Abstract
γ-Secretase is a fascinating, multi-subunit, intramembrane-cleaving protease that is now being considered as a therapeutic target for a number of diseases. Potent, orally bioavailable γ-secretase inhibitors (GSIs) have been developed and tested in humans with Alzheimer's disease (AD) and cancer. Preclinical studies also suggest the therapeutic potential for GSIs in other disease conditions. However, due to inherent mechanism based-toxicity of non-selective inhibition of γ-secretase, clinical development of GSIs will require empirical testing with careful evaluation of benefit versus risk. In addition to GSIs, compounds referred to as γ-secretase modulators (GSMs) remain in development as AD therapeutics. GSMs do not inhibit γ-secretase, but modulate γ-secretase processivity and thereby shift the profile of the secreted amyloid β peptides (Aβ) peptides produced. Although GSMs are thought to have an inherently safe mechanism of action, their effects on substrates other than the amyloid β protein precursor (APP) have not been extensively investigated. Herein, we will review the current state of development of GSIs and GSMs and explore pertinent biological and pharmacological questions pertaining to the use of these agents for select indications.
Keywords: Presenilin, γ-secretase, γ-secretase inhibitor, γ-secretase modulator, cancer, Alzheimer's disease, amyloid β protein, APP, Notch, intramembrane proteolysis
1. Introduction
In concert with three other proteins, APH1, PEN2, and Nicastrin, presenilin 1 (PSEN1) or 2 (PSEN2) function as the catalytic core of the intramembrane cleaving protease called γ-secretase [1-3]. This multi-subunit protease cleaves within the transmembrane domains (TMDs) of over 100 type 1 membrane proteins [4]. γ-Secretase was originally identified as the protease responsible for the generation of Aβ, and thus considered a prime therapeutic target in Alzheimer's disease (AD) [5-6]. However, it was soon recognized that γ-secretase catalyzed cleavages regulate a variety of signaling events by untethering the cytoplasmic domain of various transmembrane proteins from the membrane, allowing these domains to transduce signals to the nucleus [7-8]. It is now clear that regulated intramembrane proteolysis carried out by γ-secretase is another means for cells to transmit and regulate signals across a lipid bilayer, though in other cases γ-secretase may also play a role in transmembrane protein turnover [9-10].
γ-Secretase is an unusual protease. It is highly promiscuous in terms of the transmembrane domain (TMD) sequences it cleaves. Although not absolute, major determinants of cleavage appear to be prior ectodomain shedding and co-localization of γ-secretase with the resulting membrane stub in select subcellular compartments [11-12]. Studies of amyloid β protein precursor (APP) and Notch 1 indicate that γ-secretase initially cleaves the TMD of a protein at a site near the cytoplasmic face of the membrane [8, 13-15]. In APP this initial cleavage is then followed by 3-5 sequential di, tri, or tetrapeptide cleavages [14]. Thus, γ-secretase cleavage of transmembrane protein results in two potentially biologically active fragments: the cytoplasmic intracellular domain and a small secreted peptide. Figure 1 illustrates the typical processing of a generic type 1 membrane protein by γ-secretase. For most substrates, the initial γ-secretase cleavage site and the extent of processivity have not been defined.
Figure 1. A model of stepwise γ-secretase cleavage.

Typical processing of a Type 1 transmembrane protein (A), by a “sheddase” followed by sequential γ-secretase cleavages results in release of multiple potentially bioactive protein fragments: the ectodomain (B), a transmembrane carboxyl terminal fragment (CTF) or stub) (C), the cytoplasmic domain (D) and an Aβ/p3 like domain (E). Thus, γ-secretase generates potentially bioactive fragments (D) by an initial cleavage and (E) by processive step-wise cleavage GSIs inhibit the initial cleavage and GSMs alter processivity.
Primarily because γ-secretase was a therapeutic target in AD, a plethora of γ-secretase inhibitors (GSIs) have been developed that effectively inhibit γ-secretase cleavage in humans [16]. Indeed, “druggability” of γ-secretase has not been an issue even when the identity of the target was unknown in these blind screens. γ-Secretase is a highly tractable therapeutic target and numerous orally-bioavailable, brain penetrant GSIs have been developed [16-17] (see Figure 2 for examples). Many of these GSIs are highly potent and show excellent bioavailability and pharmacokinetic properties. In AD the efficacy of GSIs has been tied to inhibition of amyloid β protein (Aβ); thus, in AD, GSIs have been conceptualized as “Aβ production inhibitors” [16]. GSIs can decrease Aβ production in human and mouse brain and chronic administration decreases Aβ deposition in amyloid β protein precursor (APP) mouse models [18-21]. These GSIs have been important tools in the AD field, but also have served as essential elements of preclinical proof of concept studies for many different disease indications. In addition to GSIs, compounds referred to as γ-secretase modulators (GSMs) that modulate processivity of γ-secretase have been identified and remain in development as potentially inherently safe ways to selectively target Aβ42 in AD.
Figure 2. Examples of GSIs.

Begacestat, BMS-708163 and ELN-475516 have been reported to be Notch-Sparing GSIs.
Herein, we will review the development status of both GSIs and GSMs. For GSIs we will largely focus on the efforts to i) repurpose these compounds for indications other than AD ii) design substrate selective GSIs. For GSMs we will discuss the current development status and open questions regarding potential utility in AD.
2. GSIs
In the mid to late 1990's, cell-based drug screens conducted by multiple groups searching for inhibitors of Aβ production identified a number of compounds that dramatically inhibited Aβ secretion and increased levels of APP carboxyl terminal fragments (CTFs) produced by prior α- or β-secretase catalyzed ectodomain shedding [22-29]. At the time the first compounds with these effects on APP processing were identified, the protease targeted was unknown, but the cleavage activity was referred to as γ-secretase. Thus, compounds with this profile were named GSIs. Because γ-secretase cleaved APP within its transmembrane domain and generated multiple Aβ peptides, there were many hypotheses regarding the nature of the activity and the proteases responsible [30-31]. Furthermore, at that time, there was general resistance to the concept that a protease could cleave peptide bonds normally present within the transmembrane domain (TMD) of a protein, fueling further speculation regarding the nature of the protease responsible. Several inhibitor studies also demonstrated that γ-secretase possessed multiple pharmacologically dissociable cleavage activities indicating that it may be more than one protease [32-33]. However, genetic, GSI binding, biochemical and mutational analyses soon demonstrated that γ-secretase was a multi-protein complex with the PSEN1 or PSEN2 acting as the catalytic core, and three accessory proteins, APH1, PEN2, and Nicastrin, needed for complex assembly and stability in cells [1-3, 34]. Although it remains formally possible that small-molecules that inhibit γ-secretase cleavage could bind one of the other subunits, GSI binding studies suggest that the target of most GSIs is PSEN1 and 2.
PSEN1 and 2 are now known to be part of a larger family of intramembrane cleaving aspartyl proteases which include five human homologs referred to as signal peptide peptidases (SPP (HM123), SPPL3, SPPL2a,b,c) [35-37]. SPPs differ from PSENs in that they cleave the transmembrane domain of type 2 as opposed to type 1 membrane proteins, and at least for SPP(HM13), the apparent lack of requirement for co-factors for activity [35, 38-39]. The differential cleavage specificity appears to be determined by the opposite orientation of the two catalytic aspartate residues between SPPs and PSEN family members. Of note, the recent crystal structure of an SPP from archaeon Methanoculleus marisnigri JR1 was established [40]. This structure showed that the catalytic aspartates residues that reside within opposing transmembrane domains are in close proximity to each other and the lipid membrane surface.
Some, but not all, GSIs inhibit signal peptide peptidases as well, though this has not been systemically studied [36, 41]. When considering biological activities of various GSIs this is an important and understudied caveat that could influence both biological response as well as potential toxicities. Indeed, it has been reported that SPP, and not PSEN1 or PSEN2, is the major binding target of a GSI in some cells, which likely reflects the fact that in most cells SPP is much more abundant then PSEN/γ-secretase [42]. Although largely outside the scope of this current review, SPP family members have been proposed to be potential therapeutic targets in malaria, various viral infections, and more recently in B-cell related disease [43-50]. Thus, GSIs which target SPPs can be useful probes to examine the biological consequences of SPP inhibition.
As noted above GSIs were initially developed as “Aβ inhibitors”. As accumulation of Aβ aggregates in the brain is proposed to trigger AD, and Aβ aggregate formation is a concentration dependent phenomena, the rationale for GSI development was strong [51]. However, there are concerns that Aβ inhibitors have to be given for a prolonged period of time, may work only as prophylactic therapies or in the protracted, asymptomatic, prodromal phase where Aβ accumulates, and will be increasingly ineffective as Aβ loads increase in the brain [52]. Indeed, this assertion is supported by several in vivo preclinical studies that show that targeting Aβ with a GSI or other modality is much more effective in a prevention paradigm [21, 53-54]. To date, the clinical experience in humans with AD is that long-term GSI treatment designed to produce moderate levels of inhibition of γ-secretase is associated with unacceptable side-effects and lack of clinical efficacy [55-58]. Thus, unless there is an unanticipated breakthrough, γ-secretase inhibition is not likely to be a viable chronic treatment strategy for AD. Furthermore because of these safety issues, it is almost certain that GSIs will not be suitable for testing in asymptomatic individuals at risk for AD.
Outside of the CNS, therapeutic inhibition of γ-secretase has been most often associated with reduced Notch 1 signaling; GSIs are often thought of in these settings as “Notch 1 inhibitors” [59-62]. γ-Secretase has now been proposed to be a therapeutic target in various cancers [59, 61, 63-81], immunologic disorders including graft versus host disease [82-83], vasculitis [84], macular degeneration [85], diabetic nephropathy [86-87], ischemic reperfusion injury in the kidney [88], ischemic stroke [89], traumatic brain injury, [90], hearing loss [91] and fibrosis [92]. It is also likely that additional disease indications may emerge. Currently, a main focus of the repurposing of γ-secretase inhibitors (GSIs) has been in cancer with multiple human trials underway (Table 1). Both GSI monotherapy and combination therapies with other agents are being explored.
Table 1. GSIs in trial for Cancer.
| Indication | GSI | Single Agent | Combination |
|---|---|---|---|
| Breast Cancer | RO4929097[194-195] MK0752[123] | Yes | Yes |
| Glioma/GBM | RO4929097 | Yes | Yes |
| Pancreatic Cancer | RO4929097 | Yes | Yes |
| Lung Cancer | RO4929097 | Yes | Yes |
| Melanoma | RO4929097 | Yes | Yes |
| Leukemia/Lymphoma | RO4929097 MK0752 PF-03084014 [64, 81] | Yes | Yes |
| Ovarian Cancer | RO4929097 | Yes | Yes |
| Kidney Cancer | RO4929097 | No | Yes |
| Colorectal Cancer | RO4929097 | No | Yes |
| Sarcomas | RO4929097 | No | Yes |
| Endometrial Cancer | RO4929097 | No | Yes |
| Prostate Cancer | RO4929097 | No | Yes |
Development of GSIs for most cancers as well as other indications has been primarily based on the premise that GSIs act by inhibiting the cleavage of Notch 1, as inhibition of γ-secretase cleavage blocks Notch 1 signaling [59, 78, 93-94]. Although studies in T-cell lymphoblastic leukemia (T-ALL) unequivocally demonstrate that Notch 1 plays a central role in T-ALL tumor development [79]; the role of Notch 1 signaling has not been as critically examined in most solid tumors. Notch 1 signaling has a normal function in maintenance, development and cell fate. It also has been shown to promote cell survival, angiogenesis and treatment resistance in numerous cancers, both through direct Notch 1 signaling and crosstalk with other key oncogenic pathways [94-99]. However, a comprehensive examination of the mechanism of action of GSIs in cancer and other indications, as well as their effects on angiogenesis and immunity in immunocompetent models has not been conducted [59, 100]. Thus, if GSIs show efficacy for cancer other indications, it may because they synergistically alter multiple signaling pathways. It is also important to consider the findings that GSI based inhibition of Notch 1 and perhaps other substrates of γ-secretase, can actually promote oncogenic transformation in certain tissues such as the skin [61]. Thus, even if acute toxicities can be managed, there are concerns that GSI like many anticancer therapies could promote other cancers.
With rare exceptions, the biological role of γ-secretase cleavage of substrates other than Notch 1 has been ignored during proof of concept preclinical repurposing studies. For example, in the GSI AD trial, many individuals noted changes in hair color, apparently due to inhibition of tyrosinase, another γ-secretase substrate [101]. In a few studies other Notch paralogs and VEGFR1 have been considered to be targets [70, 102-103]. Given that tools are not readily available to perform facile, detailed studies on the impact of γ-secretase cleavage on γ-secretase substrates other than APP and Notch 1; it will be important to develop these tools to better understand the biological consequences of GSI based therapies [4]. Indeed, the biology of γ-secretase is complex and our understanding of it suffers from the lamplight effect. As a field we have largely focused only on what we can easily see -cleavage of Notch 1 and APP [104].
Sub-unit composition and subcellular localization of γ-secretase within the target cell may influence both activity on a given substrate and response to a given GSI. Zhao and colleagues have shown that sulfonamide based GSIs selectively inhibit PSEN1 over PSEN2, whereas the GSIs DAPT and L685458 showed minimal selectivity [105]. Similarly, recent data suggest that the GSI MRK-560 preferentially targets PSEN1 over PSEN2 and that this selectivity, at least in mice, increases the tolerability of this GSI [106]. De Strooper and colleagues also show that heterogeneity with respect to the Aph1 subunit is important with respect to viability and overall phenotype of mice, indicating that selective targeting of Aph1b γ-secretase complexes may be less toxic [107]; however, it is not clear whether selective targeting of Aph1b γ-secretase complexes is feasible. Other studies also provide some evidence that various γ-secretase complexes may have differential sensitivity to GSI and different substrate preferences [108-110]. In many cases it is not clear if substrate preference, differential spatiotemporal expression patterns of the various γ-secretase subunits, or a combination of these factors, contributes to the phenotype. Outside of AD, the fact that γ-secretase is a heterogeneous activity has largely been ignored.
3. Avoiding GSI toxicity, Substrate Selective GSIs and other strategies
Given the toxicities associated with inhibition of Notch 1, especially those associated with altered proliferation and maturation of gut epithelium, there has been considerable effort to develop APP selective inhibitors or at least inhibitors that would reduce in vivo toxicity sufficiently to enable significant reduction of Aβ production in the brain [111]. Unfortunately, so called “Notch-sparing” APP selective GSIs that can be shown to preferentially inhibit γ-secretase APP relative to Notch 1 in preclinical studies have not shown reduced toxicity nor increased Aβ lowering in humans [112-119]. Furthermore, recent studies suggest that one potential issue is that substrate selectivity has not always been assessed in matched assays, and when GSIs inhibitory profiles are examined in matched assays substrate selectivity is absent [117, 120]. Thus, it is possible that the apparent substrate selectivity is really attributable to different in vitro assays. Discrepant IC50 values may arise when using reporter assays for γ-secretase activity that rely on translocation of endogenous or transcription factor tagged intracellular domains of a given substrate. In these assays, γ-secretase cleavage liberates the intracellular domain and enables it to translocate to the nucleus where it activates a reporter gene [121-122]. It is not clear that these reporter assays accurately reflect effects on cleavage, as there are often large differnces between inhibitor IC50s in the reporter assays versus direct cleavage assays. One thought is that the overexpression of and cleavage of substrate may over-saturate the transcriptional reporter; thus; significant inhibition of cleavage can be observed before the reporter activity is decreased.
Based on the collective experience to date, it is likely that a much more comprehensive assessment of GSI selectivity for inhibition of multiple substrates, as well as biomarkers to track inhibition of non-APP substrates in vivo will be needed to support future efforts to develop substrate selective GSIs. Similarly, development of GSIs designed to target a specific γ-secretase complex will be facilitated by development of such biomarkers that can facilely track inhibition of multiple substrates. The most likely biomarkers candidates are the Aβ-like peptides produced by γ-secretase cleavage, as these are likely to be present in body fluids and detectable using sandwich ELISAs or mass-spectrometry. Alternatively one might collect peripheral blood and examine the accumulation of substrate derived carboxyl terminal fragments in peripheral leukocytes, but this will likely limit the number of substrates that could be assayed. Until such assays are available we will not know whether we can predict with any accuracy the differential in vivo activity of a given GSI using current preclinical models.
With the repurposing of GSIs for cancer, one of the key findings from the early human trials is that subacute dosing with GSIs is reasonably well-tolerated especially when dosing regimens are altered so that dosing is not continuous but intermittent [123]. Alternatively, administration of glucocortocoids with GSI dramatically attenuated gastrointestinal toxicity [124]. In this regard, development of additional tools that better enable assessment of GSI activity on multiple substrates may help to optimize individual dosing so that maximal clinical benefit is achieved while minimizing side-effects. Of course in cancer as opposed to AD, there is generally a willingness to accept some level of toxicity if there are any signs of efficacy.
One important question that remains is whether all current GSIs are biologically equivalent. Though many GSIs currently being used for cancer trials are considered “pan-GSI inhibitors” this labeling may be a misnomer. GSI inhibitory activity is often only established for Aβ and Notch 1 [16]. The net action of GSIs may be influenced by multiple factors within a target cell [9, 107, 109, 125-126]. These factors not only include the variable subunit composition of the γ-secretase complexes but also a) the expression of the substrate in the target cell, b) the location of the substrate, c) sheddase expression, and d) activation of the sheddase. Thus, GSI action could be unexpectedly influenced by any of these factors. Clearly, given the investment in repurposing GSIs, additional studies directly comparing biological actions of various GSI used in clinical trials in various model systems are warranted.
γ-Secretase cleavage is remarkably promiscuous, but somehow regulated [9]. To our knowledge there is no type 1 membrane protein which has been shown to be processed by a sheddase in which the membrane stub is not subsequently processed by γ-secretase. Furthermore, mutational studies and comparison of the TMD sequences cut by γ-secretase reveal that there is little sequence specificity to γ-cleavage [32, 127-130]. Clearly, ligand binding induced ectodomain shedding is one regulatory step in γ-cleavage, but for many constitutively shed and processed proteins there must be other ways of regulating signaling [131]. Previously, we and others have shown that γ-secretase cleavage of APP was located in cholesterol rich buoyant membranes (lipid rafts) and that cholesterol depletion blocked cleavage [12, 132-134]. Subsequently, a number of labs have confirmed these findings for APP, and recently this was extended to show that γ-secretase interacts with tetraspanins and this interaction facilitates γ-secretase localization in raft domains [135-136]. Another study suggested that Notch 1 γ-secretase cleavage occurs on the cell surface whereas APP occurs in intracellular compartments [131]. These data indicated that co-localization of ectodomain-shed substrate CTF and γ-secretase, helped to regulate cleavage. In contrast during development, γ-secretase appears to be active in both raft and non-raft membranes. How the potential altered localization of γ-secretase influences activity and response to a GSI is not known, but again an area worthy of further study.
4. GSMs
γ-Secretase cleavage of APP generates a number of Aβ peptides [32, 137]. In most cells Aβ1-37, 38, 39, and 42 are produced at low levels (typically each represents 5-20% of total Aβ detected) and the major species generated is Aβ1-40 (typically over 50% of total Aβ). Other Aβ peptides can also be variably detected at low levels including Aβ1-34, 1-36, 1-41 and 1-43. Shifts in the relative production of these various Aβ peptides towards Aβ1-42, is tightly associated with risk for AD [138-139]. Mutations in APP and PSEN1/2 that elevate the relative level of Aβ42 by even as little as 30% deterministically cause early onset AD [140], and it now appears that the deposition process likely begins 20 years before the onset of dementia in these individuals [141]. Seminal biochemical studies show that Aβ1-42 aggregates into amyloid fibrils and other assemblies much more readily than Aβ1-40 [142-143], and transgenic modeling studies show that AD-associated APP and PSEN mutations increases Aβ42 levels and accelerate Aβ deposition [144-145]. In addition other studies using various fusion protein strategies to express Aβ1-42 and Aβ1-40 in the absence of APP overexpression show that Aβ42 is required to drive Aβ deposition, and that Aβ1-40 may actually inhibit Aβ deposition [146-149].
Some early studies of peptidic GSIs showed that although they reduced total Aβ levels and increased APP CTF; they also shifted the profile of Aβ species produced, in some cases increasing the absolute level of longer Aβ1-41,42, and 43 [30, 32-33]. As discussed earlier these data fueled some speculation that multiple proteases contributed to the generation of the various Aβ peptides. Subsequently, we identified a subset of non-steroidal anti-inflammatory agents (NSAIDs) such as ibuprofen, sulindac and indomethacin, as prototypic agents capable of lowering Aβ42 selectively in vitro and in vivo by targeting the γ-secretase complex [150-152]. The classic signature of these first generation GSMs differed from previous GSIs in that they did not alter total Aβ production, or increase APP CTFs and no alterations in the generation of several other γ-secretase substrates, but instead decreased Aβ1-42 levels and increased Aβ1-38 [152]. These data suggested that it may be possible to use GSMs as therapeutic agents for AD as they would selectively target the longer more pathogenic forms of Aβ. In addition to these classic GSMs, other compounds referred to as inverse GSMs (iGSMs) were also identified [153-154]. These compounds were often structurally related to the GSMs but typically lacked an acidic group, and increased, rather than lowering, Aβ1-42. In some, but not all cases, these compounds also decreased levels of shorter Aβ peptides including (Aβ1-37, 38, 39). Since the initial identification of NSAID-based GSMs there has been a major effort to improve potency, pharmacokinetic properties and find new non-acidic classes of GSMs. This has led to the identification of a number of acidic GMSs with dramatic increases in potency and brain penetrance, and non-acidic GSMs based around a common scaffold that are also much more potent than the first generation NSAID-based GSMs (Figure 3) [155-172]. More recently triterpenoid natural product derived GSMs have been identified [173-174]. Overall there are subtle differences between these GSMs that may be therapeutically important. While the more potent acidic GSMs show the classic GSM signature of lowering Aβ1-42 and increasing Aβ1-38 [175], nonacidic GSMs increase Aβ1-37 and 1-38 and lower both Aβ1-40 and 1-42 [171]. In contrast, the triterpenoid GSM appears to be even more distinct lowering both Aβ1-38 and 1-42 [173-174]. These differential properties and comparison to GSIs are described in Table 2.
Figure 3. Examples of GSMs.
Indomethacin is an NSAID GSM, GSM-1 is an example of a potent 2nd generation acidic GSM and E2012 is an example of a non-acidic GSM.
Table 2.
Comparison of Effects of GSIs, GSMs and iGSMs
| Total Aβ | Aβ1-37 | Aβ1-38 | Aβ1-40 | Aβ1-42 | APP CTF | Notch | EC50↓ Aβ42A | Binding Target | |
|---|---|---|---|---|---|---|---|---|---|
| GSI | ↓ | ↓ | ↓ | ↓ | ↓ | ↑ | ↓ | ∼500 pM | PSEN1/2 |
| NSAID-GSM | = | = | ↑ | = | ↓ | = | = | ∼10 μM | APP/Substrate |
| 2nd Generation Acidic-GSMs | = | = | ↑ | = | ↓ | = | = | ∼50 nM | PSEN |
| Non-Acidic | = | ↑ | ↑ | = | ↓ | = | = | ∼50 nM | PSEN/PEN2 |
| Triterpenoid | = | = | ↓ | = | ↓ | = | = | ∼50 nM | ? |
| iGSM | = | ↓or = | ↓or = | = | ↑ | = | = | ∼10 μM | APP/Substrate |
These EC50s are approximations of the most potent compounds reported.
Until recently the mechanism of how GSMs shifted γ-secretase cleavage was poorly understood. Today, building on the sequential cleavage model developed by Ihara and colleagues [14], there is evidence that GSMs act as processivity enhancers, and iGSMs as inhibitors of processivity [120]. At least for APP and Notch, neither acidic,non-acidic GSMs nor iGSMS significantly alter the initial γ-secretase cleavage site [150, 152, 176-177], but they do appear to primarily alter the subsequent number of processivity cleavage events. For classic GSMs this means that they increase cleavage of Aβ1-42 to Aβ1-38; for many iGSMs, they appear to do the opposite. Although not directly established non-acidic GSMs appear would be proposed to increase processivity of Aβ1-40 and Aβ1-42 to Aβ1-37 and Aβ1-38. The novel triterpenoid GSMs recently described by Satori Pharmaceuticals, are somewhat unusual as they decrease both Aβ1-38 and Aβ1-42 [173-174]. How this unusual type of modulation occurs is unclear, but could be accounted for either by effect on the substrate product line that is determined by the initial γ-secretase cleavage (i.e. a shift to cleavage that initially generates Aβ1-49) or decreased processivity along the Aβ1-42 product line. If the latter is the case, then an even longer Aβ peptide should accumulate.
Notably, for the more well studied GSMs there is clear evidence that these compounds do not alter Notch 1 processing to any great extent[129, 150, 152, 178]. They preserve Notch 1 signaling and thus are thought to be inherently safe. Although it is almost certain that GSMs will to varying extents modulate cleavage of some other γ-secretase substrates, there is no evidence to date that this will have significant biological impact. GSMs are not likely to alter signaling events mediated by the initial γ-cleavage and subtle shifts in the length of the short Aβ like fragments produced by subsequent γ-cleavages are unlikely to have significant liabilities.
There remains some controversy over the binding site of GSMs. Low potency NSAID-based GSMs have been shown to bind substrate [171, 179-180], whereas more potent acidic GSMs and non-acidic GSMs have been shown to bind PSEN1 or PEN-2 [171, 181-183]. The target of the triterpenoid natural product has not been established. Notably, there is evidence that the effect of GSMs is substrate selective and that sequences within substrate dramatically influence processivity of γ-secretase [129-130]. Collectively these binding studies and substrate selective effects of GSMs suggest that there may tripartite interactions between GSM, γ-secretase and substrate that subtly alter processivity perhaps by altering residence time of the substrate within the active site of γ-secretase. Another area of some controversy is whether APP and PS1 mutations influence the potency of GSMs. Though potency of 1st generation GSM were reported to be altered by AD-linked PS1 and APP mutations, other studies with more potent GSMs showed little effect on potency [175, 184].
5. Clinical Development of GSMs
A Phase III study of the NSIAD based GSM Tarenflurbil (R-flurbiprofen) in patients with mild AD showed no benefit on cognitive or functional outcomes[185]. Tarenflurbil is a weak GSM with low CNS penetrance limiting general inferences based on this trial with respect to clinical efficacy of more potent, CNS penetrant GSMs [11, 151, 160, 186]. Many companies and academic groups have ongoing GSM development programs and several more potent GSM have entered early phase clinical trials. TheGSM, CHF5074, based on R-flurbiprofen developed by Chiesi Pharmaceuticals has advanced the furthest in human testing completing a phase II trial [157]. Recent data suggest that while this compound may possess GSM activity it may have a rich pharmacology and may have additional mechanism(s) of action besides altering Aβ42 [187-189]. One clear challenge in the development of potent GSMs that have been recently reviewed [160], is the balance between liophillicity and potency. Although potency and brain penetration have been dramatically improved in 2nd and 3rd generation GSMs, this has been associated with increases in lipophillicty of the compounds. It has been proposed that this lipophillicity as well as potential glucuronidation of acidic GSMs contributes to what are thought to be off-target, primarily liver toxicities [160].
As with the GSIs, there are again major concerns that GSM therapy in patients with symptomatic AD is almost certain to fail, unless the compound has additional mechanism of action not linked to Aβ that prove to beneficial [52]. Although Aβ1-42 or other longer Aβ peptides are critical for initiating Aβ deposition, shorter Aβ accumulate in the AD brain [190]. Thus, shifting Aβ cleavage after nucleation events have occurred is not likely to dramatically alter Aβ deposition kinetics. Thus, a major challenge is whether a sufficiently safe GSM can be developed that can potentially be used as a prophylactic agent in asymptomatic individuals. A final issue of clinical relevance relates to the biology of the shorter Aβ peptides. There has been little systematic investigation of these peptides. Although one report suggested that Aβ1-38 may behave in vitro like Aβ1-42, this important finding has not been reproduced [191]. Further study of these short peptides may reveal unique properties that might help to guide development of GSMs. For example Aβ1-40 appears to act an inhibitor of Aβ1-42 nucleation and aggregation in vivo[192-193], but whether Aβ1-37 or 1-38 inhibit aggregation in vivo is unknown. Given that non-acidic GSMs decrease Aβ1-40 whereas acidic GSMs do not, the distinct action of these two classes of GSMs could have major impact on efficacy.
6. Conclusions
From a biological perspective γ-secretase is both a fascinating and complex enzyme that is increasingly being scrutinized as a therapeutic target for conditions other than AD. There are many gaps in our knowledge that need to be answered to optimally move forward with development of GSIs for cancer and other indications and the continued development of GSMs for AD. Future studies addressing the contribution of other substrates to effects of GSIs and even GSMs, and efforts to determine if all GSI are biologic equivalents certainly will be key steps in these development efforts.
Highlights.
γ-Secretase is a therapeutic target for a number of diseases.
γ-Secretase is a highly druggable therapeutic target.
Unique properties of γ-secretase inhibitors and modulators may influence efficacy.
Acknowledgments
This work was supported by P01 AG20206 (EHK, TEG) and P01 AG025531 (BAO, TEG, LM).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.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. 2003;422:438–441. doi: 10.1038/nature01506. [DOI] [PubMed] [Google Scholar]
- 2.De Strooper B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma- Secretase complex. Neuron. 2003;38:9–12. doi: 10.1016/s0896-6273(03)00205-8. [DOI] [PubMed] [Google Scholar]
- 3.Wolfe MS, Kopan R. Intramembrane proteolysis: theme and variations. Science. 2004;305:1119–1123. doi: 10.1126/science.1096187. [DOI] [PubMed] [Google Scholar]
- 4.Haapasalo A, Kovacs DM. The Many Substrates of Presenilin/gamma-Secretase. J Alzheimers Dis. 2011 [Google Scholar]
- 5.Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, McKay DM, Tintner R, Frangione B, et al. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science. 1992;258:126–129. doi: 10.1126/science.1439760. [DOI] [PubMed] [Google Scholar]
- 6.Haass C, Schlossmacher MG, Hung AY, Vigo-Pelfrey C, Mellon A, Ostaszewski BL, Lieberburg I, Koo EH, Schenk D, Teplow DB, et al. Amyloid beta-peptide is produced by cultured cells during normal metabolism [see comments] Nature. 1992;359:322–325. doi: 10.1038/359322a0. [DOI] [PubMed] [Google Scholar]
- 7.Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain [see comments] Nature. 1998;393:382–386. doi: 10.1038/30756. [DOI] [PubMed] [Google Scholar]
- 8.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. 1999;398:518–522. doi: 10.1038/19083. [DOI] [PubMed] [Google Scholar]
- 9.Kopan R, Ilagan MX. Gamma-secretase: proteasome of the membrane? Nat Rev Mol Cell Biol. 2004;5:499–504. doi: 10.1038/nrm1406. [DOI] [PubMed] [Google Scholar]
- 10.Golde TE, Eckman CB. Physiologic and pathologic events mediated by intramembranous and juxtamembranous proteolysis. Sci STKE. 2003;2003 doi: 10.1126/stke.2003.172.re4. [DOI] [PubMed] [Google Scholar]
- 11.De Strooper B, Vassar R, Golde T. The secretases: enzymes with therapeutic potential in Alzheimer disease. Nat Rev Neurol. 2010;6:99–107. doi: 10.1038/nrneurol.2009.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Vetrivel KS, Cheng H, Kim SH, Chen Y, Barnes NY, Parent AT, Sisodia SS, Thinakaran G. Spatial segregation of gamma-secretase and substrates in distinct membrane domains. J Biol Chem. 2005;280:25892–25900. doi: 10.1074/jbc.M503570200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gu Y, Misonou H, Sato T, Dohmae N, Takio K, Ihara Y. Distinct intramembrane cleavage of the beta-amyloid precursor protein family resembling gamma-secretase-like cleavage of Notch. J Biol Chem. 2001;276:35235–35238. doi: 10.1074/jbc.C100357200. [DOI] [PubMed] [Google Scholar]
- 14.Takami M, Nagashima Y, Sano Y, Ishihara S, Morishima-Kawashima M, Funamoto S, Ihara Y. gamma-Secretase: successive tripeptide and tetrapeptide release from the transmembrane domain of beta-carboxyl terminal fragment. J Neurosci. 2009;29:13042–13052. doi: 10.1523/JNEUROSCI.2362-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pinnix I, Musunuru U, Tun H, Sridharan A, Golde T, Eckman C, Ziani-Cherif C, Onstead L, Sambamurti K. A novel gamma -secretase assay based on detection of the putative C- terminal fragment-gamma of amyloid beta protein precursor. J Biol Chem. 2001;276:481–487. doi: 10.1074/jbc.M005968200. [DOI] [PubMed] [Google Scholar]
- 16.Golde TE, Petrucelli L, Lewis J. Targeting Abeta and tau in Alzheimer's disease, an early interim report. Exp Neurol. 2010;223:252–266. doi: 10.1016/j.expneurol.2009.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wolfe MS. Inhibition and modulation of gamma-secretase for Alzheimer's disease. Neurotherapeutics. 2008;5:391–398. doi: 10.1016/j.nurt.2008.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bateman RJ, Siemers ER, Mawuenyega KG, Wen G, Browning KR, Sigurdson WC, Yarasheski KE, Friedrich SW, Demattos RB, May PC, Paul SM, Holtzman DM. A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann Neurol. 2009;66:48–54. doi: 10.1002/ana.21623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Siemers ER, Dean RA, Friedrich S, Ferguson-Sells L, Gonzales C, Farlow MR, May PC. Safety, tolerability, and effects on plasma and cerebrospinal fluid amyloid-beta after inhibition of gamma-secretase. Clin Neuropharmacol. 2007;30:317–325. doi: 10.1097/WNF.0b013e31805b7660. [DOI] [PubMed] [Google Scholar]
- 20.Dovey HF, John V, Anderson JP, Chen LZ, de P, Andrieu Saint, Fang LY, Freedman SB, Folmer B, Goldbach E, Holsztynska EJ, Hu KLK, Johnson-Wood L, Kennedy SL, Kholodenko D, Knops JE, Latimer LH, Lee M, Liao Z, Lieberburg IM, Motter RN, Mutter LC, Nietz J, Quinn KP, Sacchi KL, Seubert PA, Shopp GM, Thorsett ED, Tung JS, Wu J, Yang S, Yin CT, Schenk DB, May PC, Altstiel LD, Bender MH, Boggs LN, Britton TC, Clemens JC, Czilli DL, Dieckman-McGinty DK, Droste JJ, Fuson KS, Gitter BD, Hyslop PA, Johnstone EM, Li WY, Little SP, Mabry TE, Miller FD, Audia JE. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J Neurochem. 2001;76:173–181. doi: 10.1046/j.1471-4159.2001.00012.x. [DOI] [PubMed] [Google Scholar]
- 21.Abramowski D, Wiederhold KH, Furrer U, Jaton AL, Neuenschwander A, Runser MJ, Danner S, Reichwald J, Ammaturo D, Staab D, Stoeckli M, Rueeger H, Neumann U, Staufenbiel M. Dynamics of Abeta turnover and deposition in different beta-amyloid precursor protein transgenic mouse models following gamma-secretase inhibition. J Pharmacol Exp Ther. 2008;327:411–424. doi: 10.1124/jpet.108.140327. [DOI] [PubMed] [Google Scholar]
- 22.Seiffert D, Bradley JD, Rominger CM, Rominger DH, Yang F, Meredith JE, Jr, Wang Q, Roach AH, Thompson LA, Spitz SM, Higaki JN, Prakash SR, Combs AP, Copeland RA, Arneric SP, Hartig PR, Robertson DW, Cordell B, Stern AM, Olson RE, Zaczek R. Presenilin-1 and -2 are molecular targets for gamma -secretase inhibitors [In Process Citation] J Biol Chem. 2000;275:34086–34091. doi: 10.1074/jbc.M005430200. [DOI] [PubMed] [Google Scholar]
- 23.Higaki JN, Chakravarty S, Bryant CM, Cowart LR, Harden P, Scardina JM, Mavunkel B, Luedtke GR, Cordell B. A combinatorial approach to the identification of dipeptide aldehyde inhibitors of beta-amyloid production. Journal of Medicinal Chemistry. 1999;42:3889–3898. doi: 10.1021/jm990009f. [DOI] [PubMed] [Google Scholar]
- 24.Higaki J, Murphy GM, Cordell B. Inhibition of Beta-Amyloid Formation By Haloperidol - a Possible Mechanism For Reduced Frequency of Alzheimers Disease Pathology in Schizophrenia. Journal of Neurochemistry. 1997;68:333–336. doi: 10.1046/j.1471-4159.1997.68010333.x. [DOI] [PubMed] [Google Scholar]
- 25.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 [In Process Citation] Nature. 2000;405:689–694. doi: 10.1038/35015085. [DOI] [PubMed] [Google Scholar]
- 26.Li YM, Lai MT, Xu M, Huang Q, DiMuzio-Mower J, Sardana MK, Shi XP, Yin KC, Shafer JA, Gardell SJ. Presenilin 1 is linked with gamma-secretase activity in the detergent solubilized state. Proc Natl Acad Sci U S A. 2000;97:6138–6143. doi: 10.1073/pnas.110126897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wolfe MS, Citron M, Diehl TS, Xia W, Donkor IO, Selkoe DJ. A substrate-based difluoro ketone selectively inhibits Alzheimer's gamma-secretase activity. Journal of Medicinal Chemistry. 1998;41:6–9. doi: 10.1021/jm970621b. [DOI] [PubMed] [Google Scholar]
- 28.Prasad CV, Vig S, Smith DW, Gao Q, Polson CT, Corsa JA, Guss VL, Loo A, Barten DM, Zheng M, Felsenstein KM, Roberts SB. 2,3-Benzodiazepin-1,4-diones as peptidomimetic inhibitors of gamma-secretase. Bioorg Med Chem Lett. 2004;14:3535–3538. doi: 10.1016/j.bmcl.2004.04.056. [DOI] [PubMed] [Google Scholar]
- 29.Barten DM, Guss VL, Corsa JA, Loo A, Hansel SB, Zheng M, Munoz B, Srinivasan K, Wang B, Robertson BJ, Polson CT, Wang J, Roberts SB, Hendrick JP, Anderson JJ, Loy JK, Denton R, Verdoorn TA, Smith DW, Felsenstein KM. Dynamics of {beta}-amyloid reductions in brain, cerebrospinal fluid, and plasma of {beta}-amyloid precursor protein transgenic mice treated with a {gamma}-secretase inhibitor. J Pharmacol Exp Ther. 2005;312:635–643. doi: 10.1124/jpet.104.075408. [DOI] [PubMed] [Google Scholar]
- 30.Murphy MP, Uljon SN, Fraser PE, Fauq A, Lookingbill HA, Findlay KA, Smith TE, Lewis PA, McLendon DC, Wang R, Golde TE. Presenilin 1 regulates pharmacologically distinct gamma -secretase activitie Implications for the role of presenilin in gamma -secretase cleavage. J Biol Chem. 2000;275:26277–26284. doi: 10.1074/jbc.M002812200. [DOI] [PubMed] [Google Scholar]
- 31.Murphy MP, Wang R, Fraser PE, Fauq A, Golde TE. An empirical model of gamma-secretase activity. Ann N Y Acad Sci. 2000;920:233–240. doi: 10.1111/j.1749-6632.2000.tb06928.x. [DOI] [PubMed] [Google Scholar]
- 32.Murphy MP, Hickman LJ, Eckman CB, Uljon SN, Wang R, Golde TE. gamma-Secretase, evidence for multiple proteolytic activities and influence of membrane positioning of substrate on generation of amyloid beta peptides of varying length. Biol Chem. 1999;274:11914–11923. doi: 10.1074/jbc.274.17.11914. [DOI] [PubMed] [Google Scholar]
- 33.Citron M, Diehl TS, Gordon G, Biere AL, Seubert P, Selkoe DJ. Evidence that the 42- and 40-amino acid forms of amyloid beta protein are generated from the beta-amyloid precursor protein by different protease activities. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:13170–13175. doi: 10.1073/pnas.93.23.13170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wolfe MS, Xia WM, Ostaszewski BL, Diehl TS, Kimberly WT, Selkoe DJ. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature. 1999;398:513–517. doi: 10.1038/19077. [DOI] [PubMed] [Google Scholar]
- 35.Weihofen A, Binns K, Lemberg MK, Ashman K, Martoglio B. Identification of signal peptide peptidase, a presenilin-type aspartic protease. Science. 2002;296:2215–2218. doi: 10.1126/science.1070925. [DOI] [PubMed] [Google Scholar]
- 36.Martoglio B, Golde TE. Intramembrane-cleaving aspartic proteases and disease: presenilins, signal peptide peptidase and their homologs. Hum Mol Genet. 2003;12(2):R201–206. doi: 10.1093/hmg/ddg303. [DOI] [PubMed] [Google Scholar]
- 37.Ponting CP, Hutton M, Nyborg A, Baker M, Jansen K, Golde TE. Identification of a novel family of presenilin homologues. Hum Mol Genet. 2002;11:1037–1044. doi: 10.1093/hmg/11.9.1037. [DOI] [PubMed] [Google Scholar]
- 38.Nyborg AC, Jansen K, Ladd TB, Fauq A, Golde TE. A signal peptide peptidase (SPP) reporter activity assay based on the cleavage of type II membrane protein substrates provides further evidence for an inverted orientation of the SPP active site relative to presenilin. J Biol Chem. 2004;279:43148–43156. doi: 10.1074/jbc.M405879200. [DOI] [PubMed] [Google Scholar]
- 39.Nyborg AC, Kornilova AY, Jansen K, Ladd TB, Wolfe MS, Golde TE. Signal peptide peptidase forms a homodimer that is labeled by an active site-directed gamma-secretase inhibitor. J Biol Chem. 2004;279:15153–15160. doi: 10.1074/jbc.M309305200. [DOI] [PubMed] [Google Scholar]
- 40.Li X, Dang S, Yan C, Gong X, Wang J, Shi Y. Structure of a presenilin family intramembrane aspartate protease. Nature. 2013;493:56–61. doi: 10.1038/nature11801. [DOI] [PubMed] [Google Scholar]
- 41.Fluhrer R, Grammer G, Israel L, Condron MM, Haffner C, Friedmann E, Bohland C, Imhof A, Martoglio B, Teplow DB, Haass C. A gamma-secretase-like intramembrane cleavage of TNFalpha by the GxGD aspartyl protease SPPL2b. Nat Cell Biol. 2006;8:894–896. doi: 10.1038/ncb1450. [DOI] [PubMed] [Google Scholar]
- 42.Iben LG, Olson RE, Balanda LA, Jayachandra S, Robertson BJ, Hay V, Corradi J, Prasad CV, Zaczek R, Albright CF, Toyn JH. Signal peptide peptidase and gamma-secretase share equivalent inhibitor binding pharmacology. J Biol Chem. 2007;282:36829–36836. doi: 10.1074/jbc.M707002200. [DOI] [PubMed] [Google Scholar]
- 43.Harbut MB, Patel BA, Yeung BK, McNamara CW, Bright AT, Ballard J, Supek F, Golde TE, Winzeler EA, Diagana TT, Greenbaum DC. Targeting the ERAD pathway via inhibition of signal peptide peptidase for antiparasitic therapeutic design. Proc Natl Acad Sci U S A. 2012;109:21486–21491. doi: 10.1073/pnas.1216016110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Parvanova I, Epiphanio S, Fauq A, Golde TE, Prudencio M, Mota MM. A small molecule inhibitor of signal peptide peptidase inhibits Plasmodium development in the liver and decreases malaria severity. PLoS One. 2009;4:e5078. doi: 10.1371/journal.pone.0005078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Targett-Adams P, Schaller T, Hope G, Lanford RE, Lemon SM, Martin A, McLauchlan J. Signal peptide peptidase cleavage of GB virus B core protein is required for productive infection in vivo. J Biol Chem. 2006;281:29221–29227. doi: 10.1074/jbc.M605373200. [DOI] [PubMed] [Google Scholar]
- 46.Bronckers AL, Guneli N, Lullmann-Rauch R, Schneppenheim J, Moraru AP, Himmerkus N, Bervoets TJ, Fluhrer R, Everts V, Saftig P, Schroder B. The intramembrane protease SPPL2A is critical for tooth enamel formation. J Bone Miner Res. 2013 doi: 10.1002/jbmr.1895. [DOI] [PubMed] [Google Scholar]
- 47.Bergmann H, Yabas M, Short A, Miosge L, Barthel N, Teh CE, Roots CM, Bull KR, Jeelall Y, Horikawa K, Whittle B, Balakishnan B, Sjollema G, Bertram EM, Mackay F, Rimmer AJ, Cornall RJ, Field MA, Andrews TD, Goodnow CC, Enders A. B cell survival, surface BCR and BAFFR expression, CD74 metabolism, and CD8- dendritic cells require the intramembrane endopeptidase SPPL2A. J Exp Med. 2013;210:31–40. doi: 10.1084/jem.20121076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Schneppenheim J, Dressel R, Huttl S, Lullmann-Rauch R, Engelke M, Dittmann K, Wienands J, Eskelinen EL, Hermans-Borgmeyer I, Fluhrer R, Saftig P, Schroder B. The intramembrane protease SPPL2a promotes B cell development and controls endosomal traffic by cleavage of the invariant chain. J Exp Med. 2013;210:41–58. doi: 10.1084/jem.20121069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Beisner DR, Langerak P, Parker AE, Dahlberg C, Otero FJ, Sutton SE, Poirot L, Barnes W, Young MA, Niessen S, Wiltshire T, Bodendorf U, Martoglio B, Cravatt B, Cooke MP. The intramembrane protease Sppl2a is required for B cell and DC development and survival via cleavage of the invariant chain. J Exp Med. 2013;210:23–30. doi: 10.1084/jem.20121072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Voss M, Fukumori A, Kuhn PH, Kunzel U, Klier B, Grammer G, Haug-Kroper M, Kremmer E, Lichtenthaler SF, Steiner H, Schroder B, Haass C, Fluhrer R. Foamy virus envelope protein is a substrate for signal peptide peptidase-like 3 (SPPL3) J Biol Chem. 2012;287:43401–43409. doi: 10.1074/jbc.M112.371369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- 52.Golde TE, Schneider LS, Koo EH. Anti-abeta therapeutics in Alzheimer's disease: the need for a paradigm shift. Neuron. 2011;69:203–213. doi: 10.1016/j.neuron.2011.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Das P, Verbeeck C, Minter L, Chakrabarty P, Felsenstein K, Kukar T, Maharvi G, Fauq A, Osborne BA, Golde TE. Transient pharmacologic lowering of Abeta production prior to deposition results in sustained reduction of amyloid plaque pathology. Mol Neurodegener. 2012;7:39. doi: 10.1186/1750-1326-7-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Garcia-Alloza M, Subramanian M, Thyssen D, Borrelli LA, Fauq A, Das P, Golde TE, Hyman BT, Bacskai BJ. Existing plaques and neuritic abnormalities in APP:PS1 mice are not affected by administration of the gamma-secretase inhibitor LY-411575. Mol Neurodegener. 2009;4:19. doi: 10.1186/1750-1326-4-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schor NF. What the halted phase III gamma-secretase inhibitor trial may (or may not) be telling us. Ann Neurol. 2011;69:237–239. doi: 10.1002/ana.22365. [DOI] [PubMed] [Google Scholar]
- 56.Cummings J. What can be inferred from the interruption of the semagacestat trial for treatment of Alzheimer's disease? Biol Psychiatry. 2010;68:876–878. doi: 10.1016/j.biopsych.2010.09.020. [DOI] [PubMed] [Google Scholar]
- 57.Samson K. NerveCenter: Phase III Alzheimer trial halted: Search for therapeutic biomarkers continues. Ann Neurol. 2010;68:A9–A12. doi: 10.1002/ana.22249. [DOI] [PubMed] [Google Scholar]
- 58.Extance A. Alzheimer's failure raises questions about disease-modifying strategies. Nat Rev Drug Discov. 2010;9:749–751. doi: 10.1038/nrd3288. [DOI] [PubMed] [Google Scholar]
- 59.Rizzo P, Osipo C, Foreman K, Golde T, Osborne B, Miele L. Rational targeting of Notch signaling in cancer. Oncogene. 2008;27:5124–5131. doi: 10.1038/onc.2008.226. [DOI] [PubMed] [Google Scholar]
- 60.Palomero T, Ferrando A. Therapeutic targeting of NOTCH1 signaling in T-cell acute lymphoblastic leukemia. Clin Lymphoma Myeloma. 2009;9(3):S205–210. doi: 10.3816/CLM.2009.s.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Li T, Wen H, Brayton C, Das P, Smithson LA, Fauq A, Fan X, Crain BJ, Price DL, Golde TE, Eberhart CG, Wong PC. Epidermal growth factor receptor and notch pathways participate in the tumor suppressor function of gamma-secretase. J Biol Chem. 2007;282:32264–32273. doi: 10.1074/jbc.M703649200. [DOI] [PubMed] [Google Scholar]
- 62.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. 2002;21:5408–5416. doi: 10.1093/emboj/cdf541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Xiao Y, Ye Y, Zou X, Jones S, Yearsley K, Shetuni B, Tellez J, Barsky SH. The lymphovascular embolus of inflammatory breast cancer exhibits a Notch 3 addiction. Oncogene. 2011;30:287–300. doi: 10.1038/onc.2010.405. [DOI] [PubMed] [Google Scholar]
- 64.Wei P, Walls M, Qiu M, Ding R, Denlinger RH, Wong A, Tsaparikos K, Jani JP, Hosea N, Sands M, Randolph S, Smeal T. Evaluation of selective gamma-secretase inhibitor PF-03084014 for its antitumor efficacy and gastrointestinal safety to guide optimal clinical trial design. Mol Cancer Ther. 2010;9:1618–1628. doi: 10.1158/1535-7163.MCT-10-0034. [DOI] [PubMed] [Google Scholar]
- 65.Wang M, Wu L, Wang L, Xin X. Down-regulation of Notch1 by gamma-secretase inhibition contributes to cell growth inhibition and apoptosis in ovarian cancer cells A2780. Biochem Biophys Res Commun. 2010;393:144–149. doi: 10.1016/j.bbrc.2010.01.103. [DOI] [PubMed] [Google Scholar]
- 66.Peignon G, Durand A, Cacheux W, Ayrault O, Terris B, Laurent-Puig P, Shroyer NF, Van Seuningen I, Honjo T, Perret C, Romagnolo B. Complex interplay between beta-catenin signalling and Notch effectors in intestinal tumorigenesis. Gut. 2011;60:166–176. doi: 10.1136/gut.2009.204719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Guo S, Liu M, Gonzalez-Perez RR. Role of Notch and its oncogenic signaling crosstalk in breast cancer. Biochim Biophys Acta. 2011;1815:197–213. doi: 10.1016/j.bbcan.2010.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Watters JW, Cheng C, Majumder PK, Wang R, Yalavarthi S, Meeske C, Kong L, Sun W, Lin J, Heyer J, Ware C, Winter C, Reilly JF, Demuth T, Clark S, Chiu MI, Robinson MO, Kohl N, Kannan K. De novo discovery of a gamma-secretase inhibitor response signature using a novel in vivo breast tumor model. Cancer Res. 2009;69:8949–8957. doi: 10.1158/0008-5472.CAN-09-1544. [DOI] [PubMed] [Google Scholar]
- 69.Rodilla V, Villanueva A, Obrador-Hevia A, Robert-Moreno A, Fernandez-Majada V, Grilli A, Lopez-Bigas N, Bellora N, Alba MM, Torres F, Dunach M, Sanjuan X, Gonzalez S, Gridley T, Capella G, Bigas A, Espinosa L. Jagged1 is the pathological link between Wnt and Notch pathways in colorectal cancer. Proc Natl Acad Sci U S A. 2009;106:6315–6320. doi: 10.1073/pnas.0813221106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Rahimi N, Golde TE, Meyer RD. Identification of ligand-induced proteolytic cleavage and ectodomain shedding of VEGFR-1/FLT1 in leukemic cancer cells. Cancer Res. 2009;69:2607–2614. doi: 10.1158/0008-5472.CAN-08-2905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mullendore ME, Koorstra JB, Li YM, Offerhaus GJ, Fan X, Henderson CM, Matsui W, Eberhart CG, Maitra A, Feldmann G. Ligand-dependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer. Clin Cancer Res. 2009;15:2291–2301. doi: 10.1158/1078-0432.CCR-08-2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Engin F, Bertin T, Ma O, Jiang MM, Wang L, Sutton RE, Donehower LA, Lee B. Notch signaling contributes to the pathogenesis of human osteosarcomas. Hum Mol Genet. 2009;18:1464–1470. doi: 10.1093/hmg/ddp057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Meng RD, Shelton CC, Li YM, Qin LX, Notterman D, Paty PB, Schwartz GK. gamma-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer Res. 2009;69:573–582. doi: 10.1158/0008-5472.CAN-08-2088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Choi JH, Park JT, Davidson B, Morin PJ, Shih Ie M, Wang TL. Jagged-1 and Notch3 juxtacrine loop regulates ovarian tumor growth and adhesion. Cancer Res. 2008;68:5716–5723. doi: 10.1158/0008-5472.CAN-08-0001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lee CW, Raskett CM, Prudovsky I, Altieri DC. Molecular dependence of estrogen receptor-negative breast cancer on a notch-survivin signaling axis. Cancer Res. 2008;68:5273–5281. doi: 10.1158/0008-5472.CAN-07-6673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhang P, Yang Y, Zweidler-McKay PA, Hughes DP. Critical role of notch signaling in osteosarcoma invasion and metastasis. Clin Cancer Res. 2008;14:2962–2969. doi: 10.1158/1078-0432.CCR-07-1992. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 77.van Es JH, Clevers H. Notch and Wnt inhibitors as potential new drugs for intestinal neoplastic disease. Trends Mol Med. 2005;11:496–502. doi: 10.1016/j.molmed.2005.09.008. [DOI] [PubMed] [Google Scholar]
- 78.Miele L, Miao H, Nickoloff BJ. NOTCH signaling as a novel cancer therapeutic target. Curr Cancer Drug Targets. 2006;6:313–323. doi: 10.2174/156800906777441771. [DOI] [PubMed] [Google Scholar]
- 79.Pear WS, Aster JC. T cell acute lymphoblastic leukemia/lymphoma: a human cancer commonly associated with aberrant NOTCH1 signaling. Curr Opin Hematol. 2004;11:426–433. doi: 10.1097/01.moh.0000143965.90813.70. [DOI] [PubMed] [Google Scholar]
- 80.Hassan KA, Wang L, Korkaya H, Chen G, Maillard I, Beer DG, Kalemkerian GP, Wicha MS. Notch Pathway Activity Identifies Cells with Cancer Stem Cell-like Properties and Correlates with Worse Survival in Lung Adenocarcinoma. Clin Cancer Res. 2013;19:1972–1980. doi: 10.1158/1078-0432.CCR-12-0370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yabuuchi S, Pai SG, Campbell NR, de Wilde RF, De Oliveira E, Korangath P, Streppel MM, Rasheed ZA, Hidalgo M, Maitra A, Rajeshkumar NV. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013 doi: 10.1016/j.canlet.2013.01.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Roderick J, Gonzales-Perez G, Kukshin CA, Dongre A, ER R, Srinivasan J, Andrzejewski C, Fauq AH, Golde TE, Miele L, LM M. Therapeutic targeting of NOTCH signaling ameliorates immune-mediated bone marrow failure. J Exp Med. 2013 doi: 10.1084/jem.20112615. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Minter LM, Turley DM, Das P, Shin HM, Joshi I, Lawlor RG, Cho OH, Palaga T, Gottipati S, Telfer JC, Kostura L, Fauq AH, Simpson K, Such KA, Miele L, Golde TE, Miller SD, Osborne BA. Inhibitors of gamma-secretase block in vivo and in vitro T helper type 1 polarization by preventing Notch upregulation of Tbx21. Nat Immunol. 2005;6:680–688. [PubMed] [Google Scholar]
- 84.Piggott K, Deng J, Warrington K, Younge B, Kubo JT, Desai M, Goronzy JJ, Weyand CM. Blocking the NOTCH pathway inhibits vascular inflammation in large-vessel vasculitis. Circulation. 2011;123:309–318. doi: 10.1161/CIRCULATIONAHA.110.936203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ablonczy Z, Prakasam A, Fant J, Fauq A, Crosson C, Sambamurti K. Pigment epithelium-derived factor maintains retinal pigment epithelium function by inhibiting vascular endothelial growth factor-R2 signaling through gamma-secretase. J Biol Chem. 2009;284:30177–30186. doi: 10.1074/jbc.M109.032391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lin CL, Wang FS, Hsu YC, Chen CN, Tseng MJ, Saleem MA, Chang PJ, Wang JY. Modulation of notch-1 signaling alleviates vascular endothelial growth factor-mediated diabetic nephropathy. Diabetes. 2010;59:1915–1925. doi: 10.2337/db09-0663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Thomas DB, Susztak K. The Notch pathway in podocytes plays a role in the development of glomerular disease. Nat Med. 2008;14:290–298. doi: 10.1038/nm1731. [DOI] [PubMed] [Google Scholar]
- 88.Huang R, Zhou Q, Veeraragoo P, Yu H, Xiao Z. Notch2/Hes-1 pathway plays an important role in renal ischemia and reperfusion injury-associated inflammation and apoptosis and the gamma-secretase inhibitor DAPT has a nephroprotective effect. Ren Fail. 2011;33:207–216. doi: 10.3109/0886022X.2011.553979. [DOI] [PubMed] [Google Scholar]
- 89.Arumugam TV, Chan SL, Jo DG, Yilmaz G, Tang SC, Cheng A, Gleichmann M, Okun E, Dixit VD, Chigurupati S, Mughal MR, Ouyang X, Miele L, Magnus T, Poosala S, Granger DN, Mattson MP. Gamma secretase-mediated Notch signaling worsens brain damage and functional outcome in ischemic stroke. Nat Med. 2006;12:621–623. doi: 10.1038/nm1403. [DOI] [PubMed] [Google Scholar]
- 90.Loane DJ, Pocivavsek A, Moussa CE, Thompson R, Matsuoka Y, Faden AI, Rebeck GW, Burns MP. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nat Med. 2009;15:377–379. doi: 10.1038/nm.1940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Mizutari K, Fujioka M, Hosoya M, Bramhall N, Okano HJ, Okano H, Edge AS. Notch inhibition induces cochlear hair cell regeneration and recovery of hearing after acoustic trauma. Neuron. 2013;77:58–69. doi: 10.1016/j.neuron.2012.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Bielesz B, Sirin Y, Si H, Niranjan T, Gruenwald A, Ahn S, Kato H, Pullman J, Gessler M, Haase VH, Susztak K. Epithelial Notch signaling regulates interstitial fibrosis development in the kidneys of mice and humans. J Clin Invest. 2010;120:4040–4054. doi: 10.1172/JCI43025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Miele L, Golde T, Osborne B. Notch signaling in cancer. Curr Mol Med. 2006;6:905–918. doi: 10.2174/156652406779010830. [DOI] [PubMed] [Google Scholar]
- 94.Purow B. Notch inhibition as a promising new approach to cancer therapy. Adv Exp Med Biol. 2012;727:305–319. doi: 10.1007/978-1-4614-0899-4_23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Milner LA, Bigas A. Notch as a mediator of cell fate determination in hematopoiesis: evidence and speculation. Blood. 1999;93:2431–2448. [PubMed] [Google Scholar]
- 96.Osborne B, Miele L. Notch and the immune system. Immunity. 1999;11:653–663. doi: 10.1016/s1074-7613(00)80140-5. [DOI] [PubMed] [Google Scholar]
- 97.Aster JC, Pear WS. Notch signaling in leukemia. Curr Opin Hematol. 2001;8:237–244. doi: 10.1097/00062752-200107000-00010. [DOI] [PubMed] [Google Scholar]
- 98.Maillard I, Adler SH, Pear WS. Notch and the immune system. Immunity. 2003;19:781–791. doi: 10.1016/s1074-7613(03)00325-x. [DOI] [PubMed] [Google Scholar]
- 99.Vo K, Amarasinghe B, Washington K, Gonzalez A, Berlin J, Dang TP. Targeting notch pathway enhances rapamycin antitumor activity in pancreas cancers through PTEN phosphorylation. Mol Cancer. 2011;10:138. doi: 10.1186/1476-4598-10-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gu JW, Rizzo P, Pannuti A, Golde T, Osborne B, Miele L. Notch signals in the endothelium and cancer “stem-like” cells: opportunities for cancer therapy. Vasc Cell. 2012;4:7. doi: 10.1186/2045-824X-4-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Wang R, Tang P, Wang P, Boissy RE, Zheng H. Regulation of tyrosinase trafficking and processing by presenilins: partial loss of function by familial Alzheimer's disease mutation. Proc Natl Acad Sci U S A. 2006;103:353–358. doi: 10.1073/pnas.0509822102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Guo S, Gonzalez-Perez RR. Notch, IL-1 and leptin crosstalk outcome (NILCO) is critical for leptin-induced proliferation, migration and VEGF/VEGFR-2 expression in breast cancer. PLoS One. 2011;6:e21467. doi: 10.1371/journal.pone.0021467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Clementz AG, Rogowski A, Pandya K, Miele L, Osipo C. NOTCH-1 and NOTCH-4 are novel gene targets of PEA3 in breast cancer: novel therapeutic implications. Breast Cancer Res. 2011;13:R63. doi: 10.1186/bcr2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.De Strooper B, Annaert W. Novel research horizons for presenilins and gamma-secretases in cell biology and disease. Annu Rev Cell Dev Biol. 2010;26:235–260. doi: 10.1146/annurev-cellbio-100109-104117. [DOI] [PubMed] [Google Scholar]
- 105.Zhao B, Yu M, Neitzel M, Marugg J, Jagodzinski J, Lee M, Hu K, Schenk D, Yednock T, Basi G. Identification of gamma-secretase inhibitor potency determinants on presenilin. J Biol Chem. 2008;283:2927–2938. doi: 10.1074/jbc.M708870200. [DOI] [PubMed] [Google Scholar]
- 106.Borgegard T, Gustavsson S, Nilsson C, Parpal S, Klintenberg R, Berg AL, Rosqvist S, Serneels L, Svensson S, Olsson F, Jin S, Yan H, Wanngren J, Jureus A, Ridderstad-Wollberg A, Wollberg P, Stockling K, Karlstrom H, Malmberg A, Lund J, Arvidsson PI, De Strooper B, Lendahl U, Lundkvist J. Alzheimer's disease: presenilin 2-sparing gamma-secretase inhibition is a tolerable Abeta peptide-lowering strategy. J Neurosci. 2012;32:17297–17305. doi: 10.1523/JNEUROSCI.1451-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Serneels L, Van Biervliet J, Craessaerts K, Dejaegere T, Horre K, Van Houtvin T, Esselmann H, Paul S, Schafer MK, Berezovska O, Hyman BT, Sprangers B, Sciot R, Moons L, Jucker M, Yang Z, May PC, Karran E, Wiltfang J, D'Hooge R, De Strooper B. gamma-Secretase heterogeneity in the Aph1 subunit: relevance for Alzheimer's disease. Science. 2009;324:639–642. doi: 10.1126/science.1171176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lee J, Song L, Terracina G, Bara T, Josien H, Asberom T, Sasikumar TK, Burnett DA, Clader J, Parker EM, Zhang L. Identification of Presenilin 1 Selective gamma-Secretase Inhibitors with Reconstituted gamma-Secretase Complexes. Biochemistry. 2011 doi: 10.1021/bi200026m. [DOI] [PubMed] [Google Scholar]
- 109.Placanica L, Chien JW, Li YM. Characterization of an atypical gamma-secretase complex from hematopoietic origin. Biochemistry. 2010;49:2796–2804. doi: 10.1021/bi901388t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wang H, Luo WJ, Zhang YW, Li YM, Thinakaran G, Greengard P, Xu H. Presenilins and gamma-secretase inhibitors affect intracellular trafficking and cell surface localization of the gamma-secretase complex components. J Biol Chem. 2004;279:40560–40566. doi: 10.1074/jbc.M404345200. [DOI] [PubMed] [Google Scholar]
- 111.Wolfe MS. gamma-Secretase as a target for Alzheimer's disease. Adv Pharmacol. 2012;64:127–153. doi: 10.1016/B978-0-12-394816-8.00004-0. [DOI] [PubMed] [Google Scholar]
- 112.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. 2008 doi: 10.1021/jm801252w. [DOI] [PubMed] [Google Scholar]
- 113.Ye XM, Konradi AW, Smith J, Aubele DL, Garofalo AW, Marugg J, Neitzel ML, Semko CM, Sham HL, Sun M, Truong AP, Wu J, Zhang H, Goldbach E, Sauer JM, Brigham EF, Bova M, Basi GS. Discovery of a novel sulfonamide-pyrazolopiperidine series as potent and efficacious gamma-secretase inhibitors (Part II) Bioorg Med Chem Lett. 2010;20:3502–3506. doi: 10.1016/j.bmcl.2010.04.148. [DOI] [PubMed] [Google Scholar]
- 114.Basi GS, Hemphill S, Brigham EF, Liao A, Aubele DL, Baker J, Barbour R, Bova M, Chen XH, Dappen MS, Eichenbaum T, Goldbach E, Hawkinson J, Lawler-Herbold R, Hu K, Hui T, Jagodzinski JJ, Keim PS, Kholodenko D, Latimer LH, Lee M, Marugg J, Mattson MN, McCauley S, Miller JL, Motter R, Mutter L, Neitzel ML, Ni H, Nguyen L, Quinn K, Ruslim L, Semko CM, Shapiro P, Smith J, Soriano F, Szoke B, Tanaka K, Tang P, Tucker JA, Ye XM, Yu M, Wu J, Xu YZ, Garofalo AW, Sauer JM, Konradi AW, Ness D, Shopp G, Pleiss MA, Freedman SB, Schenk D. Amyloid precursor protein selective gamma-secretase inhibitors for treatment of Alzheimer's disease. Alzheimers Res Ther. 2010;2:36. doi: 10.1186/alzrt60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Albright CF, Dockens RC, Meredith JE, Jr, Olson RE, Slemmon R, Lentz KA, Wang JS, Denton RR, Pilcher G, Rhyne PW, Raybon JJ, Barten DM, Burton C, Toyn JH, Sankaranarayanan S, Polson C, Guss V, White R, Simutis F, Sanderson T, Gillman KW, Starrett JE, Jr, Bronson J, Sverdlov O, Huang SP, Castaneda L, Feldman H, Coric V, Zaczek R, Macor JE, Houston J, Berman RM, Tong G. Pharmacodynamics of selective inhibition of gamma-secretase by avagacestat. J Pharmacol Exp Ther. 2013;344:686–695. doi: 10.1124/jpet.112.199356. [DOI] [PubMed] [Google Scholar]
- 116.Tong G, Castaneda L, Wang JS, Sverdlov O, Huang SP, Slemmon R, Gu H, Wong O, Li H, Berman RM, Smith C, Albright C, Dockens RC. Effects of single doses of avagacestat (BMS-708163) on cerebrospinal fluid Abeta levels in healthy young men. Clin Drug Investig. 2012;32:761–769. doi: 10.1007/s40261-012-0006-4. [DOI] [PubMed] [Google Scholar]
- 117.Crump CJ, Castro SV, Wang F, Pozdnyakov N, Ballard TE, Sisodia SS, Bales KR, Johnson DS, Li YM. BMS-708,163 targets presenilin and lacks notch-sparing activity. Biochemistry. 2012;51:7209–7211. doi: 10.1021/bi301137h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.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. 2012;69:1430–1440. doi: 10.1001/archneurol.2012.2194. [DOI] [PubMed] [Google Scholar]
- 119.Kurosumi M, Nishio Y, Osawa S, Kobayashi H, Iwatsubo T, Tomita T, Miyachi H. Novel Notch-sparing gamma-secretase inhibitors derived from a peroxisome proliferator-activated receptor agonist library. Bioorg Med Chem Lett. 2010;20:5282–5285. doi: 10.1016/j.bmcl.2010.06.131. [DOI] [PubMed] [Google Scholar]
- 120.Chavez-Gutierrez L, Bammens L, Benilova I, Vandersteen A, Benurwar M, Borgers M, Lismont S, Zhou L, Van Cleynenbreugel S, Esselmann H, Wiltfang J, Serneels L, Karran E, Gijsen H, Schymkowitz J, Rousseau F, Broersen K, De Strooper B. The mechanism of gamma-Secretase dysfunction in familial Alzheimer disease. EMBO J. 2012;31:2261–2274. doi: 10.1038/emboj.2012.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Karlstrom H, Bergman A, Lendahl U, Naslund J, Lundkvist J. A sensitive and quantitative assay for measuring cleavage of presenilin substrates. J Biol Chem. 2002;277:6763–6766. doi: 10.1074/jbc.C100649200. [DOI] [PubMed] [Google Scholar]
- 122.Gunyuzlu PL, White WH, Davis GL, Hollis GF, Toyn JH. A yeast genetic assay for caspase cleavage of the amyloid-beta precursor protein. Biotechnol. 2000;15:29–37. doi: 10.1385/MB:15:1:29. [DOI] [PubMed] [Google Scholar]
- 123.Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT, Froehlich AM, Paranilam J, Hayes DF, Wicha MS, Chang JC. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19:1512–1524. doi: 10.1158/1078-0432.CCR-11-3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Real PJ, Tosello V, Palomero T, Castillo M, Hernando E, de Stanchina E, Sulis ML, Barnes K, Sawai C, Homminga I, Meijerink J, Aifantis I, Basso G, Cordon-Cardo C, Ai W, Ferrando A. Gamma-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia. Nat Med. 2009;15:50–58. doi: 10.1038/nm.1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Golde TE, Eckman CB, Younkin SG. Biochemical detection of Abeta isoforms: implications for pathogenesis, diagnosis, and treatment of Alzheimer's disease. Biochim Biophys Acta. 2000;1502:172–187. doi: 10.1016/s0925-4439(00)00043-0. [DOI] [PubMed] [Google Scholar]
- 126.Selkoe D, Kopan R. Notch and Presenilin: regulated intramembrane proteolysis links development and degeneration. Annu Rev Neurosci. 2003;26:565–597. doi: 10.1146/annurev.neuro.26.041002.131334. [DOI] [PubMed] [Google Scholar]
- 127.Lichtenthaler SF, Ida N, Multhaup G, Masters CL, Beyreuther K. Mutations in the transmembrane domain of APP altering gamma-secretase specificity. Biochemistry. 1997;36:15396–15403. doi: 10.1021/bi971071m. [DOI] [PubMed] [Google Scholar]
- 128.Lichtenthaler SF, Wang R, Grimm H, Uljon SN, Masters CL, Beyreuther K. Mechanism of the cleavage specificity of Alzheimer's disease gamma-secretase identified by phenylalanine-scanning mutagenesis of the transmembrane domain of the amyloid precursor protein. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:3053–3058. doi: 10.1073/pnas.96.6.3053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sagi SA, Lessard CB, Winden KD, Maruyama H, Koo JC, Weggen S, Kukar TL, Golde TE, Koo EH. Substrate sequence influences gamma-secretase modulator activity, role of the transmembrane domain of the amyloid precursor protein. J Biol Chem. 2011;286:39794–39803. doi: 10.1074/jbc.M111.277228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kukar TL, Ladd TB, Robertson P, Pintchovski SA, Moore B, Bann MA, Ren Z, Jansen-West K, Malphrus K, Eggert S, Maruyama H, Cottrell BA, Das P, Basi GS, Koo EH, Golde TE. Lysine 624 of the amyloid precursor protein (APP) is a critical determinant of amyloid beta peptide length: support for a sequential model of gamma-secretase intramembrane proteolysis and regulation by the amyloid beta precursor protein (APP) juxtamembrane region. J Biol Chem. 2011;286:39804–39812. doi: 10.1074/jbc.M111.274696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Tarassishin L, Yin YI, Bassit B, Li YM. Processing of Notch and amyloid precursor protein by gamma-secretase is spatially distinct. Proc Natl Acad Sci U S A. 2004;101:17050–17055. doi: 10.1073/pnas.0408007101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wahrle S, Das P, Nyborg AC, McLendon C, Shoji M, Kawarabayashi T, Younkin LH, Younkin SG, Golde TE. Cholesterol-Dependent gamma-Secretase Activity in Buoyant Cholesterol- Rich Membrane Microdomains. Neurobiol Dis. 2002;9:11–23. doi: 10.1006/nbdi.2001.0470. [DOI] [PubMed] [Google Scholar]
- 133.Vetrivel KS, Cheng H, Lin W, Sakurai T, Li T, Nukina N, Wong PC, Xu H, Thinakaran G. Association of gamma -secretase with lipid rafts in post-golgi and endosome membranes. J Biol Chem. 2004 doi: 10.1074/jbc.M407986200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Vetrivel KS, Gong P, Bowen JW, Cheng H, Chen Y, Carter M, Nguyen PD, Placanica L, Wieland FT, Li YM, Kounnas MZ, Thinakaran G. Dual roles of the transmembrane protein p23/TMP21 in the modulation of amyloid precursor protein metabolism. Mol Neurodegener. 2007;2:4. doi: 10.1186/1750-1326-2-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wakabayashi T, Craessaerts K, Bammens L, Bentahir M, Borgions F, Herdewijn P, Staes A, Timmerman E, Vandekerckhove J, Rubinstein E, Boucheix C, Gevaert K, De Strooper B. Analysis of the gamma-secretase interactome and validation of its association with tetraspanin-enriched microdomains. Nat Cell Biol. 2009;11:1340–1346. doi: 10.1038/ncb1978. [DOI] [PubMed] [Google Scholar]
- 136.Hur JY, Welander H, Behbahani H, Aoki M, Franberg J, Winblad B, Frykman S, Tjernberg LO. Active gamma-secretase is localized to detergent-resistant membranes in human brain. FEBS J. 2008;275:1174–1187. doi: 10.1111/j.1742-4658.2008.06278.x. [DOI] [PubMed] [Google Scholar]
- 137.Wang R, Sweeney D, Gandy SE, Sisodia SS. The profile of soluble amyloid beta protein in cultured cell medi Detection and quantification of amyloid beta protein and variants by immunoprecipitation-mass spectrometry. Journal of Biological Chemistry. 1996;271:31894–31902. doi: 10.1074/jbc.271.50.31894. [DOI] [PubMed] [Google Scholar]
- 138.Younkin SG. The role of A beta 42 in Alzheimer's disease. Journal of Physiology, Paris. 1998;92:289–292. doi: 10.1016/s0928-4257(98)80035-1. [DOI] [PubMed] [Google Scholar]
- 139.Golde TE. The pathogenesis of Alzheimer's disease and the role of Abeta42. CNS Spectr. 2007;12:4–6. doi: 10.1017/s1092852900025876. [DOI] [PubMed] [Google Scholar]
- 140.Selkoe DJ. Alzheimer's disease: genes, proteins, and therapy. Physiol Rev. 2001;81:741–766. doi: 10.1152/physrev.2001.81.2.741. [DOI] [PubMed] [Google Scholar]
- 141.Jack CR, Jr, Lowe VJ, Weigand SD, Wiste HJ, Senjem ML, Knopman DS, Shiung MM, Gunter JL, Boeve BF, Kemp BJ, Weiner M, Petersen RC. Serial PIB and MRI in normal, mild cognitive impairment and Alzheimer's disease: implications for sequence of pathological events in Alzheimer's disease. Brain. 2009 doi: 10.1093/brain/awp062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jarrett JT, Berger EP, TL P., Jr The carboxy terminus of ß amyloid protein is critical for the seeding of amyloid formation: Implications for pathogenesis of Alzheimer's disease. Biochem. 1993;32:4693–4697. doi: 10.1021/bi00069a001. [DOI] [PubMed] [Google Scholar]
- 143.Jarrett JT, TL P., Jr Seeding “one dimensional crystallization” of amyloid: A pathogenic mechanism in Alzheimer's disease and Scrapie? Cell. 1993;73:1055–1058. doi: 10.1016/0092-8674(93)90635-4. [DOI] [PubMed] [Google Scholar]
- 144.Duff K, Eckman C, Zehr C, Yu X, Prada CM, Perez-tur J, Hutton M, Buee L, Harigaya Y, Yager D, Morgan D, Gordon MN, Holcomb L, Refolo L, Zenk B, Hardy J, Younkin S. Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature. 1996;383:710–713. doi: 10.1038/383710a0. [DOI] [PubMed] [Google Scholar]
- 145.Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F, Guido T, Hagoplan S, Johnson-Wood K, Khan K, Lee M, Leibowitz P, Lieberburg I, Little S, Masliah E, McConlogue L, Montoya-Zavala M, Mucke L, Paganini L, Penniman E, Power M, Schenk D, Seubert P, Snyder B, Sorlano F, Tan H, Vitale J, Wadsworth S, Wolozin B, Zhao J. Alzheimer-type neuropathology in transgenic mice overexpressing V717F ß-amyloid precursor protein. Nature. 1995;373:523–527. doi: 10.1038/373523a0. [DOI] [PubMed] [Google Scholar]
- 146.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. 2004;101:6623–6628. doi: 10.1073/pnas.0400895101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Greeve I, Kretzschmar D, Tschape JA, Beyn A, Brellinger C, Schweizer M, Nitsch RM, Reifegerste R. Age-dependent neurodegeneration and Alzheimer-amyloid plaque formation in transgenic Drosophila. J Neurosci. 2004;24:3899–3906. doi: 10.1523/JNEUROSCI.0283-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.McGowan E, Onstead L, Pickford F, Kim J, Beard J, Murphy MP, Yu C, Das P, Golde TE. Selective Overexpression of Abeta42, but not Abeta40, in the secretory pathway is sufficient for plaque deposition in mice. Society for Neuroscience Abstracts, Program. 2003;877(14) [Google Scholar]
- 149.Herzig MC, Winkler DT, Burgermeister P, Pfeifer M, Kohler E, Schmidt SD, Danner S, Abramowski D, Sturchler-Pierrat C, Burki K, van Duinen SG, Maat-Schieman ML, Staufenbiel M, Mathews PM, Jucker M. Abeta is targeted to the vasculature in a mouse model of hereditary cerebral hemorrhage with amyloidosis. Nat Neurosci. 2004;7:954–960. doi: 10.1038/nn1302. [DOI] [PubMed] [Google Scholar]
- 150.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. 2003;278:31831–31837. doi: 10.1074/jbc.M303592200. [DOI] [PubMed] [Google Scholar]
- 151.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. 2003;112:440–449. doi: 10.1172/JCI18162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.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. 2001;414:212–216. doi: 10.1038/35102591. [DOI] [PubMed] [Google Scholar]
- 153.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. 2005;11:545–550. doi: 10.1038/nm1235. [DOI] [PubMed] [Google Scholar]
- 154.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. 2004;88:337–348. doi: 10.1111/j.1471-4159.2004.02154.x. [DOI] [PubMed] [Google Scholar]
- 155.Rogers K, Felsenstein KM, Hrdlicka L, Tu Z, Albayya F, Lee W, Hopp S, Miller MJ, Spaulding D, Yang Z, Hodgdon H, Nolan S, Wen M, Costa D, Blain JF, Freeman E, De Strooper B, Vulsteke V, Scrocchi L, Zetterberg H, Portelius E, Hutter-Paier B, Havas D, Ahlijanian M, Flood D, Leventhal L, Shapiro G, Patzke H, Chesworth R, Koenig G. Modulation of gamma-secretase by EVP-0015962 reduces amyloid deposition and behavioral deficits in Tg2576 mice. Mol Neurodegener. 2012;7:61. doi: 10.1186/1750-1326-7-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.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 B, Strooper A. Leon, 1-(3′,4′-Dichloro-2-fluoro[1,1′-biphenyl]-4-yl)-cyclopropanecarboxylic 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. 2007;323:822–830. doi: 10.1124/jpet.107.129007. [DOI] [PubMed] [Google Scholar]
- 157.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. 2007;55:318–328. doi: 10.1016/j.phrs.2006.12.010. [DOI] [PubMed] [Google Scholar]
- 158.Portelius E, Van Broeck B, Andreasson U, Gustavsson MK, Mercken M, Zetterberg H, Borghys H, Blennow K. Acute effect on the Abeta isoform pattern in CSF in response to gamma-secretase modulator and inhibitor treatment in dogs. J Alzheimers Dis. 2010;21:1005–1012. doi: 10.3233/JAD-2010-100573. [DOI] [PubMed] [Google Scholar]
- 159.Qin J, Dhondi P, Huang X, Mandal M, Zhao Z, Pissarnitski D, Zhou W, Aslanian R, Zhu Z, Greenlee W, Clader J, Zhang L, Cohen-Williams M, Jones N, Hyde L, Palani A. Discovery of fused 5,6-bicyclic heterocycles as gamma-secretase modulators. Bioorg Med Chem Lett. 2011;21:664–669. doi: 10.1016/j.bmcl.2010.12.012. [DOI] [PubMed] [Google Scholar]
- 160.Gijsen HJ, Mercken M. gamma-Secretase Modulators: Can We Combine Potency with Safety? Int J Alzheimers Dis. 2012;2012:295207. doi: 10.1155/2012/295207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.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. 2010;20:1306–1311. doi: 10.1016/j.bmcl.2009.08.072. [DOI] [PubMed] [Google Scholar]
- 162.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. 2010;20:755–758. doi: 10.1016/j.bmcl.2009.11.034. [DOI] [PubMed] [Google Scholar]
- 163.Li H, Qin J, Dhondi P, Zhou W, Vicarel M, Bara T, Cole D, Josien H, Pissarnitski D, Zhu Z, Palani A, Aslanian R, Clader J, Czarniecki M, Greenlee W, Cohen-Williams M, Hyde L, Song L, Zhang L, Chu I, Huang X. The discovery of fused oxadiazepines as gamma secretase modulators for treatment of Alzheimer's disease. Bioorg Med Chem Lett. 2013;23:466–471. doi: 10.1016/j.bmcl.2012.11.055. [DOI] [PubMed] [Google Scholar]
- 164.Sun ZY, Asberom T, Bara T, Bennett C, Burnett D, Chu I, Clader J, Cohen-Williams M, Cole D, Czarniecki M, Durkin J, Gallo G, Greenlee W, Josien H, Huang X, Hyde L, Jones N, Kazakevich I, Li H, Liu X, Lee J, Maccoss M, Mandal MB, McCracken T, Nomeir A, Mazzola R, Palani A, Parker EM, Pissarnitski DA, Qin J, Song L, Terracina G, Vicarel M, Voigt J, Xu R, Zhang L, Zhang Q, Zhao Z, Zhu X, Zhu Z. Cyclic hydroxyamidines as amide isosteres: discovery of oxadiazolines and oxadiazines as potent and highly efficacious gamma-secretase modulators in vivo. J Med Chem. 2012;55:489–502. doi: 10.1021/jm201407j. [DOI] [PubMed] [Google Scholar]
- 165.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. 2010;20:2279–2282. doi: 10.1016/j.bmcl.2010.02.008. [DOI] [PubMed] [Google Scholar]
- 166.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. 2010;20:1269–1271. doi: 10.1016/j.bmcl.2009.11.101. [DOI] [PubMed] [Google Scholar]
- 167.Lubbers T, Flohr A, Jolidon S, David-Pierson P, Jacobsen H, Ozmen L, Baumann K. Aminothiazoles as gamma-secretase modulators. Bioorg Med Chem Lett. 2011;21:6554–6558. doi: 10.1016/j.bmcl.2011.08.060. [DOI] [PubMed] [Google Scholar]
- 168.Xin Z, Peng H, Zhang A, Talreja T, Kumaravel G, Xu L, Rohde E, Jung MY, Shackett MN, Kocisko D, Chollate S, Dunah AW, Snodgrass-Belt PA, Arnold HM, Taveras AG, Rhodes KJ, Scannevin RH. Discovery of 4-aminomethylphenylacetic acids as gamma-secretase modulators via a scaffold design approach. Bioorg Med Chem Lett. 2011;21:7277–7280. doi: 10.1016/j.bmcl.2011.10.047. [DOI] [PubMed] [Google Scholar]
- 169.Bischoff F, Berthelot D, De Cleyn M, Macdonald G, Minne G, Oehlrich D, Pieters S, Surkyn M, Trabanco AA, Tresadern G, Van Brandt S, Velter I, Zaja M, Borghys H, Masungi C, Mercken M, Gijsen HJ. Design and synthesis of a novel series of bicyclic heterocycles as potent gamma-secretase modulators. J Med Chem. 2012;55:9089–9106. doi: 10.1021/jm201710f. [DOI] [PubMed] [Google Scholar]
- 170.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. 2011;163:375–389. doi: 10.1111/j.1476-5381.2011.01207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.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. 2010;67:769–780. doi: 10.1016/j.neuron.2010.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Narlawar R, Perez Revuelta BI, Baumann K, Schubenel R, Haass C, Steiner H, Schmidt B. N-Substituted carbazolyloxyacetic acids modulate Alzheimer associated gamma-secretase. Bioorg Med Chem Lett. 2007;17:176–182. doi: 10.1016/j.bmcl.2006.09.061. [DOI] [PubMed] [Google Scholar]
- 173.Hubbs JL, Fuller NO, Austin WF, Shen R, Creaser SP, McKee TD, Loureiro RM, Tate B, Xia W, Ives J, Bronk BS. Optimization of a natural product-based class of gamma-secretase modulators. J Med Chem. 2012;55:9270–9282. doi: 10.1021/jm300976b. [DOI] [PubMed] [Google Scholar]
- 174.Loureiro RM, Dumin JA, McKee TD, Austin WF, Fuller NO, Hubbs JL, Shen R, Jonker J, Ives J, Bronk BS, Tate B. Efficacy of SPI-1865, a novel gamma-secretase modulator, in multiple rodent models. Alzheimers Res Ther. 2013;5:19. doi: 10.1186/alzrt173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.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 A 38 and A 42 Is Independently and Differentially Affected by Familial Alzheimer Disease-associated Presenilin Mutations and {gamma}-Secretase Modulation. J Biol Chem. 2008;283:677–683. doi: 10.1074/jbc.M708754200. [DOI] [PubMed] [Google Scholar]
- 176.Czirr E, Cottrell BA, Leuchtenberger S, Kukar T, Ladd TB, Esselmann H, Paul S, Schubenel R, Torpey JW, Pietrzik CU, Golde TE, Wiltfang J, Baumann K, Koo EH, Weggen S. Independent generation of Abeta 42 and Abeta 38 peptide species by gamma -secretase. J Biol Chem. 2008 doi: 10.1074/jbc.M802912200. [DOI] [PubMed] [Google Scholar]
- 177.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. 2003;278:30748–30754. doi: 10.1074/jbc.M304824200. [DOI] [PubMed] [Google Scholar]
- 178.Weggen S, Eriksen JL, Sagi SA, Pietrzik CU, Golde TE, Koo EH. A beta42-lowering nonsteroidal anti-inflammatory drugs preserve intramembrane cleavage of the amyloid precursor protein and ErbB-4 receptor and signaling through the APP intracellular domain. J Biol Chem. 2003 May 31;2003 doi: 10.1074/jbc.M304824200. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 179.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. 2008;453:925–929. doi: 10.1038/nature07055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Richter L, Munter LM, Ness J, Hildebrand PW, Dasari M, Unterreitmeier S, Bulic B, Beyermann M, Gust R, Reif B, Weggen S, Langosch D, Multhaup G. Amyloid beta 42 peptide (Abeta42)-lowering compounds directly bind to Abeta and interfere with amyloid precursor protein (APP) transmembrane dimerization. Proc Natl Acad Sci U S A. 2010;107:14597–14602. doi: 10.1073/pnas.1003026107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pozdnyakov N, Murrey HE, Crump CJ, Pettersson M, Ballard TE, Am Ende CW, Ahn K, Li YM, Bales KR, Johnson DS. gamma-Secretase Modulator (GSM) Photoaffinity Probes Reveal Distinct Allosteric Binding Sites on Presenilin. J Biol Chem. 2013;288:9710–9720. doi: 10.1074/jbc.M112.398602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.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. 2011;2:705–710. doi: 10.1021/cn200098p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.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. 2011;30:4815–4824. doi: 10.1038/emboj.2011.372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Czirr E, Leuchtenberger S, Dorner-Ciossek C, Schneider A, Jucker M, Koo EH, Pietrzik CU, Baumann K, Weggen S. Insensitivity to Abeta42-lowering nonsteroidal anti-inflammatory drugs and gamma-secretase inhibitors is common among aggressive presenilin-1 mutations. J Biol Chem. 2007;282:24504–24513. doi: 10.1074/jbc.M700618200. [DOI] [PubMed] [Google Scholar]
- 185.Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, Zavitz KH. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA. 2009;302:2557–2564. doi: 10.1001/jama.2009.1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kukar T, Prescott S, Eriksen JL, Holloway V, Murphy MP, Koo EH, Golde TE, Nicolle MM. Chronic administration of R-flurbiprofen attenuates learning impairments in transgenic amyloid precursor protein mice. BMC Neurosci. 2007;8:54. doi: 10.1186/1471-2202-8-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sivilia S, Lorenzini L, Giuliani A, Gusciglio M, Fernandez M, Baldassarro VA, Mangano C, Ferraro L, Pietrini V, Baroc MF, Viscomi AR, Ottonello S, Villetti G, Imbimbo BP, Calza L, Giardino L. Multi-target action of the novel anti-Alzheimer compound CHF5074: in vivo study of long term treatment in Tg2576 mice. BMC Neurosci. 2013;14:44. doi: 10.1186/1471-2202-14-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Poli G, Corda E, Lucchini B, Puricelli M, Martino PA, Dall'ara P, Villetti G, Bareggi SR, Corona C, Costassa EV, Gazzuola P, Iulini B, Mazza M, Acutis P, Mantegazza P, Casalone C, Imbimbo BP. Therapeutic effect of CHF5074, a new gamma-secretase modulator, in a mouse model of scrapie. Prion. 2012;6:62–72. doi: 10.4161/pri.6.1.18317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Imbimbo BP, Frigerio E, Breda M, Fiorentini F, Fernandez M, Sivilia S, Giardino L, Calza L, Norris D, Casula D, Shenouda M. Pharmacokinetics and Pharmacodynamics of CHF5074 After Short-term Administration in Healthy Subjects. Alzheimer Dis Assoc Disord. 2012 doi: 10.1097/WAD.0b013e3182622ace. [DOI] [PubMed] [Google Scholar]
- 190.Moore BD, Chakrabarty P, Levites Y, Kukar TL, Baine AM, Moroni T, Ladd TB, Das P, Dickson DW, Golde TE. Overlapping profiles of Abeta peptides in the Alzheimer's disease and pathological aging brains. Alzheimers Res Ther. 2012;4:18. doi: 10.1186/alzrt121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Kuperstein I, Broersen K, Benilova I, Rozenski J, Jonckheere W, Debulpaep M, Vandersteen A, Segers-Nolten I, Van Der Werf K, Subramaniam V, Braeken D, Callewaert G, Bartic C, D'Hooge R, Martins IC, Rousseau F, Schymkowitz J, De Strooper B. Neurotoxicity of Alzheimer's disease Abeta peptides is induced by small changes in the Abeta42 to Abeta40 ratio. EMBO J. 2010;29:3408–3420. doi: 10.1038/emboj.2010.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.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. 2007;27:627–633. doi: 10.1523/JNEUROSCI.4849-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Wang R, Wang B, He W, Zheng H. Wild-type presenilin 1 protects against Alzheimer disease mutation-induced amyloid pathology. J Biol Chem. 2006;281:15330–15336. doi: 10.1074/jbc.M512574200. [DOI] [PubMed] [Google Scholar]
- 194.He W, Luistro L, Carvajal D, Smith M, Nevins T, Yin X, Cai J, Higgins B, Kolinsky K, Rizzo C, Packman K, Heimbrook D, Boylan JF. High tumor levels of IL6 and IL8 abrogate preclinical efficacy of the gamma-secretase inhibitor, RO4929097. Mol Oncol. 2011 doi: 10.1016/j.molonc.2011.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Luistro L, He W, Smith M, Packman K, Vilenchik M, Carvajal D, Roberts J, Cai J, Berkofsky-Fessler W, Hilton H, Linn M, Flohr A, Jakob-Rotne R, Jacobsen H, Glenn K, Heimbrook D, Boylan JF. Preclinical profile of a potent gamma-secretase inhibitor targeting notch signaling with in vivo efficacy and pharmacodynamic properties. Cancer Res. 2009;69:7672–7680. doi: 10.1158/0008-5472.CAN-09-1843. [DOI] [PMC free article] [PubMed] [Google Scholar]

