trials of Aβ vaccine in pre-symptomatic members from an extended Colombian family carrying a PS1 mutation. Specific GSMs reversing familial mutant PS1/γ-secretase activity may be ideally positioned for those subjects [135]. Anti-Amyloid Treatment in Asymptomatic AD Trial (A4) with brain amyloid imaging has enrolled over a thousand asymptomatic subjects. Therapeutic development with brain imaging and cognitive function as efficacy readouts has been pursued [136], and new Aβ-reducing approaches might be effective in patients with MCI and in pre-symptomatic AD patients. With recent development of GSMs such as BPN-15606 [132] or endogenous cholesterol metabolite cholestenoic acid [137], it does not take 20 years to wait for AD patients converting from pre-symptomatic to symptomatic stages while testing efficacy of GSMs. Advancements in brain imaging and fluid biomarkers will greatly facilitate the discovery of disease modifying therapeutics for AD.

Acknowledgements

This study was supported by the award I21BX003807 and IO1 BX003527 from the Biomedical Laboratory Research and Development Service of the Veterans Affairs Office of Research and Development (WX) and the Cure Alzheimer’s Fund (WX). The views expressed in this article are those of the author and do not represent the views of the US Department of Veterans Affairs or the US Government.

Abbreviation:

AD

Alzheimer’s disease

FAD

familial AD

GSI

γ-secretase inhibitor

GSM

γ-secretase modulator

GSK3β

glycogen synthase kinase-3β

KO

knockout

References

  • [1].Selkoe DJ, Alzheimer disease: mechanistic understanding predicts novel therapies, Ann. Intern. Med 140 (2004) 627–638. [DOI] [PubMed] [Google Scholar]
  • [2].Haass C, Schlossmacher M, Hung AY, Vigo-Pelfrey C, Mellon A, Ostaszewski B, Lieberburg I, Koo EH, Schenk D, Teplow D, Selkoe D, Amyloid b-peptide is produced by cultured cells during normal metabolism, Nature 359 (1992) 322–325. [DOI] [PubMed] [Google Scholar]
  • [3].Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai X, McKay DM, Tintner R, Frangione B, Younkin SG, Production of the Alzheimer amyloid b protein by normal proteolytic processing, Science 258 (1992) 126–129. [DOI] [PubMed] [Google Scholar]
  • [4].Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, Stefansson H, Sulem P, Gudbjartsson D, Maloney J, Hoyte K, Gustafson A, Liu Y, Lu Y, Bhangale T, Graham RR, Huttenlocher J, Bjornsdottir G, Andreassen OA, Jonsson EG, Palotie A, Behrens TW, Magnusson OT, Kong A, Thorsteinsdottir U, Watts RJ, Stefansson K, A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline, Nature 488 (2012) 96–99. [DOI] [PubMed] [Google Scholar]
  • [5].Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, Fagan AM, Morris JC, Mawuenyega KG, Cruchaga C, Goate AM, Bales KR, Paul SM, Bateman RJ, Holtzman DM, Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance, Sci. Transl. Med 3 (2011) 89ra57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Portelius E, Westman-Brinkmalm A, Zetterberg H, Blennow K, Determination of beta-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitationmass spectrometry, J. Proteome Res 5 (2006) 1010–1016. [DOI] [PubMed] [Google Scholar]
  • [7].Vigo-Pelfrey C, Lee D, Keim PS, Lieberburg I, Schenk D, Characterization of bamyloid peptide from human cerebrospinal fluid, J. Neurochem. 61 (1993) 1965–1968. [DOI] [PubMed] [Google Scholar]
  • [8].Jarrett JT, Berger EP, Lansbury PT Jr., The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease, Biochemistry 32 (1993) 4693–4697. [DOI] [PubMed] [Google Scholar]
  • [9].Walsh DM, Selkoe DJ, Deciphering the molecular basis of memory failure in Alzheimer’s disease, Neuron 44 (2004) 181–193. [DOI] [PubMed] [Google Scholar]
  • [10].Kumar-Singh S, Theuns J, Van Broeck B, Pirici D, Vennekens K, Corsmit E, Cruts M, Dermaut B, Wang R, Van Broeckhoven C, Mean age-of-onset of familial alzheimer disease caused by presenilin mutations correlates with both increased Abeta42 and decreased Abeta40, Hum. Mutat 27 (2006) 686–695. [DOI] [PubMed] [Google Scholar]
  • [11].Deng Y, Tarassishin L, Kallhoff V, Peethumnongsin E, Wu L, Li YM, Zheng H, Deletion of presenilin 1 hydrophilic loop sequence leads to impaired gamma-secretase activity and exacerbated amyloid pathology, J. Neurosci 26 (2006) 3845–3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Kim J, Onstead L, Randle S, Price R, Smithson L, Zwizinski C, Dickson DW, Golde T, McGowan E, Abeta40 inhibits amyloid deposition in vivo, J. Neurosci 27 (2007) 627–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Asami-Odaka A, Ishibashi Y, Kikuchi T, Kitada C, Suzuki N, Long amyloid betaprotein secreted from wild-type human neuroblastoma IMR-32 cells, Biochemistry 34 (1995) 10272–10278. [DOI] [PubMed] [Google Scholar]
  • [14].Jarrett JT, Berger EP, Lansbury PT Jr., The C-terminus of the beta protein is critical in amyloidogenesis, Ann. N. Y. Acad. Sci 695 (1993) 144–148. [DOI] [PubMed] [Google Scholar]
  • [15].Murray MM, Bernstein SL, Nyugen V, Condron MM, Teplow DB, Bowers MT, Amyloid beta protein: Abeta40 inhibits Abeta42 oligomerization, J. Am. Chem. Soc 131 (2009) 6316–6317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Yan Y, Wang C, Abeta40 protects non-toxic Abeta42 monomer from aggregation, J. Mol. Biol 369 (2007) 909–916. [DOI] [PubMed] [Google Scholar]
  • [17].Jan A, Gokce O, Luthi-Carter R, Lashuel HA, The ratio of monomeric to aggregated forms of Abeta40 and Abeta42 is an important determinant of amyloid-beta aggregation, fibrillogenesis, and toxicity, J. Biol. Chem 283 (2008) 28176–28189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Iijima K, Liu HP, Chiang AS, Hearn SA, Konsolaki M, Zhong Y, Dissecting the pathological effects of human Abeta40 and Abeta42 in Drosophila: a potential model for Alzheimer’s disease, Proc. Natl. Acad. Sci. U.S.A 101 (2004) 6623–6628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, Yu C, Skipper L, Murphy MP, Beard J, Das P, Jansen K, Delucia M, Lin WL, Dolios G, Wang R, Eckman CB, Dickson DW, Hutton M, Hardy J, Golde T, Abeta42 is essential for parenchymal and vascular amyloid deposition in mice, Neuron 47 (2005) 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Wolfe MS, When loss is gain: reduced presenilin proteolytic function leads to increased Abeta42/Abeta40. Talking Point on the role of presenilin mutations in Alzheimer disease, EMBO Rep. 8 (2007) 136–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].De Strooper B, Loss-of-function presenilin mutations in Alzheimer disease. Talking Point on the role of presenilin mutations in Alzheimer disease, EMBO Rep. 8 (2007) 141–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Hutton M, Lendon C, Rizzu P, Baker M, Froelich S, Houlden H, Pickering-Brown S, Chakraverty S, Isaacs A, Grover A, Hackett J, Adamson J, Lincoln S, Dickson D, Davies P, Peterson R, Stevens M, de Graafif E, Wauters E, van Baren J, Hillebrand M, Joosse M, Kwon J, Nowotny P, Che L, Norton J, Morris J, Ree L, Trojanowski J, Basun H, Lannfelt L, Neystat M, Fahn S, Dark F, Tannenberg T, Dodd P, Hayward N, Kwok J, Schofield P, Andreadis A, Snowden J, Craufurd D, Neary D, Owen F, Oostra B, Hardy J, Goate A, van Swieten J, Mann D, Lynch T, Heutink P, Association of missense and 5’-splice-site mutations in tau with the inherited FTDP-17, Nature 393 (1998) 702–705. [DOI] [PubMed] [Google Scholar]
  • [23].Lewis J, Dickson DW, Lin WL, Chisholm L, Corral A, Jones G, Yen SH, Sahara N, Skipper L, Yager D, Eckman C, Hardy J, Hutton M, McGowan E, Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP, Science 293 (2001) 1487–1491. [DOI] [PubMed] [Google Scholar]
  • [24].Gotz J, Chen F, van Dorpe J, Nitsch RM, Formation of neurofibrillary tangles in P3011 tau transgenic mice induced by Abeta 42 fibrils, Science 293 (2001) 1491–1495. [DOI] [PubMed] [Google Scholar]
  • [25].Lee EB, Leng LZ, Zhang B, Kwong L, Trojanowski JQ, Abel T, Lee VM, Targeting amyloid-beta peptide (Abeta) oligomers by passive immunization with a conformation-selective monoclonal antibody improves learning and memory in Abeta precursor protein (APP) transgenic mice, J. Biol. Chem 281 (2006) 4292–4299. [DOI] [PubMed] [Google Scholar]
  • [26].Flaherty DB, Soria JP, Tomasiewicz HG, Wood JG, Phosphorylation of human tau protein by microtubule-associated kinases: GSK3beta and cdk5 are key participants, J. Neurosci. Res 62 (2000) 463–472. [DOI] [PubMed] [Google Scholar]
  • [27].Reynolds CH, Betts JC, Blackstock WP, Nebreda AR, Anderton BH, Phosphorylation sites on Tau identified by nanoelectrospray mass spectrometry, J. Neurochem 74 (2000) 1587–1595. [DOI] [PubMed] [Google Scholar]
  • [28].Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van Gorp S, Nazor KL, Boscolo FS, Carson CT, Laurent LC, Marsala M, Gage FH, Remes AM, Koo EH, Goldstein LS, Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells, Nature 482 (2012) 216–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Liu Q, Waltz S, Woodruff G, Ouyang J, Israel MA, Herrera C, Sarsoza F, Tanzi RE, Koo EH, Ringman JM, Goldstein LS, Wagner SL, Yuan SH, Effect of potent gamma-secretase modulator in human neurons derived from multiple presenilin 1-induced pluripotent stem cell mutant carriers, JAMA Neurol. 71 (2014) 1481–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Yahata N, Asai M, Kitaoka S, Takahashi K, Asaka I, Hioki H, Kaneko T, Maruyama K, Saido TC, Nakahata T, Asada T, Yamanaka S, Iwata N, Inoue H, Anti-abeta drug screening platform using human iPS cell-derived neurons for the treatment of Alzheimer’s disease, PLoS One 6 (2011) e25788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E, Development of tau aggregation inhibitors for Alzheimer’s disease, Angew. Chem. Int. Ed. Engl 48 (2009) 1740–1752. [DOI] [PubMed] [Google Scholar]
  • [32].Hardy J, Allsop D, Amyloid deposition as the central event in the aetiology of Alzheimer’s disease, Trends Pharmac. 12 (1991) 383–388. [DOI] [PubMed] [Google Scholar]
  • [33].Hardy J, Selkoe DJ, The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics, Science 297 (2002) 353–356. [DOI] [PubMed] [Google Scholar]
  • [34].Wolfe MS, Xia W, Ostaszewski BL, Diehl TS, Kimberly WT, Selkoe DJ, Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and g-secretase activity, Nature 398 (1999) 513–517. [DOI] [PubMed] [Google Scholar]
  • [35].Francis R, McGrath G, Zhang J, Ruddy D, Sym M, Apfeld J, Nicoll M, Maxwell M, Hai B, Ellis MC, Parks AL, Xu W, Li J, Gurney M, Myers RL, Himes CS, Hiebsch R, Ruble C, Nye JS, Curtis D, Aph-1 and pen-2 are required for Notch pathway signaling, g-secretase cleavage of bAPP and presenilin protein accumulation, Dev. Cell 3 (2002) 85–97. [DOI] [PubMed] [Google Scholar]
  • [36].Crystal A, Morais VA, Pierson TC, Pijak DS, Carlin D, Lee VM, Dorns RW, Membrane topology of gamma-secretase component PEN-2, J. Biol. Chem 278 (2003) 20117–20123. [DOI] [PubMed] [Google Scholar]
  • [37].Baulac S, LaVoie MJ, Kimberly WT, Strahle J, Wolfe MS, Selkoe DJ, Xia W, Functional gamma-secretase complex assembly in Golgi/trans-Golgi network: interactions among presenilin, nicastrin, Aph1, Pen-2, and gamma-secretase substrates, Neurobiol. Dis 14 (2003) 194–204. [DOI] [PubMed] [Google Scholar]
  • [38].De Strooper B, Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-secretase complex, Neuron 38 (2003) 9–12. [DOI] [PubMed] [Google Scholar]
  • [39].Hu Y, Fortini M, Different cofactor activities in gamma-secretase assembly: evidence for a nicastrin-Aph-1 subcomplex, J. Cell. Biol 161 (2003) 685–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kimberly W, LaVoie M, Ostaszewski BL, Ye W, Wolfe MS, Selkoe DJ, Gamma- secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2, Proc. Natl. Acad. Sci. U.S.A 100 (2003) 6382–6387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Luo WJ, Wang H, Li H, Kim BS, Shah S, Lee HJ, Thinakaran G, Kim TW, Yu G, Xu H, PEN-2 and APH-1 coordinately regulate proteolytic processing of presenilin 1, J. Biol. Chem 278 (2003) 7850–7854. [DOI] [PubMed] [Google Scholar]
  • [42].Takasugi N, Tomita T, Hayashi I, Tsuruoka M, Niimura M, Takahashi Y, Thinakaran G, Iwatsubo T, The role of presenilin cofactors in the gamma-secretase complex, Nature 422 (2003) 438–441. [DOI] [PubMed] [Google Scholar]
  • [43].Edbauer D, Winkler E, Regula JT, Pesold B, Steiner H, Haass C, Reconstitution of gamma-secretase activity, Nat. Cell. Biol 5 (2003) 486–488. [DOI] [PubMed] [Google Scholar]
  • [44].Ahn K, Shelton CC, Tian Y, Zhang X, Gilchrist ML, Sisodia SS, Li YM, Activation and intrinsic gamma-secretase activity of presenilin 1, Proc. Natl. Acad. Sci. U.S.A 107 (2010) 21435–21440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Lessard CB, Wagner SL, Koo EH, And four equals one: presenilin takes the gamma-secretase role by itself, Proc. Natl. Acad. Sci. U.S.A 107 (2010) 21236–21237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Li YM, Xu M, Lai MT, Huang Q, Castro JL, DiMuzio-Mower J, Harrison T, Lellis C, Nadin A, Neduvelil JG, Register RB, Sardana MK, Shearman MS, Smith AL, Shi XP, Yin KC, Shafer JA, Gardell SJ, Photoactivated gamma-secretase inhibitors directed to the active site covalently label presenilin 1, Nature 405 (2000) 689–694. [DOI] [PubMed] [Google Scholar]
  • [47].Siemers E, Skinner M, Dean RA, Gonzales C, Satterwhite J, Farlow M, Ness D, May PC, Safety, tolerability, and changes in amyloid beta concentrations after administration of a gamma-secretase inhibitor in volunteers, Clin. Neuropharmacol 28 (2005) 126–132. [DOI] [PubMed] [Google Scholar]
  • [48].Fleisher AS, Raman R, Siemers ER, Becerra L, Clark CM, Dean RA, Farlow MR, Galvin JE, Peskind ER, Quinn JF, Sherzai A, Sowell BB, Aisen PS, Thal LJ, Phase 2 safety trial targeting amyloid beta production with a gamma-secretase inhibitor in Alzheimer disease, Arch. Neurol 65 (2008) 1031–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, He F, Sun X, Thomas RG, Aisen PS, Siemers E, Sethuraman G, Mohs R, A phase 3 trial of semagacestat for treatment of Alzheimer’s disease, N. Engl. J. Med 369 (2013) 341–350. [DOI] [PubMed] [Google Scholar]
  • [50].Panelos J, Massi D, Emerging role of Notch signaling in epidermal differentiation and skin cancer, Cancer Biol. Ther 8 (2009) 1986–1993. [DOI] [PubMed] [Google Scholar]
  • [51].Shelton CC, Zhu L, Chau D, Yang L, Wang R, Djaballah H, Zheng H, Li YM, Modulation of gamma-secretase specificity using small molecule allosteric inhibitors, Proc. Natl. Acad. Sci. U.S.A 106 (2009) 20228–20233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Kukar T, Murphy MP, Eriksen JL, Sagi SA, Weggen S, Smith TE, Ladd T, Khan MA, Kache R, Beard J, Dodson M, Merit S, Ozols VV, Anastasiadis PZ, Das P, Fauq A, Koo EH, Golde TE, Diverse compounds mimic Alzheimer disease-causing mutations by augmenting Abeta42 production, Nat. Med 11 (2005) 545–550. [DOI] [PubMed] [Google Scholar]
  • [53].Cole DC, Stock JR, Kreft AF, Antane M, Aschmies SH, Atchison KP, Casebier DS, Comery TA, Diamantidis G, Ellingboe JW, Harrison BL, Hu Y, Jin M, Kubrak DM, Lu P, Mann CW, Martone RL, Moore WJ, Oganesian A, Riddell DR, Sonnenberg-Reines J, Sun SC, Wagner E, Wang Z, Woller KR, Xu Z, Zhou H, Jacobsen JS, (S)-N-(5-Chlorothiophene-2-sulfonyl)-beta,betadiethylalaninol a Notch-1-sparing gamma-secretase inhibitor, Bioorg. Med. Chem. Lett 19 (2009) 926–929. [DOI] [PubMed] [Google Scholar]
  • [54].Martone RL, Zhou H, Atchison K, Comery T, Xu JZ, Huang X, Gong X, Jin M, Kreft A, Harrison B, Mayer SC, Aschmies S, Gonzales C, Zaleska MM, Riddell DR, Wagner E, Lu P, Sun SC, Sonnenberg-Reines J, Oganesian A, Adkins K, Leach MW, Clarke DW, Huryn D, Abou-Gharbia M, Magolda R, Bard J, Frick G, Raje S, Forlow SB, Balliet C, Burczynski ME, Reinhart PH, Wan HI, Pangalos MN, Jacobsen JS, Begacestat (GSI-953): a novel, selective thiophene sulfonamide inhibitor of amyloid precursor protein gamma-secretase for the treatment of Alzheimer’s disease, J. Pharmacol. Exp. Ther 331 (2009) 598–608. [DOI] [PubMed] [Google Scholar]
  • [55].Mayer SC, Kreft AF, Harrison B, Abou-Gharbia M, Antane M, Aschmies S, Atchison K, Chlenov M, Cole DC, Comery T, Diamantidis G, Ellingboe J, Fan K, Galante R, Gonzales C, Ho DM, Hoke ME, Hu Y, Huryn D, Jain U, Jin M, Kremer K, Kubrak D, Lin M, Lu P, Magolda R, Martone R, Moore W, Oganesian A, Pangalos MN, Porte A, Reinhart P, Resnick L, Riddell DR, Sonnenberg-Reines J, Stock JR, Sun SC, Wagner E, Wang T, Woller K, Xu Z, Zaleska MM, Zeldis J, Zhang M, Zhou H, Jacobsen JS, Discovery of begacestat, a Notch-1-sparing gamma-secretase inhibitor for the treatment of Alzheimer’s disease, J. Med. Chem 51 (2008) 7348–7351. [DOI] [PubMed] [Google Scholar]
  • [56].Gillman KW, Starrett JE, Parker MF, Xie K, Bronson JJ, Marcin LR, McElhone KE, Bergstrom CP, Mate RA, Williams R, Meredith JE, Burton CR, Barten DM, Toyn JH, Roberts SB, Lentz KA, Houston JG, Zaczek R, Albright CF, Decicco CP, Macor JE, Olson RE, Discovery and evaluation of BMS-708163, a potent, selective and orally bioavailable g-secretase inhibitor, ACS Med. Chem. Lett 1 (2010) 120–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Crump CJ, Castro SV, Wang F, Pozdnyakov N, Ballard TE, Sisodia SS, Bales KR, Johnson DS, Li Y-M, BMS-708,163 targets presenilin and lacks Notch-sparing activity, Biochemistry 51 (2012) 7209–7211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Tong G, Wang JS, Sverdlov O, Huang SP, Slemmon R, Croop R, Castaneda L, Gu H, Wong O, Li H, Berman RM, Smith C, Albright CF, Dockens RC, Multicenter, randomized, double-blind, placebo-controlled, single-ascending dose study of the oral gamma-secretase inhibitor BMS-708163 (avagacestat): tolerability profile, pharmacokinetic parameters, and pharmacodynamic markers, Clin. Ther 34 (2012) 654–667. [DOI] [PubMed] [Google Scholar]
  • [59].Coric V, van Dyck CH, Salloway S, Andreasen N, Brody M, Richter RW, Soininen H, Thein S, Shiovitz T, Pilcher G, Colby S, Rollin L, Dockens R, Pachai C, Portelius E, Andreasson U, Blennow K, Soares H, Albright C, Feldman HH, Berman RM, Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease, Arch. Neurol 13 (2012) 1–12. [DOI] [PubMed] [Google Scholar]
  • [60].De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS, Ray WJ, Goate A, Kopan R, A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain, Nature 398 (1999) 518–522. [DOI] [PubMed] [Google Scholar]
  • [61].Okochi M, Steiner H, Fukumori A, Tanii H, Tomita T, Tanaka T, Iwatsubo T, Kudo T, Takeda M, Haass C, Presenilins mediate a dual intramembranous gamma-secretase cleavage of Notch-1, EMBO J. 21 (2002) 5408–5416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Fortini ME, Gamma-secretase-mediated proteolysis in cell-surface-receptor signalling, Nat. Rev. Mol. Cell. Biol 3 (2002) 673–684. [DOI] [PubMed] [Google Scholar]
  • [63].Kopan R, Goate A, Aph-2/Nicastrin: an essential component of gamma-secretase and regulator of Notch signaling and Presenilin localization, Neuron 33 (2002) 321–324. [DOI] [PubMed] [Google Scholar]
  • [64].Xia W, Wolfe M, Intramembrane proteolysis by presenilin and presenilin-like proteases, J. Cell. Sci 116 (2003) 2839–2844. [DOI] [PubMed] [Google Scholar]
  • [65].Haapasalo A, Kovacs DM, The many substrates of presenilin/gamma-secretase, J. Alzheimers Dis 25 (2011) 3–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Searfoss GH, Jordan WH, Calligaro DO, Galbreath EJ, Schirtzinger LM, Berridge R, Gao H, Higgins MA, May PC, Ryan TP, Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma-secretase inhibitor, J. Biol. Chem 278 (2003) 46107–46116. [DOI] [PubMed] [Google Scholar]
  • [67].Wong GT, Manfra D, Poulet FM, Zhang Q, Josien H, Bara T, Engstrom L, Pinzon-Ortiz M, Fine JS, Lee HJ, Zhang L, Higgins GA, Parker EM, Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation, J. Biol. Chem 279 (2004) 12876–12882 Epub 12004 Jan 12876. [DOI] [PubMed] [Google Scholar]
  • [68].Campbell WA, Yang H, Zetterberg H, Baulac S, Sears JA, Liu T, Wong ST, Zhong TP, Xia W, Zebrafish lacking Alzheimer presenilin enhancer 2 (Pen-2) demonstrate excessive p53-dependent apoptosis and neuronal loss, J. Neurochem 96 (2006) 1423–1440. [DOI] [PubMed] [Google Scholar]
  • [69].Zetterberg H, Campbell WA, Yang HW, Xia W, The cytosolic loop of the gamma -secretase component presenilin enhancer 2 (Pen-2) protects zebrafish embryos from apoptosis, J. Biol. Chem 281 (2006) 11933–11939. [DOI] [PubMed] [Google Scholar]
  • [70].Geling A, Steiner H, Willem M, Bally-Cuif L, Haass C, A gamma-secretase inhibitor blocks Notch signaling in vivo and causes a severe neurogenic phenotype in zebrafish, EMBO Rep. 3 (2002) 688–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Yang T, Arslanova D, Gu Y, Augelli-Szafran C, Xia W, Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein, Mol. Brain 1 (2008) 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Yang T, Arslanova D, Xu X, Li YM, Xia W, In vivo manifestation of Notch related phenotypes in zebrafish treated with Alzheimer’s amyloid reducing gamma-secretase inhibitors, J. Neurochem 113 (2010) 1200–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Arslanova D, Yang T, Xu X, Wong ST, Augelli-Szafran CE, Xia W, Phenotypic analysis of images of zebrafish treated with Alzheimer’s gamma-secretase in-hibitors, BMC Biotechnol. 10 (2010) 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Heilig EA, Xia W, Shen J, Kelleher RJ 3rd, A presenilin-1 mutation identified in familial Alzheimer disease with cotton wool plaques causes a nearly complete loss of gamma-secretase activity, J. Biol. Chem 285 (2010) 22350–22359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Yu H, Saura C, Choi S, Sun L, Yang X, Handler M, Kawarabayashi T, Younkin L, Fedeles B, Wilson M, Younkin S, Kandel E, Kirkwood A, Shen J, APP processing and synaptic plasticity in presenilin-1 conditional knockout mice, Neuron 31 (2001) 713–726. [DOI] [PubMed] [Google Scholar]
  • [76].Saura CA, Choi SY, Beglopoulos V, Malkani S, Zhang D, Rao BS, Chattarji S, Kelleher RJ 3rd, Kandel ER, Duff K, Kirkwood A, Shen J, Loss of presenilin function causes impairments of memory and synaptic plasticity followed by age-dependent neurodegeneration, Neuron 42 (2004) 23–36. [DOI] [PubMed] [Google Scholar]
  • [77].Tabuchi K, Chen G, Sudhof TC, Shen J, Conditional forebrain inactivation of nicastrin causes progressive memory impairment and age-related neurodegeneration, J. Neurosci 29 (2009) 7290–7301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Weggen S, Eriksen JL, Das P, Sagi SA, Wang R, Pietrzik CU, Findlay KA, Smith TE, Murphy MP, Bulter T, Kang DE, Marquez-Sterling N, Golde TE, Koo EH, A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity, Nature 414 (2001) 212–216. [DOI] [PubMed] [Google Scholar]
  • [79].Lessard CB, Cottrell BA, Maruyama H, Suresh S, Golde TE, Koo EH, Gamma-secretase modulators and APH1 isoforms modulate gamma-secretase cleavage but not position of epsilon-cleavage of the amyloid precursor protein (APP), PLoS One 10 (2015) e0144758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Ohki Y, Higo T, Uemura K, Shimada N, Osawa S, Berezovska O, Yokoshima S, Fukuyama T, Tomita T, Iwatsubo T, Phenylpiperidine-type gamma-secretase modulators target the transmembrane domain 1 of presenilin 1, EMBO J. 30 (2011) 4815–4824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Crump CJ, Fish BA, Castro SV, Chau DM, Gertsik N, Ahn K, Stiff C, Pozdnyakov N, Bales KR, Johnson DS, Li YM, Piperidine acetic acid based gamma-secretase modulators directly bind to presenilin-1, ACS Chem. Neurosci 2 (2011) 705–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Ebke A, Luebbers T, Fukumori A, Shirotani K, Haass C, Baumann K, Steiner H, Novel gamma-secretase enzyme modulators directly target presenilin protein, J. Biol. Chem 286 (2011) 37181–37186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Kukar TL, Ladd TB, Bann MA, Fraering PC, Narlawar R, Maharvi GM, Healy B, Chapman R, Welzel AT, Price RW, Moore B, Rangachari V, Cusack B, Eriksen J, Jansen-West K, Verbeeck C, Yager D, Eckman C, Ye W, Sagi S, Cottrell BA, Torpey J, Rosenberry TL, Fauq A, Wolfe MS, Schmidt B, Walsh DM, Koo EH, Golde TE, Substrate-targeting gamma-secretase modulators, Nature 453 (2008) 925–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Lue LF, Walker DG, Rogers J, Modeling microglial activation in Alzheimer’s disease with human postmortem microglial cultures, Neurobiol. Aging 22 (2001) 945–956. [DOI] [PubMed] [Google Scholar]
  • [85].Janelsins MC, Mastrangelo MA, Park KM, Sudol KL, Narrow WC, Oddo S, LaFerla FM, Callahan LM, Federoff HJ, Bowers WJ, Chronic neuron-specific tumor necrosis factor-alpha expression enhances the local inflammatory environment ultimately leading to neuronal death in 3xTg-AD mice, Am. J. Pathol 173 (2008) 1768–1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Sheedy FJ, Grebe A, Rayner KJ, Kalantari P, Ramkhelawon B, Carpenter SB, Becker CE, Ediriweera HN, Mullick AE, Golenbock DT, Stuart LM, Latz E, Fitzgerald KA, Moore KJ, CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation, Nat. Immunol 14 (2013) 812–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, Golenbock DT, The NALP3 inflammasome is involved in the innate immune response to amyloid-beta, Nat. Immunol 9 (2008) 857–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT, NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice, Nature 493 (2013) 674–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J, TREM2 variants in Alzheimer’s disease, N. Engl. J. Med 368 (2013) 117–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, Rujescu D, Hampel H, Giegling I, Andreassen OA, Engedal K, Ulstein I, Djurovic S, Ibrahim-Verbaas C, Hofman A, Ikram MA, van Duijn CM, Thorsteinsdottir U, Kong A, Stefansson K, Variant of TREM2 associated with the risk of Alzheimer’s disease, N. Engl. J. Med 368 (2013) 107–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE, Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta, Neuron 78 (2013) 631–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A, Morris MC, Evans DA, Johnson K, Sperling RA, Schneider JA, Bennett DA, De Jager PL, CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology, Nat. Neurosci 16 (2013) 848–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R, Fluder E, Clurman B, Melquist S, Narayanan M, Suver C, Shah H, Mahajan M, Gillis T, Mysore J, MacDonald ME, Lamb JR, Bennett DA, Molony C, Stone DJ, Gudnason V, Myers AJ, Schadt EE, Neumann H, Zhu J, Emilsson V, Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease, Cell 153 (2013) 707–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Oehlrich D, Berthelot DJC, Gijsen HJM, g-Secretase modulators as potential disease modifying anti-Alzheimer’s drugs, J. Med. Chem 54 (2011) 669–698. [DOI] [PubMed] [Google Scholar]
  • [95].Crump CJ, Johnson DS, Li YM, Target of gamma-secretase modulators, presenilin marks the spot, EMBO J. 30 (2011) 4696–4698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Weggen S, Eriksen JL, Sagi SA, Pietrzik CU, Golde TE, Koo EH, Abeta42-lowering nonsteroidal anti-inflammatory drugs preserve intramembrane cleavage of the amyloid precursor protein (APP) and ErbB-4 receptor and signaling through the APP intracellular domain, J. Biol. Chem 278 (2003) 30748–30754. [DOI] [PubMed] [Google Scholar]
  • [97].Gasparini L, Rusconi L, Xu H, del Soldato P, Ongini E, Modulation of beta-amyloid metabolism by non-steroidal anti-inflammatory drugs in neuronal cell cultures, J. Neurochem 88 (2004) 337–348. [DOI] [PubMed] [Google Scholar]
  • [98].Qin W, Ho L, Pompl PN, Peng Y, Zhao Z, Xiang Z, Robakis NK, Shioi J, Suh J, Pasinetti GM, Cyclooxygenase (COX)-2 and COX-1 potentiate beta-amyloid peptide generation through mechanisms that involve gamma-secretase activity, J. Biol. Chem 278 (2003) 50970–50977 Epub 52003, Sep 50924. [DOI] [PubMed] [Google Scholar]
  • [99].Yan Q, Zhang J, Liu H, Babu-Khan S, Vassar R, Biere AL, Citron M, Landreth G, Anti-inflammatory drug therapy alters beta-amyloid processing and deposition in an animal model of Alzheimer’s disease, J. Neurosci 23 (2003) 7504–7509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLendon DC, Ozols VV, Jessing KW, Zavitz KH, Koo EH, Golde TE, NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower abeta 42 in vivo, J. Clin. Invest 112 (2003) 440–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Weggen S, Eriksen JL, Sagi SA, Pietrzik CU, Ozols V, Fauq A, Golde TE, Koo EH, Evidence that nonsteroidal anti-inflammatory drugs decrease amyloid beta 42 production by direct modulation of gamma-secretase activity, J. Biol. Chem 278 (2003) 31831–31837. [DOI] [PubMed] [Google Scholar]
  • [102].Zhou Y, Su Y, Li B, Liu F, Ryder JW, Wu X, Gonzalez-DeWhitt PA, Gelfanova V, Hale JE, May PC, Paul SM, Ni B, Nonsteroidal anti-inflammatory drugs can lower amyloidogenic Abeta42 by inhibiting Rho, Science 302 (2003) 1215–1217. [DOI] [PubMed] [Google Scholar]
  • [103].Imbimbo BP, Del Giudice E, Cenacchi V, Volta R, Villetti G, Facchinetti F, Riccardi B, Puccini P, Moretto N, Grassi F, Ottonello S, Leon A, In vitro and in vivo profiling of CHF5022 and CHF5074 two beta-amyloid1-42 lowering agents, Pharmacol. Res 55 (2007) 318–328. [DOI] [PubMed] [Google Scholar]
  • [104].Imbimbo BP, Hutter-Paier B, Villetti G, Facchinetti F, Cenacchi V, Volta R, Lanzillotta A, Pizzi M, Windisch M, CHF5074, a novel gamma-secretase modulator, attenuates brain beta-amyloid pathology and learning deficit in a mouse model of Alzheimer’s disease. Br. J. Pharmacol 156 (2009) 982–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Imbimbo BP, Giardino L, Sivilia S, Giuliani A, Gusciglio M, Pietrini V, Del Giudice E, D’Arrigo A, Leon A, Villetti G, Calza L, CHF5074, a novel gamma-secretase modulator, restores hippocampal neurogenesis potential and reverses contextual memory deficit in a transgenic mouse model of Alzheimer’s disease, J. Alzheimers Dis 20 (2010) 159–173. [DOI] [PubMed] [Google Scholar]
  • [106].Ross J, Sharma S, Winston J, Nunez M, Bottini G, Franceschi M, Scarpini E, Frigerio E, Fiorentini F, Fernandez M, Sivilia S, Giardino L, Calza L, Norris D, Cicirello H, Casula D, Imbimbo BP, CHF5074 reduces biomarkers of neuroinflammation in patients with mild cognitive impairment: A 12-week, double-blind, placebo- controlled study, Curr. Alzheimer Res 10 (2013) 742–753. [DOI] [PubMed] [Google Scholar]
  • [107].Ross J, Sharma S, Chatterjee A, Winston J, Bottini G, Franceschi M, Fernandez M, Giardino L, Calza L, Norris D, Cicirello H, Imbimbo BP, Sustained cognitive benefit in patients with mild cognitive impairment (MCI) upon prolonged treatment with CHF5074, Alzheimer’s Dementia (2013) 03–06-05. [Google Scholar]
  • [108].Zhao C, Ling Z, Newman MB, Bhatia A, Carvey PM, TNF-alpha knockout and minocycline treatment attenuates blood-brain barrier leakage in MPTP-treated mice, Neurobiol. Dis 26 (2007) 36–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Chen G, Goeddel DV, TNF-R1 signaling: a beautiful pathway, Science 296 (2002) 1634–1635. [DOI] [PubMed] [Google Scholar]
  • [110].Imbimbo BP, Del Giudice E, Colavito D, D’Arrigo A, Dalle Carbonare M, Villetti G, Facchinetti F, Volta R, Pietrini V, Baroc MF, Serneels L, De Strooper B, Leon A, l-(3’,4’-Dichloro-2-fluoro[1,1’-biphenyl]-4-yl)-cyclopropane-carboxylic acid (CHF5074), a novel gamma-secretase modulator, reduces brain beta-amyloid pathology in a transgenic mouse model of Alzheimer’s disease without causing peripheral toxicity, J. Pharmacol. Exp. Ther 323 (2007) 822–830. [DOI] [PubMed] [Google Scholar]
  • [111].Ling IF, Golde TE, Galasko DR, Koo EH, Modulation of Abeta42 in vivo by gamma-secretase modulator in primates and humans, Alzheimers Res. Ther 7 (2015) 55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Hashimoto TI, Hagiwara A, Murata H, Takenaka OMT, E2012: A novel Gamma-secretase modulator; Pharmacology part, International Conference on Alzheimer’s Disease 2010 (2010) Abstract P2011–2236. [Google Scholar]
  • [113].Amino HH, Murata H, Watanabe Y, Sasaki H, Miyagawa TT, E2012: A novel Gamma-secretase modulator; Mechanism of action, International Conference on Alzheimer’s Disease 2010 (2010) Abstract P2013–2307. [Google Scholar]
  • [114].Kounnas MZ, Danks AM, Cheng S, Tyree C, Ackerman E, Zhang X, Ahn K, Nguyen P, Comer D, Mao L, Yu C, Pleynet D, Digregorio PJ, Velicelebi G, Stauderman KA, Comer WT, Mobley WC, Li YM, Sisodia SS, Tanzi RE, Wagner SL, Modulation of gamma-secretase reduces beta-amyloid deposition in a transgenic mouse model of Alzheimer’s disease, Neuron 67 (2010) 769–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Fischer C, Shah S, Hughes BL, Nikov GN, Crispino JL, Middleton RE, Szewczak AA, Munoz B, Shearman MS, Quinazolinones as gamma-secretase modulators, Bioorg. Med. Chem. Lett 21 (2011) 773–776. [DOI] [PubMed] [Google Scholar]
  • [116].Fischer C, Zultanski SL, Zhou H, Methot JL, Brown WC, Mampreian DM, Schell AJ, Shah S, Nuthall H, Hughes BL, Smotrov N, Kenific CM, Cruz JC, Walker D, Bouthillette M, Nikov GN, Savage DF, Jeliazkova-Mecheva VV, Diaz D, Szewczak AA, Bays N, Middleton RE, Munoz B, Shearman MS, Triazoles as gamma-secretase modulators, Bioorg Med. Chem. Lett 21 (2011) 4083–4087. [DOI] [PubMed] [Google Scholar]
  • [117].Rivkin A, Ahearn SP, Chichetti SM, Hamblett CL, Garcia Y, Martinez M, Hubbs JL, Reutershan MH, Daniels MH, Siliphaivanh P, Otte KM, Li C, Rosenau A, Surdi LM, Jung J, Hughes BL, Crispino JL, Nikov GN, Middleton RE, Moxham CM, Szewczak AA, Shah S, Moy LY, Kenific CM, Tanga F, Cruz JC, Andrade P, Angagaw MH, Shomer NH, Miller T, Munoz B, Shearman MS, Purine derivatives as potent gamma-secretase modulators, Bioorg. Med. Chem. Lett 20 (2010) 2279–2282. [DOI] [PubMed] [Google Scholar]
  • [118].Tomita T, Secretase inhibitors and modulators for Alzheimer’s disease treatment, Expert Rev. Neurother 9 (2009) 661–679. [DOI] [PubMed] [Google Scholar]
  • [119].Rivkin A, Ahearn SP, Chichetti SM, Kim YR, Li C, Rosenau A, Kattar SD, Jung J, Shah S, Hughes BL, Crispino JL, Middleton RE, Szewczak AA, Munoz B, Shearman MS, Piperazinyl pyrimidine derivatives as potent gamma-secretase modulators. Bioorg. Med. Chem. Lett 20 (2010) 1269–1271. [DOI] [PubMed] [Google Scholar]
  • [120].Stanton MG, Hubbs J, Sloman D, Hamblett C, Andrade P, Angagaw M, Bi G, Black RM, Crispino J, Cruz JC, Fan E, Farris G, Hughes BL, Kenific CM, Middleton RE, Nikov G, Sajonz P, Shah S, Shomer N, Szewczak AA, Tanga F, Tudge MT, Shearman M, Munoz B, Fluorinated piperidine acetic acids as gamma-secretase modulators, Bioorg. Med. Chem. Lett 20 (2010) 755–758. [DOI] [PubMed] [Google Scholar]
  • [121].Hall A, Elliott RL, Giblin GM, Hussain I, Musgrave J, Naylor A, Sasse R, Smith B, Piperidine-derived gamma-secretase modulators, Bioorg. Med. Chem. Lett 20 (2010) 1306–1311. [DOI] [PubMed] [Google Scholar]
  • [122].Hussain I, Harrison DC, Hawkins J, Chapman T, Marshall I, Facci L, Ahmed S, Brackenborough K, Skaper SD, Mead TL, Smith BB, Giblin GM, Hall A, Gonzalez MI, Richardson JC, TASTPM mice expressing amyloid precursor protein and presenilin-1 mutant transgenes are sensitive to gamma-secretase modulation and amyloid-beta lowering by GSM-10h, Neurodegener. Dis 8 (2011) 15–24. [DOI] [PubMed] [Google Scholar]
  • [123].Hawkins J, Harrison DC, Ahmed S, Davis RP, Chapman T, Marshall I, Smith B, Mead TL, Medhurst A, Giblin GM, Hall A, Gonzalez MI, Richardson J, Hussain I, Dynamics of Abeta42 reduction in plasma, CSF and brain of rats treated with the gamma-secretase modulator, GSM-10h, Neurodegener. Dis 8 (2011) 455–464. [DOI] [PubMed] [Google Scholar]
  • [124].Page RM, Baumann K, Tomioka M, Perez-Revuelta BI, Fukumori A, Jacobsen H, Flohr A, Luebbers T, Ozmen L, Steiner H, Haass C, Generation of Abeta38 and Abeta42 is independently and differentially affected by familial Alzheimer disease-associated presenilin mutations and gamma-secretase modulation, J. Biol. Chem 283 (2008) 677–683. [DOI] [PubMed] [Google Scholar]
  • [125].Chesworth RS, Rogers G, Felsenstein K, Sarma K, Baruah BBBV, Nikonov S, Huang G, Zhang G, Mueller RAM, Tu F, Nolan Z, Wen S, Spaulding MDC, Hrdlicka F, Yang L, Leventhal Z, Hopp SML, Patzke MJ, Koenig H, Vulsteke G, De Strooper VBA, Lee M, Hodgdon W, Costa H, Sigel ED, Gamma secretase modulators that acutely reduce soluble Aβ 42 levels in Non-transgenic rodents, 239th National Meeting of the American Chemical Society (2010). [Google Scholar]
  • [126].Felsenstein KMS, Yang D, Hodgdon Z, Costa H, Nolan SWD, Lee M, Hrdlicka W, Catana L, Albayya FT, Patzke Z, Chesworth H, Shapiro R, Zaninovic M, Pharmacokinetic and pharmacodynamic analysis of the gamma-secretase modulator (GSM) EVP-0015962, International Conference on Alzheimer’s Disease (2010). [Google Scholar]
  • [127].Rogers KL, Hopp L, Miller S, Zhiyong M-J, Hrdlicka YLL, Hodgdon W, Nolan H, Wen S, Koperniak MTS, Catana D, Chesworth F, Shapiro RGC, Ahlijanian D, Koenig M, Felsenstein G, The gamma secretase modulator EVP-0015962 improves cognitive deficits in Tg2576 mice concomitant with decreases in Aβ42, International Conference on Alzheimer’s Disease (2010). [Google Scholar]
  • [128].Van Broeck B, Chen JM, Treton G, Desmidt M, Hopf C, Ramsden N, Karran E, Mercken M, Rowley A, Chronic treatment with a novel gamma-secretase modulator, JNJ-40418677, inhibits amyloid plaque formation in a mouse model of Alzheimer’s disease, Br. J. Pharmacol 163 (2011) 375–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Dockens R, Wang JS, Castaneda L, Sverdlov O, Huang SP, Slemmon R, Gu H, Wong O, Li H, Berman RM, Smith C, Albright CF, Tong G, A. Placebo-Controlled, Multiple ascending dose study to evaluate the safety, pharmacokinetics and pharmacodynamics of avagacestat (BMS-708163) in healthy young and elderly subjects, Clin. Pharmacokinet 51 (2012) 681–693. [DOI] [PubMed] [Google Scholar]
  • [130].Toyn JH, Boy KM, Raybon J, Meredith JE Jr, Robertson AS, Guss V, Hoque N, Sweeney F, Zhuo X, Clarke W, Snow K, Denton RR, Zuev D, Thompson LA, Morrison J, Grace J, Berisha F, Furlong M, Wang JS, Lentz KA, Padmanabha R, Cook L, Wei C, Drexler DM, Macor JE, Albright CF, Gasior M, Olson RE, Hong Q, Soares HD, AbuTarif M, Ahlijanian MK, Robust translation of gamma-secretase modulator pharmacology across preclinical species and human subjects, J. Pharmacol. Exp. Ther 358 (2016) 125–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Soares HD, Gasior M, Toyn JH, Wang JS, Hong Q, Berisha F, Furlong MT, Raybon J, Lentz KA, Sweeney F, Zheng N, Akinsanya B, Berman RM, Thompson LA, Olson RE, Morrison J, Drexler DM, Macor JE, Albright CF, Ahlijanian MK, AbuTarif M, The gamma-secretase modulator BMS -932481, modulates Abeta peptides in the plasma and cerebrospinal fluid of healthy volunteers, J. Pharmacol. Exp. Ther 358 (2016) 138–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Wagner SL, Rynearson KD, Duddy SK, Zhang C, Nguyen PD, Becker A, Vo U, Masliah D, Monte L, Klee JB, Echmalian CM, Xia W, Quinti L, Johnson G, Lin JH, Kim DY, Mobley WC, Rissman RA, Tanzi RE, Pharmacological and toxicological properties of the potent oral gamma-secretase modulator BPN-15606, J. Pharmacol. Exp. Ther 362 (2017) 31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, Chen H, Hooli B, Asselin C, Muffat J, Klee JB, Zhang C, Wainger BJ, Peitz M, Kovacs DM, Woolf CJ, Wagner SL, Tanzi RE, Kim DY, A three-dimensional human neural cell culture model of Alzheimer’s disease, Nature 515 (2014) 274–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Golde TE, Schneider LS, Koo EH, Anti-abeta therapeutics in Alzheimer’s dis-ease: the need for a paradigm shift, Neuron 69 (2011) 203–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Blain JF, Bursavich MG, Freeman EA, Hrdlicka LA, Hodgdon HE, Chen T, Costa DE, Harrison BA, Kapadnis S, Murphy DA, Nolan S, Tu Z, Tang C, Burnett DA, Patzke H, Koenig G, Characterization of FRM-36143 as a new gamma-secretase modulator for the potential treatment of familial Alzheimer’s disease, Alzheimers Res. Ther 8 (2016) 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, O’Gorman J, Qian F, Arastu M, Li M, Chollate S, Brennan MS, Quintero-Monzon O, Scannevin RH, Arnold HM, Engber T, Rhodes K, Ferrero J, Hang Y, Mikulskis A, Grimm J, Hock C, Nitsch RM, Sandrock A, The antibody aducanumab reduces abeta plaques in Alzheimer’s disease, Nature 537 (2016) 50–56. [DOI] [PubMed] [Google Scholar]
  • [137].Jung JI, Price AR, Ladd TB, Ran Y, Park HJ, Ceballos-Diaz C, Smithson LA, Hochhaus G, Tang Y, Akula R, Ba S, Koo EH, Shapiro G, Felsenstein KM, Golde TE, Cholestenoic acid, an endogenous cholesterol metabolite, is a potent gamma-secretase modulator, Mol. Neurodegener 10 (2015) 29. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES