You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/51068164

??-Secretase Inhibitors and Modulators for the Treatment of Alzheimer's


Disease: Disappointments and Hopes

Article  in  Current topics in medicinal chemistry · April 2011


DOI: 10.2174/156802611795860942 · Source: PubMed

CITATIONS READS

124 450

2 authors:

Bruno P. Imbimbo Giuseppe A.M. Giardina


Chiesi Farmaceutici S.p.A. Istituto Italiano di Tecnologia
248 PUBLICATIONS   6,317 CITATIONS    117 PUBLICATIONS   1,439 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Mutated form of Human Nerve Growth Factor View project

GreatAGE Study: Aging in Population View project

All content following this page was uploaded by Bruno P. Imbimbo on 24 March 2014.

The user has requested enhancement of the downloaded file.


Current Topics in Medicinal Chemistry, 2011, 11, 1555-1570 1555

-Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s


Disease: Disappointments and Hopes

Bruno P. Imbimbo1,* and Giuseppe A.M. Giardina2

1
Research & Development Department, Chiesi Farmaceutici, Parma and 2NiKem Research, Baranzate, Italy

Abstract: According to the -amyloid (A) hypothesis, compounds that inhibit or modulate secretase, the pivotal en-
zyme that generates A, are potential therapeutics for Alzheimer’s disease (AD). Studies in both transgenic and non-
transgenic animal models of AD have indicated that secretase inhibitors, administered by the oral route, are able to
lower brain A concentrations. However, scanty data are available on the effects of these compounds on brain A deposi-
tion after chronic administration. Behavioral studies are also scarce with only one study indicating positive cognitive ef-
fects of a peptidomimetic compound acutely administered (DAPT). -Secretase inhibitors may cause abnormalities in the
gastrointestinal tract, thymus, spleen and skin in experimental animals and in man. These toxic effects are likely due to
inhibition of Notch cleavage, a transmembrane receptor involved in regulating cell-fate decisions. Some non-steroidal
anti-inflammatory drugs (NSAIDs) and other small organic molecules have been found to modulate secretase shifting
its cleavage activity from longer to shorter -amyloid species without affecting Notch cleavage. Long-term histopa-
thological and behavioral animal studies are available with these NSAIDs (mainly ibuprofen) but it is unclear if the ob-
served in vivo effects on A brain pathology and learning depend on their activity on -secretase or on other biological
targets. The most studied -secretase inhibitor, semagacestat (LY-450139), was shown to dose-dependently decrease the
generation of A in the cerebrospinal fluid of healthy humans. Unfortuantely, two large Phase 3 clinical trials of sema-
gacestat in mild-to-moderate AD patients were prematurely interrupted because of the observation of detrimental effects
on cognition and functionality in patients receiving the drug compared to those receiving placebo. These detrimental ef-
fects were mainly ascribed to the inhibition of Notch processing and to the accumulation of the neurotoxic precursor of
A (the carboxy-terminal fragment of APP, or CTF) resulting from the block of the -secretase cleavage activity on
APP. Two large Phase 3 studies in mild AD patients with tarenflurbil (R-flurbiprofen), a putative -secretase modulator,
were also completely negative. The failure of tarenflurbil was ascribed to low potency and brain penetration. New Notch-
sparing -secretase inhbitors and more potent, more brain penetrant -secretase modulators are being developed with the
hope of overcoming the previous setbacks.
Keywords: -secretase inhibitors, -secretase modulators, -amyloid, alzheimer’s disease.

INTRODUCTION disease. This hypothesis was conceptualized in 1991 by


Hardy and Allsop [1]. The updated version of this theory
Alzheimer’s disease (AD) is the most common cause of
says that the oligomeric forms of A1-42 are the main cause
dementia. Neuritic plaques, the characteristic lesions found of neuronal death in AD [2,3].
in the brain of AD patients, are composed mainly of aggre-
gates of a peptide with 40 or 42 amino acid residues known -Secretase is formed by at least four proteins: presenilin
as -amyloid (A) Fig. (1). The A peptide is the result of (PS), nicastrin, anterior pharynx-defective 1 (Aph-1) and
the metabolic processing of a complex transmembrane gly- presenilin enhancer 2 (Pen-2). Presenilins are of exceptional
coprotein known as amyloid precursor protein (APP). APP pathophysiological importance since more than 150 autoso-
may be metabolically processed according to two pathways mal dominant point mutations are known in these proteins,
Fig. (1). In the so-called non-amyloidogenic pathway, the - all of which cause aggressive early-onset AD. These muta-
secretase enzyme cleaves APP within the A sequence and tions result in increased production of A142, the highly self-
releases extracellularly its soluble N-terminal fragment, aggregating and neurotoxic form of A, or an increased
sAPP, that appears to exert neuroprotective activity. In the A1-42 to A1-40 ratio. Thus, the inhibition of the catalytic
amyloidogenic pathway, the -secretase enzyme releases unit (presenilin) of the -secretase enzymatic complex ap-
sAPP plus a 12 kDa protein fragment (C99 or CTF), pears to be a logical strategy for contrasting A accumula-
which in turn is cleaved by the -secretase enzyme giving tion in the brain of AD patients and indeed many potent,
way to A. The correlation among A histopathologic orally active and brain penetrant compounds have been syn-
lesions, brain cell death and cognitive deficiency in AD thesized and developed [4]. Unfortunately, -secretase
represents the so called “A cascade hypothesis” of the cleaves many other important substrates other than APP [5],
one of the most relevant from toxicological point of view
being Notch. Thus, -secretase inhibitors block the process-
*Address correspondence to this author at the Research & Development
Department, Chiesi Farmaceutici, Via Palermo 26/A, 43100 Parma, Italy;
ing of other proteins and this is the main cause of toxicity in
Tel: +39 0521 279278; Fax:+39 0521 279579; preclinical testing and represents a major source of concern
E-mail: b.imbimbo@chiesigroup.com in clinical trials. The discovery that some non-steroidal anti-

1568-0266/11 $58.00+.00 © 2011 Bentham Science Publishers Ltd.


1556 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

Fig. (1). Schematic overview of APP processing. The top panel shows the APP region comprising the transmembrane sequence (underlined,
bold) and the sequences of A1-40 (D1-V40) and A1-42 (D1-A42) peptides. The -secretase cleaves at D1 and Y10. The -secretase cleaves at
Lys16, and the -secretase cleaves at Val40 and/or Ala42. Below the sequence is a representation of APP with the residue numbers of interest
in - and -secretase cleavage. Panel A represents the non-amyloidogenic -secretase pathway in which sAPP and C83 are generated. Sub-
sequent hydrolysis by the -secretase produces a p3 peptide that does not form amyloid deposits. Panel B represents the amyloidogenic path-
way in which cleavage of APP by the -secretase to liberate sAPP and C99 is followed by -secretase processing to release -amyloid pep-
tides (A1-40 and A1-42) found in plaque deposits (from John et al, 2003).

inflammatory drugs (NSAIDs) may modulate the cleavage olytic event. Nicastrin is a 709- amino acid type 1 membrane
activity of -secretase on APP without interfering with the glycoprotein with a large ectodomain that may act
cleavage of other substrates [6], have lead to intensive efforts
in designing -secreatase modulators that appear much more
attractive from safety point of view than traditional -
secretase inhibitors [7]. This article reviews the profile of -
secretase inhibitors and modulators that have reached the
clinic and will discuss the pharmacological and clinical is-
sues of this new class of anti-AD compounds both in terms
of safety and efficacy.
THE STRUCTURE OF THE -SECRETASE COM-
PLEX
-Secretase is an unusual aspartyl protease that cleaves its
substrates within the transmembrane region. This enzyme
was shown to consist of four protein components: presenilin
(PS) 1 or 2 (which contains the catalytic domain), nicastrin
(which may serve to dock substrates), Aph-1 (anterior phar-
ynx-defective 1), and Pen-2 (presenilin enhancer-2) in a
1:1:1:1 ratio Fig. (2) [8]. Presenilin-1 (PS-1) and preseneni-
lin-2 (PS-2) are 476- and 448-amino acid proteins consisting
of nine transmembrane domains (TMDs) and are endoprote-
olytically cleaved into a 30 kDa N-terminal fragment and a
20 kDa C-terminal fragment, each of which contains an
Fig. (2). Schematic structure of the -secretase enzymatic complex
aspartate active site. The cleavage occurs in the large cyto-
and of its processing of the C99 substrate (nicastrin = green, Aph-1
plasmic loop between TMD6 and TMD7 within a short hy-
= blue, Pen-2 = red, presenilin N-terminal = yellow, presenilin C-
drophobic domain that is believed to dive into the mem-
terminal = light blue) (from Kreft et al, 2009).
brane. This endoproteolysis is believed to be an autoprote-
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1557

as a -secretase substrate receptor. It is hypothesized that the cognitive or behavioral effects of the drug in animal models
free N-terminus of the -secretase substrate(s) first binds to of AD [20]. The lack of cognitive effects of semagacestat in
the ectodomain of nicastrin, which may facilitate its interac- animals could be linked to the fact that the drug has neuro-
tion with the docking site on PS which is followed by reloca- toxic effects in vivo. A study employing in vivo two-photon
tion to the active site on PS where it is cleaved. Pen-2 is the imaging showed that dendritic spines get irreversibly lost in
smallest component (10 kDa) of -secretase and is thought the cerebral cortex of wild-type mice after only 4 days of
to be required for the stabilization of the PS fragments in the treatment with semagacestat (30 mg/kg s.c.). The same ex-
-secretase complex. Aph-1 is a seven-TMD protein ( 20 periments carried out in APP-deficient mice suggested that
kDa) whose function in -secretase is currently unclear. It APP-cleavage products (probably an accumulation of C-
has been found that Aph-1 can exist as two splice variants: terminal fragments), are critically involved [21].
Aph-1a and Aph-1b [9]. Thus, several different -secretase Many studies with semagacestat have been conducted in
complexes are possible depending on whether they contain man. A Phase 1 study evaluated the safety and tolerability
PS-1 or PS-2 and whether they contain Aph-1a or Aph-1b. It and biomarker responses to single oral doses of 60, 100, or
has been proposed that nicastrin and Aph-1 form an initial 140 mg in 31 healthy male and female volunteers (40 years
complex that sequentially adds PS and Pen-2. The first high and older) [22]. No clinically significant adverse events or
resolution (12 Å) structure of -secretase have been recently laboratory changes were observed in this study. A dose-
obtained [10] and revealed a globular structure with a num- proportional increase in drug exposure was observed in
ber of cavities that are open to either the extracellular space plasma and in CSF with an estimated CNS penetration of
or the cytosol as well as an almost continuous surface groove 8%. Peak drug plasma levels occurred at 1 h and then de-
at the membrane region that could be a substrate entry site.
clined with a half-life of 2.5 h. A dose-dependent decrease in
plasma A1-40 levels was also demonstrated with maximum
-SECRETASE INHBITORS THAT HAVE REACHED inhibition (-73%) at 6 h after the administration of the 140-
CLINICAL DEVELOPMENT mg dose. A rebound effect on plasma A1-40 levels was ob-
In 2001 Eli Lilly firstly reported the in vivo inhibition of served at 8-12 hours after administration and lasted for at
brain A with DAPT, a peptidomimetic -secretase inhibitor least the 24 h. Cerebrospinal fluid concentrations of A were
[11]. Several other non-peptidic, orally available, secretase unchanged 4 hours after drug administration. In a second
inhibitors have been synthesized [12]. Nevertheless, data on Phase 1 study, LY-450139 was administered to 37 healthy
the cognitive effects of single and prolonged administration men and women (45 years and older) for up to 14 days at
of -secretase inhibitors in animal models of AD are scanty, doses of 5, 20, 40 and 50 mg once daily [23]. Two subjects
the only published study being one on DAPT [13]. In this in the 50-mg dose group developed possibly drug-related ad-
study, single oral doses of DAPT (100 mg/kg) reversed the verse events and discontinued treatment. The first subject had
contextual fear-conditioning deficit of Tg2576 mice only significant increases in serum amylase and lipase and com-
when administered before training. Peak inhibitions of about plained of moderate abdominal pain. The other subject re-
30% in soluble brain A140 and A142 levels were measured ported diarrhea that was positive for occult blood. The
at 4 and 8 hours, respectively, after dosing. plasma half-life of LY-450139 was found to be approxi-
Historically, the first -secretase inhibitor publicly re- mately 2.5 h and peak plasma concentrations were achieved
ported to reach the clinic is a compound synthesized at Bris- about 1 hour after administration. The 50-mg dose caused a
tol-Myers Squibb (BMS-299897). The drug was later aban- maximal 40% reduction in total plasma A that returned to
doned. At least six other secretase inhibitors (LY-450139, baseline within 8 hours. After returning to baseline, plasma
PF-3084014, MK-0752, BMS-708163, GSI-953 and A levels increased to about 300% of baseline values at 15
ELND006) reached the clinic (Table 1). For only one com- hours before slowly declining again. At lower doses, smaller
pound (LY-450139) clinical data have been fully published. and shorter decreases in plasma A were observed, although
Most of the information on the other compounds derives the subsequent plasma A increases were similar. No signifi-
from congress communications. cant changes in cerebrospinal fluid A levels were detected.
LY-450139 has been extensively evaluated in AD pa-
LY-450139 (Semagacestat) tients. In a first randomized, placebo-controlled trial, 70 pa-
The benzoazepinone derivative LY-450139 Fig. (3) is a tients received the drug for 6 weeks (30 mg once a day for 1
-secretase inhibitor developed at Eli-Lilly. LY-450139 is week followed by 40 mg once a day for 5 weeks) [24]. Six
only 3-fold selective in inhibiting APP and Notch cleavage patients taking LY-450139 reported diarrhea. A 76-year-old
(APP IC50 = 15 nM, Notch EC50 = 49 nM) [14]. LY-450139 man on LY-450139 had gastrointestinal bleeding associated
is the best documented -secretase inhibitor that has reached to a Barrett esophagus, a clinical condition characterized by
clinical testing and for which clinical studies have been fully the conversion of normal squamous cells into abnormal spe-
published. In experimental animals, the effects of LY- cialized columnar cells. Approximately 4 months after dis-
450139 on A levels in brain, cerebrospinal fluid (CSF) and continuing treatment, the patient developed endocarditis and
plasma were well characterized in transgenic mice [15], non- approximately 1 month thereafter, died. In the LY-450139-
transgenic mice [16], guinea pigs [17] and dogs [18]. Never- treated group, circulating CD69, T lymphocytes, eosinophils,
theless, the drug failed to show a statistically significant ef- and serum concentrations of potassium and inorganic phos-
fect on brain plaque deposition in chronic studies in trans- phorus showed statistically significant changes, although
genic mice expressing mutated human APPV717F (PDAPP these findings were reported as “clinically irrelevant”.
mice) [19]. More importantly, no data are available on the Plasma A1-40 concentrations of patients taking the com-
1558 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

Table 1. -Secretase Inhibitors that have Reached Clinical Development for the Treatment of Alzheimer’s Disease (see Figure 3 for
Chemical Structures)

Compound Main Refer-


Pros Cons Development Company
Status ences

Worsens dose-dependly cognitive and


functional decay of AD patients.
Decreases production of newly syn- Increases skin cancer in AD patients.
Semagacestat
thesized A in the CSF of healthy Is neurotoxic (reduced dendritic spine Discontinued Eli-Lilly [21-27]
(LY-450139)
humans density) in mice. Lack of data on
behavioral effects in animal models
of AD.

Notch sparing.
Lack of data on brain plaque deposi-
Good brain penetration.
tion in transgenic mice
Long-lasting effects on A levels in
Lack of data on behavioral effects in Discontinued
PF-3084014 animals. Pfizer [38]
animal models of AD. for AD
No rebound effect on plasma
Unfavourable PK/PD profile. In hu-
A in animals
mans

Decreases A1 -40 levels in CSF of Inhibits Notch cleavage. Significant Discontinued
MK-0752 Merck [29]
healthy volunteers gastro-intestinal toxicity in humans for AD

Lack of data on brain plaque deposi-


Notch sparing. tion in transgenic mice. Bristol
BMS-708163 Decreases A levels in CSF of Lack of data on behavioral effects in Phase 2 Myers [32,34,35]
healthy volunteers animal models of AD Squibb
Poor tolerability in AD patients

Lack of data on brain plaque deposi-


Notch sparing.
tion in transgenic mice.
Begacestat Good brain penetration
Does not decrease A levels in CSF Phase 2 Wyeth [39,40]
(GSI-953) Improves memory in a transgenic
of AD patients. Causes rebounds on
mouse model of AD
plasma A40 levels in man

Notch sparing. Produces late rebounds in plasma


Good brain penetration A in animals.
ELND-006 Phase 1 Elan [44]
Decrease brain A burden in trans- Lack of data on behavioral effects in
genic mice animal models of AD.

F
OH O
H H H
N N N
N N N
N
H Me H
O Me O F O N

Semagacestat (LY-450139) PF-3084014

CF3

CF3 O O
CF3 H2N S
N
OH O Cl
NH
Cl S S
N
O O F O
N
Begacestat (GSI-953) BMS-708163

MK-0752 (no structure disclosed) ELND-006 (no structure disclosed)

Fig. (3). Chemical structure of -secretase inhibitors that have reached clinical development.
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1559

pound decreased significantly by 38% compared to baseline. po, qd, titrated to 140 mg po, qd) for 21 months, with the
The lowest concentration was achieved approximately 3 option of enrolling in an open-label extension trial for further
hours after administration of a 40-mg dose and returned to treatment. Patients taking symptomatic treatments for AD
baseline approximately 7 hours postdose. A140 concentra- were permitted to continue treatment. The trial also incorpo-
tions in cerebrospinal fluid did not decrease significantly. rated a randomized delayed start design, similar to IDEN-
Another study evaluated the safety, tolerability, and A TITY. The primary outcome measures of efficacy were the
ADAS-Cog scale for cognition and the ADCS-ADL scale for
response to LY-450139 in 51 AD patients treated for 14
functionality. Secondary endpoints include other dementia
weeks [25]. Patients were randomized to receive placebo (n
rating scales, A levels in plasma and CSF and other brain
= 15) or LY-450139 (n = 36). Patients on LY-450139 re-
biomarkers determined by neuroimaging. The dose titration
ceived 60 mg/day for 2 weeks, then 100 mg/day for 6 weeks,
based on patient tolerability was designed to provide a more
and then either 100 or 140 mg/day for 6 additional weeks.
Forty-three patients completed the study. There were 7 cases 'real-world simulation' of semagacestat. Eli Lilly estimated
the trial would be completed by June 2012 [20]. Unfor-
of skin rashes and 3 reports of hair color change in the drug
tunetly, on August 17, 2010, Eli Lilly announced that the
treatment groups. There were 3 adverse event-related discon-
preliminary results from the two Phase 3 trials showed se-
tinuations, including 1 transient bowel obstruction. Com-
magacestat worsened clinical measures of cognition and the
pared to placebo, A140 plasma concentrations were reduced
ability to perform activities of daily living compared to pla-
by 58% in the 100-mg group and 65% in the 140-mg group.
No significant reduction was seen in cerebrospinal fluid A cebo. In addition, data showed semagacestat was associated
with an increased risk of skin cancer compared with those
levels. No differences were seen in cognitive or functional
who received placebo. The detrimental effects of semagaces-
measures between placebo-and LY-450139-treated patients.
tat appeared to be dose-dependent [27]. These negative find-
The effects of semagacestat (100, 140 and 280 mg) on ings lead the company to interrupt the development of sema-
A synthesis and clearance in CSF of 20 healthy volunteers gacestat, although the two studies are still ongoing in double-
have been investigated with a relatively new technique, blind conditions to follow up the cognitive and clinical con-
based on the intravenous administration of a stable isotope- ditions of the patients [27].
labeled aminoacid (13C6-leucine) [26]. CSF was collected
hourly with a lumbar catheter. A dose-dependent decrease in MK-0752
levels of newly-generated A was observed; this was statis-
tically significant at all doses versus placebo. The mean de- Merck is developing a -secretase inhibitor (MK-0752,
crease in A generation was 47, 52 and 84% over a 12-h no structure disclosed) that does not distinguish between
period with doses of 100, 140 and 280 mg, respectively. APP and Notch. Doses of 30 or 60 mg/kg orally to rhesus
There was a significant inverse relationship between sema- monkeys produced high plasma levels (Cmax of 32 or 88 M
gacestat concentrations in the CSF and the amount of newly and an AUC of 477 or 858 Mh, respectively) but unfortu-
generated A in CSF (r = -0.646; p = 0.002). Furthermore, nately spleen and ileum toxicity due to inhibition of Notch
the AUC of CSF A (measured by ELISA) tended to de- signalling [28]. A Phase 1 study evaluated the safety, toler-
crease in this time period, with significance from placebo ability, pharmacokinetics and pharmacodynamics of single
achieved in the 280-mg group (48.2% decrease). Interest- oral doses (from 110 to 1000 mg) of MK-0752 in 27 healthy
ingly, there was a rebound effect on CSF A142 concentra- young men [29]. The drug was generally well tolerated.
tions at later times (24 to 36 h); compared with placebo, a 2- Drug plasma levels increased proportionally to the dose,
fold increase was observed at 30 h with the 280-mg dose. peaked at 3-4 hours and then declined with a half-life of
Clearance of A was unaffected by semagacestat. about 20 hours. MK-0752 CSF levels were similar to un-
bound plasma concentrations, suggesting a good penetration
In April 2008, Eli Lilly has initiated its first Phase 3 of the drug into the CNS. MK-0752 Doses of 500 mg sig-
clinical trial (IDENTITY) of semagacestat in about 1,500 nificantly inhibited for 12 hours A1-40 concentrations in
mild to moderate AD patients with the hope of slowing dis- cerebrospinal fluid with a peak inhibitory effect of 35%. Af-
ease progression. Patients would be treated with semagaces- ter 1000 mg, cerebrospinal fluid A140 inhibition was sus-
tat (100 or 140 mg po, qd) for 21 months, with the option of tained over 24 hours. Plasma A1-40 concentrations also
enrolling in an open-label extension trial for further treat- showed a dose-dependent decrease but were followed by a
ment. Patients taking symptomatic treatments for AD were later rebound over baseline levels.
permitted to continue treatment. The trial incorporated a
'randomized delayed start' design, which means that patients Although MK-0752 was initially developed as a treat-
initially assigned to the placebo arm will be administered ment for AD, in mid 2005, Phase 1 trials in patients with
semagacestat sometime before the end of the 21-month pe- advanced breast cancer and in patients with T-cell acute
riod to assess the effects on disease progression. The primary lymphoblastic leukemia (T-ALL) were initiated and results
outcome measures of efficacy were the ADAS-Cog scale for were presented in June 2006 [30]. Indeed, there is increasing
cognition and the ADCS-ADL scale for functionality. Sec- interest in the applicability of -secretase inhibitors to the
ondary endpoints include A levels in plasma and CSF and treatment of cancer. MK-0752 has been shown to inhibit -
other brain biomarkers determined by neuroimaging [20]. Eli secretase-mediated cleavage of Notch with an IC50 of 55 nM.
Lilly initially estimated the trial would be completed by Increasing evidence implicates the Notch pathway in normal
March 2012. A second Phase 3 trial, called IDENTITY-2, T-cell lymphopoiesis and the pathogenesis of several human
was also started in about 1,100 patients with mild to moder- malignancies. Prolonged activation of the Notch signal
ate AD. Patients would be treated with semagacestat (60 mg transduction pathway occurs in more than 50% of patients
1560 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

with T-cell acute lymphoblastic leukemias and is important tients with prodromal AD was started in May 2009 and is
in the pathogenesis of the disease. Preclinical studies indi- still recruiting patients (ClinicalTrials.gov Identifier
cate that pharmacologic inhibition of -secretase activity NCT00890890). Initially, the study was planned to last 52
suppresses T-cell acute lymphoblastic leukemias cell growth weeks and later was amended to prolong treatment to 104
and induces apoptosis by preventing cleavage of Notch, thus weeks. Dosing regimens were initially 50, 100 and 125
preventing prolonged activation in downstream pathways. mg/day. Later due to tolerability problems, the two upper
Unfortunately, studies with MK-0752 in pediatric and adult doses were discontinued.
patients with T-cell acute lymphoblastic leukemias and acute
myeloid leukemia reported that drug was associated with PF-3084014
gastrointestinal toxicity and fatigue without substantive
clinical activity [30]. The novel aminotetraline derivative PF-3084014 Fig. (3)
is a potent, Notch-sparing, -secretase inhibitor in develop-
An intermittent dosing schedule appears to reduce toxic- ment at Pfizer. In a cell-free assay, PF-3084014 appears to
ity while demonstrating adequate target inhibition, warrant- be a non-competitive but reversible inhibitor of human -
ing further evaluation [31]. In February 2008, the Phase 1 secretase activity with an IC50 of 6.2 nM [36]. In a whole-
trial of MK-0752 in combination with docetaxel began in 30 cell assay, PF-3084014 displays an IC50 of 1.3 nM. In fetal
patients with locally advanced or metastatic breast cancer. thymus organ culture assay, PF-3084014 appears to be a
The primary outcome was dose limiting toxicity. At that time weak inhibitor of Notch signaling with an IC50 of 1915 nM.
the estimated study completion date was March 2012. In The APP to Notch selectivity ratio is 1,473. In guinea-pigs,
May 2008, an open-label, uncontrolled pilot study was initi- dose-response inhibition of total A levels was observed in
ated in 20 women with early stage, estrogen receptor positive plasma, CSF and brain after subcutaneous administration
breast cancer to establish the safety and tolerability of MK- (0.03-10 mg/kg). At the highest dose (10 mg/kg), A levels
0752 in the pre-surgical setting in combination with ta- were reduced by 70% in brain and plasma, and by 50% in
moxifen or letrozole. The study was expected to be com- CSF, which was maintained at 30 hours post-dose. No late
pleted in late 2010. rebound effects on plasma A were observed. Drug levels in
the brain were similar to that measured in plasma. Studies in
BMS-708163 young (plaque-free) Tg2576 transgenic mice showed that
brain, CSF, and plasma levels of A were inhibited dose-
The benzene sulfonamide BMS-708163 Fig. (3), in
dependently following doses of 1 to 18 mg/kg. At the highest
which an oxadiazolylbenzyl group is attached to the sul-
dose (18 mg/kg), A levels were reduced by 78% in brain,
fonamido nitrogen, is a potent, Notch-sparing, -secretase
inhibitor in development at Bristol-Myers Squibb. In vitro, 72% in CSF, and 92% in plasma. A1-40 was most potently
inhibited in all compartments. A1-42 showed approximately
the drug shows a 193-fold selectivity versus Notch cleavage
20% less reduction than A1-40 in all compartments [36].
(A1-40 IC50 = 0.3 nM and Notch EC50 = 58 nM) [32]. Stud-
However, dose-dependent increases in A11-40 and A1-43
ies in rats have shown that BMS-708163 dose-dependently
were seen at doses that potently inhibited A1-40 and A1-42.
decreases CSF and brain A1-40 levels without causing
In addition, PF-3084014, like previously described gamma-
Notch-related gastrointestinal and lymphoid toxicity [33]. In
dogs receiving 2 mg/kg orally a decrease in brain and CSF secretase inhibitors, preferentially reduced A1-40 relative to
A1-42 [36].
A1-40 levels were observed for at least 24 hours and peak
inhibition around 75% in CSF and 50% in the cortex [32]. In healthy volunteers enrolled in a Phase 1 study, single
doses were associated with mild and self-limiting adverse
Studies in healthy young subjects have indicated that
events, no apparent clinically relevant changes in vital signs
BMS-708163 is well tolerated up to 400 mg after single ad-
ministration and up to 150 mg/day after multiple doses for and no significant ECG trends [37]. Doses of 1-120 mg were
safe and well tolerated, and the maximum tolerated dose was
28 days [33]. After oral administration, BMS-708163 ap-
not identified. Mean pharmacokinetic variables with the sin-
pears to be quickly absorbed (Tmax = 1-2 h), to produce sys-
gle 120 mg dose included an oral clearance of approximately
temic exposure proportional to the dose (up to 200 mg) and
16 mL/min/kg, an half-life of approximately 19 hours, a
to be slowly eliminated (terminal half-life  40 h). The ef-
mean Cmax of approximately 21 nM and an average steady-
fects of BMS-708163 in CSF A levels in humans were
evaluated after both single and multiple oral doses. Twelve state concentration of approximately 4 nM [37]. However, an
analysis of pharmacokinetic/pharmacodynamic data from a
hours after a single administration, doses of 200 and 400 mg
study of multiple oral dosing in humans led to the decision to
to young subjects produced 37% and 40% reductions, re-
end development of the compound for AD [38]. Plasma drug
spectively in CSF A140 levels versus baseline. The corre-
and A concentrations collected from 18 healthy volunteers
sponding inhibitory values for CSF A142 levels were 32%
given 40 or 90 mg dose once daily for 14 days were used to
and 34%, respectively. Exposure for BMS-708163 in CSF
was much lower (< 1%) than in plasma [34]. After multiple model the relationship between these concentrations. Phar-
macokinetic-pharmacodynamic profiles for these doses were
administrations for 4 weeks (50-150 mg/day), steady-state
used to extrapolate the profile at higher doses. This popula-
trough CSF A140 levels were reduced dose-dependently
tion pharmacokinetic-pharmacodynamic analysis yielded the
compared to baseline values [35]. A 24-week dose-range
finding that exposure levels needed to reach the prespecified
(25, 50, 100 and 125 mg/day) finding Phase 2 study in 209
area above the effect curve target for plasma inhibition of
mild-to-moderate AD patients has been completed in June
2010 but data have not been released (ClincalTrials.gov A140 on Day 14 relative to Day 1 were 2- to 3-fold higher
than exposure limits based on animal toxicology data. As a
Identifier NCT00810147). A 104-week Phase 2 study in pa-
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1561

result, much higher doses than those previously used would line were similar to those of A1-40 in magnitude and dura-
be need to attain a high probability of technical success [38]. tion. No significant effects of the drug on CSF A1-40 levels
PF-3084014 has now entered clinical study as an anti- were observed in either AD patients or young volunteers
[40].
cancer agent. A Phase 1 study in patients with advanced
solid tumors and leukemia, with an estimated enrollment of A translational medicine study was subsequently per-
60, is currently recruiting patients (ClinicalTrials.gov Identi- formed on begacestat, comparing pharmacokinetic/ pharma-
fier NCT00878189). codynamic biomarker relationships in Tg2576 mice and hu-
mans [41]. It was found that a 10 mg/kg dose in Tg2576
Begacestat (GSI-953) mice produces different drug exposures and inhibitiory ef-
fects on  in plasma (AUC0-3 = 5951 ng·h/mL, 9-20% 
The thiophene sulfonamide GSI-953 Fig. (3) is a potent inhibition), brain (AUC0-3 = 9338 ng·h/mL, 22-33%  inhi-
-secretase inhibitor in development at Weyth. This com- bition) and CSF (AUC0-3 = 350 ng·h/mL, no  inhibition).
pound inhibits A production with low nM potency in both In AD patients receiving a 450 mg dose, drug exposures and
cellular (1-40 EC50 = 14.8 nM, 1-42 EC50 = 12.4 nM) and  inhibitory effects in plasma (AUC0-3 = 2334 ng·h/mL,
cell-free (IC50 = 8 nM) assays [39]]. The compound deter- 28%  inhibition) and CSF (AUC0-3 = 240 ng·h/mL, no 
mines CTF elevation in cellular assay with an EC50 = 6.6. inhibition) were correlated with the Tg2576 results and sug-
nM. Cellular assays of Notch cleavage reveal that this com- gested that the exposure in human brain would lead to AUC0-
pound is 17-fold selective for the inhibition of APP cleavage
3 = 2400 ng·h/mL, which should produce brain  inhibition
(Notch EC50 = 208.5 nM) [39]. In Tg2576 mice, a 100 mg/kg similar to what was observed in plasma. The apparent dis-
doses of GSI-953 markedly reduced A1-40 and A1-42 levels connect between CSF and brain  lowering in Tg2576
in both CSF (maximum  90% inhibition) and in brain mice may be explained by the lower drug levels observed in
(maximum  60% inhibition). At 30 mg/kg, brain A levels CSF compared to brain [41].
were significantly reduced for 24 h with maximum effects
between 4 and 6 h. The minimal efficacious doses were 1 No clinical trials of begacestat in AD patients are pres-
mg/kg on brain A1-40 and 2.5 mg/kg on brain A1-42. Impor- ently registered in ClinicalTrials.gov web site.
tantly, this compound has been reported to attenuate dose-
dependently contextual memory deficit in Tg2576 transgenic ELND-006
mice with the effect being significant at 10 mg/kg [39]. In ELND-006 is a -secretase inhibitor in development at
mice, the drug showed a very good blood-brain barrier pene-
Elan Pharmaceuticals. The chemical structure of ELND-006
tration with brain-to-plasma exposure ratios ranging from 1.1
is still undisclosed. Like BMS-708163, ELN-006 is claimed
to 1.3. Interestingly, CSF drug levels were lower than those
to be a “APP-selective” -secretase inhibitor. These com-
measured in plasma (5-10%) [39]. No behavioral studies are
pounds are thought to interact with -secretase outside the
available on begacestat.
catalytic site, but the precise binding site and mode of action
In a first-in-man study in healthy human volunteers, oral is not known. The potency and in vivo characterization of the
administration of single doses of begacestat (3-600 mg) pro- agent after oral administration was recently described. In
duced dose-dependent transient reductions in plasma A1- cellular and enzymatic assays, ELND-006 displayed selectiv-
40 levels with maximum inhibition of 40% with 600 mg at 2 ity for APP, inhibiting APP and Notch cleavage with IC50
h. Rebounds in plasma A1-40 levels were visible at later values of 0.34 and 5.3 nM, respectively, and inhibiting A
times [39]. Other studies have described the pharmacoki- production and Notch signaling in cells with IC50 values of
netic, pharmacodynamic and tolerability profile of GSI-953 1.1 and 81.6 nM, respectively [42]. Brain/plasma ratios ex-
after single oral administration in young subjects and in AD ceeded 1 in both rodents and non-human primates, indicating
patients [40]. GSI-953 was well-tolerated and no dose- a very good brain penetration [42, 43]. After oral administra-
limiting adverse events were observed. Begacestat was rap- tion (0.3-30 mg/kg) to wild-type mice, PDAPP mice, wild-
idly absorbed in both young subjects and AD patients (Tmax = type rats or wild-type guinea pigs, ELND-006 was associ-
1-2 h). Drug plasma levels increased proportionally to the ated with significant reductions in CSF A [42]. In cynomol-
dose. Plasma elimination half-life was also similar in the two gus monkeys, an oral dose regimen of 0.3 mg/kg/ day for 13
populations (7-8 h). Drug concentrations in CSF were 10- weeks produced a decrease of brain A levels of at least 25%
fold lower than in plasma in both young and AD subjects for approximately 24 h [43]. ELND-006 concentrations in
[40]. For both young healthy subjects and AD patients, a plasma needed to reduce A in brain were consistent across
biphasic pattern of plasma A140 concentrations was ob- species. Similarly to other -secretase inhibitors, ELND-006
served, with an initial reduction below baseline for approxi- administration determined late rebounds in plasma A levels
mately 4 hours, followed by a second phase of increased in both rodent and non-human primate [42, 43]. Studies in
concentrations above baseline lasting up to 48 h before re- PDAPP transgenic mice indicated that treatment with 12.5
turning to baseline. Maximum inhibition in plasma A140 mg/kg/day for 13 weeks significantly reduced hippocampal
concentrations was observed at 2 h with a 28% reduction in amyloid burden and brain A levels but not of dystrophic
AD subjects and 33% reduction in young subjects. Notably, neurites [44]. More prolonged treatment with the same dose
initial reductions in plasma concentrations were less pro- significantly reduced both plaque burden and dystrophic
nounced for A142 than A140. Maximum reductions in neuritis but not brain A levels measured by Elisa [44]. Ac-
plasma A142 were only 7% in AD subjects and 17% in cording to Elan, ELND-006 is currently being evaluated in
young subjects. Subsequent increases in A142 above base- Phase 1 studies.
1562 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

-SECRETASE MODULATORS THAT HAVE REAC- studies have shown that tarenflurbil has quite low potency in
HED OR ARE APPROCHING CLINICAL DEVEL- inhibiting the secretion of A1-42 in vitro (IC50  250 M)
OPMENT [48] as well as a poor CNS penetration in rodents (cerebro-
spinal fluid to plasma ratio of 1.3%) [49] with inadequate
Based on the initial observation that some NSAIDs pref-
brain concentrations achieved in vivo ( 2 M) [50, 51]. The
erentially inhibit A1-42 secretion without affecting Notch promising pharmacological results obtained with tarenflurbil
processing [6], a new class of drugs called -secretase modu-
by the Mayo Clinic group after short-term administration in
lators with both NSAID-like and non-NSAID structure has
transgenic mice [50] have never been confirmed by other
been developed in the last 5 years [7]. At least three of these
groups [52,49]. The neuropathological and behavioral effects
-secretase modulators have entered clinical trials (tarenflur-
of tarenflurbil after chronic administration in the transgenic
bil, E2012, CHF5074) Table 2, Fig. (4). It has been proposed
mouse [51] are inconsistent and questionable from a meth-
that NSAID-like -secretase modulators interact with the odological point of view. In these studies [51], the control
substrate (APP) rather than with the enzyme (-secretase)
groups of three different experiments using animals of dif-
[45]. However, the Merck group has shown that tarenflurbil
ferent ages (12 and 19 months) were pooled. In addition,
and sulindac sulfide could non-competitively block binding
several animals from control and drug-treated groups were
of -secretase inhibitors to -secretase [46]. An additional
excluded from the analyses. The positive behavioral effects
level of complexity recently uncovered is that APP binds to
of tarenflurbil observed in two pooled experiments of 16-
-secretase as a dimer and must partially unwind for - week duration, were not coupled with significant lowering
secretase processing [47].
effects on A plaque burden or on A1-40 or A1-42 brain lev-
Tarenflurbil els [51]. The positive effects of the drug on A deposition in
a third experiment of only 2-week duration, were not cou-
Tarenflurbil Fig. (4) is a biphenylpropionic acid single pled with any behavioral effect [51].
isomer developed by Myriad Genetics. The drug is the R
entantiomer of flurbiprofen. Tarenflurbil has been developed In healthy elderly human subjects, a three-week treat-
with the rationale of maintaining the A1-42 lowering proper- ment with tarenflurbil (200, 400 or 800 mg b.i.d.) did not
ties of flurbiprofen while removing its anti-COX activity that modify A1-40 or A1-42 levels in CSF [53]. Overall, there
is ascribable only to the S entantiomer and thus reducing were no significant effects of the drug on plasma A levels,
gastro-intestinal toxicity of the original drug. Preclinical although an inverse relationship was found between peak

Table 2. -Secretase Modulators that have Reached or are Approaching Clinical Development for the Treatment of Alzheimer’s
Disease (see Figure 4 for Chemical Structures)

Compound Main
Pros Cons Development Company
Status References

Poor in vitro potency. Poor brain


Tarenflur-
Good safety profile in AD patients penetration Discontinued Myriad Genetics [54-56]
bil
Not effective in two Phase 3 trials

Lenticular opacity in rats Lack of data


Notch sparing
on brain plaque deposition in trans-
E2012 No accumulation of CTF No re- Discontinued Eisai [57,58,60]
genic mice.
bound effects on human plasma
A Lack of data on behavioral effects

Notch sparing
No accumulation of CTF Low micromolar inhibitory potency
CHF5074 Attenuates brain pathology in tg mice on A1-42 Phase 2 Chiesi [61-64]
Improves cognitve deficit in tg mice Limited brain penetration
Neuroprotective activity in vivo

Lack of data on brain plaque deposi- ClinicalTri-


Good safety margin in animals
E2212 tion in transgenic mice. Lack of data Phase 1 Eisai als.gov Identifier
High inhibitory potency on A142.
on behavioral effects NCT0122125

Notch sparing
Decreases brain A aggregates in
Cognitive effects in transgenic mice
EVP-15962 transgenic mice. Preclinical En Vivo [66,67]
occur at relatively high doses
Improves cognitve deficit in trans-
genic mice.

Nanomolar potency.
Neuro Genetic
NPG-328 Notch-sparing Lack of data on behavioral effects Preclinical [68]
Pharmaceuticals
Attenuates brain pathology in tg mice
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1563

O Me
Me
N

O N F
F HO N
O

Tarenflurbil (R-Flurbiprofen) Me E2012

N N
CF3
N
Cl
O N
N
Cl F HO N
O
CHF5074 Possible structure of E2212
Me

Cl O
CF3
O S
O
N
HO N H

N
N
O

Me
Possible structure of EVP-15962 NGP-328

Fig. (4). Chemical structure of -secretase modulators that have reached or are approaching clinical development.

tarenflurbil concentrations and A1-42 levels in plasma (r = mg b.i.d. (p = 0.042) but again, the decay in the placebo-
0.38, p = 0.016) suggesting that at concentrations around 180 treated patients (2.13 ± 0.28 points) was unexpectedly simi-
M, tarenflurbil may decrease A1-42 levels in plasma as lar to that observed in moderate patients (2.20 ± 0.58 points).
expected by its in vitro potency (IC50  250 M). Unfortu- The cognitive measure ADAS-Cog (Alzheimer’s Disease
nately, the drug reached much lower concentrations in CSF Assessment Scale Cognitive Subscale) showed the expected
samples collected 2-4 hours from the last dose (1.2 M after mean decays in mild and moderate patients treated with pla-
800 mg b.i.d.) and no significant effects were detected on cebo (4.02 ± 0.99 and 6.86 ± 1.79 points, respectively) and
CSF A1-42 levels even at peak levels [53]. It cannot be ex- no significant effects of tarenflurbil were observed. There
cluded that later CSF sampling could allow the detection of were no effects at all of tarenflurbil on MMSE (Mini-Mental
some effects of tarenflurbil on CSF A levels as occurred State Examination), another measure of cognition. Moder-
with traditional -secretase inhibitors [26]. ately affected patients who received the same dose of taren-
flurbil (800 mg b.i.d.) had highly significant greater clinical
From a clinical point of view, an accurate examination of
deterioration compared to placebo on the CDR-SB scale
the results of a 12-month Phase 2 study with tarenflurbil in
(4.63 ± 0.57 vs 2.20 ± 0.58 points, p = 0.003).
AD patients [54] reveals that the apparent positive effects of
the drug at a dose of 800 mg b.i.d. in mildly affected patients The detrimental effects of tarenflurbil on the global clini-
were likely due to an anomalous deterioration rate observed cal status of the AD patients were again observed in a 18-
in patients treated with placebo, rather than to the drug itself. month, placebo-controlled, Phase 3 study in 1,684 patients
On the ADCS-ADL (Alzheimer’s Disease Cooperative with mild AD [55]. Patients receiving 800 mg b.i.d. had a
Study Activities of Daily Living) scale, mild patients had a significantly higher deterioration than placebo on the CDR-
mean decay in 12 months of 8.57 ± 1.32 points on placebo vs SB scale at the end of the 18-month treatment period (p =
4.60 ± 1.30 points on tarenflurbil 800 mg b.i.d., the differ- 0.004). There were no significant differences in all the other
ence being statistically significant (p = 0.033). However, the cognitive, functional and behavioral variables between taren-
decay observed in the placebo-treated mild patients (8.57 ± flurbil and placebo. Another large 18-month Phase 3 study
1.32 points) was very similar to that observed in the moder- confimed that tarenflurbil is not effective in mild AD pa-
ately affected patients treated with placebo (8.31 ± 3.03 tients [56].
points). This is abnormal since it is expected that mildly af-
fected patients have less decline than moderately affected E2012
patients. On the CDR-SB (Clinical Dementia Rating Sum of
Boxes) scale, mild patients had a mean decay of 2.13 ± 0.28 The imidazylbenzyliden derivative E2012 Fig. (4) is a -
points on placebo vs 1.33 ± 0.28 points on tarenflurbil 800 secretase developed by Eisai in a partnership with TorreyPi-
1564 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

nes Therapeutics. E2012 represents the first -secretase impairment in the hippocampus, normalized synaptophysin
modulator with a non-NSAID structure entering clinical de- levels in the cortex and completely reversed contextual
velopment. In 2005, Eisai disclosed a novel chemical class memory deficit [63]. In October 2009 CHF5074 has been
of -secretase modulators characterized by the presence of an advanced into Phase 1 clinical trials and the results of a First-
arylimidazole moiety as exemplified by E2012. This chemi- in-Man study in 84 healthy mal subjects has been reported at
cal backbone has prompted intense research activity ICAD 2010 [64]. The maximum tolerated dose was found to
throughout the industry. In in vitro studies, MALDI-TOF be 800 mg. The pharmacokinetics of CHF5074 appeared
analysis showed that E2012 decreased A1-40 and A1-42 and linear in the studied dose range (25-1000 mg) with plasma
increased A1-38 without changing total A levels [57]]. exposure increasing in a predictable dose-proportional man-
E2012 did not induce the accumulation of APP-CTFs and ner [64]. A safety and tolerability mutiple ascending dose
did not inhibit Notch processing up to 3 M [57]. In rat cor- study has been completed (ClinicalTrials.gov Identifier:
tical neurons, E2012 inhibited A1-42 and A1-40 with an IC50 NCT01203384) and the drug has recently started Phase 2
of 92 and 330 nM, respectively [58]. In rats, a 30 mg/kg dose clinical development.
maximally decreased CSF and brain A1-42 levels by 47%
and 43%, respectively [58]. Treatment of dogs with E2012 E2212
(20 and 80 mg/kg) led to a dose-dependent increase in CSF
levels of A1-37, while A1-39, A1-40 and A1-42 decreased Eisai recently reported that E2012 will not be developed
[59]. In mid 2006, E2012 entered Phase 1 clinical develop- further in favor of an improved compound, E2212 whose
ment. In February 2007, the Phase 1 study was put on hold structure has not been disclosed [65]. Based on the analysis
because lenticular opacity was observed in a high-dose group of Eisai’s patent literature, E2212 could have a quite original
of a 13-week safety preclinical study in rats. At the time phenyldihydrobenzotriazole group replacing the benzylpyri-
when the study was suspended, no medical concerns were done moiety of E2012 in the right end side of the molecule
reported. Lenticular opacity was not observed in a later 13- and an imidazylpyridinyliden instead of the imidazylben-
week safety study in monkeys. An additional 13-week mul- zyliden group in the left end side Fig. (4). Indeed, the com-
tiple dosing study in rats did not reveal ocular toxicity and pany has published in the past few years several patent ap-
the clinical hold on E2012 was lifted in April 2008. The re- plications on interesting arylimidazole derivatives. E2212
sults of a Phase 1 study were recently reported [60]. Single was claimed to be more potent both in vitro and in vivo than
doses ranging from 2.5 to 400 mg were studied. Dose- E2012 and to have a wider safety margin [65]. An IND for
dependent reductions in plasma A1-42 and A1-40 levels were E2212 was submitted in November 2009, and a First-in-Man
observed with maximal inhibitions with the 400 mg dose of study was started in January 2010. On October 25, 2010 the
50% and 30%, respectively without a rebound effect [60]. study was still recruiting subjects (ClinicalTrials.gov Identi-
Eisai recently reported that E2012 will not be developed fier: NCT01221259).
further in favor of an improved compound, E2212.
EVP-15962
CHF5074 EVP-15962 is a -secretase modulator in development at
CHF5074 Fig. (4) is a novel dichloro substituted EnVivo Pharmaceuticals. The chemical structure of EVP-
biphenyl spirocyclopropyl acetic acid derivative in which the 15962 has not been disclosed, but based on analysis of En-
anti-cyclooxygenase activity has been removed and anti- Vivo’s patent literature a biphenyl cyclopropyl propionic
A1-42 secretory properties potentiated [49]. In human neuro- acid derivative structure could be hypothesized Fig. (4).
glioma cells overexpressing the Swedish mutant form of EVP-15962 could derive from the linear propionic acid de-
APP (H4swe), CHF5074 lowered A1-42 secretion with an rivatives like tarenflurbil or CHF5074 by shifting the distal
IC50 of 3.6 M [61]. At 5 M, no effects were observed on phenyl group from the para to the meta position while re-
Notch intracellular cleavage in human embryonic kidney 293 placing the steric hindrance in para with the lipophilic -
APPswe cells [61]. In 14-month-old Tg2576 mice expressing OCH2CF3 group. In rat cortical neurons, EVP-15962 inhib-
human mutated APP with point mutations at the -secretase ited A1-42 production (IC50 = 0.42 M) and increased A1-38
cleavage sites (K670N/M671 or “Swedish mutations”), secretion without affecting Notch signalling [66]. Similar
chronic treatment with CHF5074 in the diet (375 ppm for 4 effects on A levels were observed in wild-type C57/BL6
months) markedly reduced brain A burden (-52% in the mice and Sprague-Dawley rats following single oral admini-
cortex; -77% in the hippocampus) without any histological stration of the agent [66]]. Chronic administration of EVP-
sign of peripheral Notch-mediated toxicity [61]. In another 15962 as a diet supplement (equivalent to 20 or 60 mg/kg/
transgenic mouse model of AD (hAPPswe+lon), expressing day) to Tg2576 mice was associated to a significant decrease
human APP with mutations at both -secretase (K670N/ in the brain concentrations of A1-42 levels and to a reduction
M671) and -secretase (V717I or “London mutation”) cleav- of A aggregates [67]. Behavioral tests of cognitive ability
age sites, chronic treatment with CHF5074-supplemented were performed using the contextual fear conditioning and
diet (375 ppm for 6 months) decreased brain A plaques (- the Morris water maze paradigms. Sub-chronic treatment (
32% in the cortex; -42% in the hippocampus) as well as mi- 3 months) of young Tg2576 mice (4-5 months of age) at-
croglia activation (-54% in the cortex; -59% in the hippo- tenuated contextual memory at both 20 and 60 mg/kg/day
campus) [62]. The same treatment also attenuated spatial doses. On the other hand, chronic treatment with EVP-15962
memory deficit (-35%) evaluated with the Morris water ( 12 months) of young Tg2576 mice (4-6 months) up to the
maze test [62]. Long-term treatment of 6-month-old Tg2576 age of 16-18 months did not show significant improvements
mice with CHF5074 for 9 months mitigated neurogenesis of contextual memory at both doses. In the Morris water
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1565

maze test, a partial recovery/blockade of the cognitive deficit -secretase activity [19]. Indeed, an in vivo study indicated
of Tg2576 animals was seen following treatment with the 60 that semagacestat is neurotoxic in mice [21]. This study em-
mg/kg/day dose [67]. Intitial clinical testing of EVP-15962 is ployed in vivo two-photon imaging and showed that den-
expected to start in 2011. dritic spines get irreversibly lost in the cerebral cortex of
wild-type mice after only 4 days of treatment with sema-
NPG-328 gacestat (30 mg/kg s.c.). The same experiments carried out
in APP-deficient mice suggested that APP-cleavage products
The imidazylphenyl 2-thiazoleamine derivative NGP-328 (probably an accumulation of C-terminal fragments), are
Fig. (4) has been recently selected at NeuroGenetic Pharma- critically involved [21]. Recent studies have shown that a
ceuticals as a clinical candidate for the development in AD. number of proteins regulate -secretase activity, including
The compound presents the same fragments in the left hand secretase activating protein (GSAP) [78], the member of
side of both E2012 and E2212 Fig. (4). In cell cultures, the p24 cargo protein family TMP21 [79] and the orphan G-
NPG-328 selectively inhibited the production of A1-42, protein coupled receptor 3 (GPR3) [80]. These proteins rep-
while favoring the generation of the shorter A1-37 and A1-38 resent potential therapeutic target for the treatment of AD
species. The compound did no show effects on the prote- because their inhibition appear to affect A production with-
olytic processing of Notch [68]. After oral dosing, NGP-328 out affecting the cleavage of Notch.
reduced the levels of A1-42 and increased the amount of
A1-38 in the plasma, cerebrospinal fluid and brains of trans- However, even Notch-sparing -secretase inhibitors (PF-
genic animals [68]. 3084014, BMS-708163, begacestat) could have neurotoxic
effects due to their inhibitory activity on amyloid precursor
DISCUSSION protein intracellular domain (AICD) release from either C-
-Secretase Inhibitors terminal fragments of APP (C83 in the non-amyloidogenic
pathway and C99 in the amyloidogenic pathway). AICD
While a number of highly potent inhibitors of secretase may have important physiological role in gene expression,
have been identified [12], serious concerns about their toxic- apoptosis, and cytoskeletal dynamics [81].
ity have been raised since -secretase can cleave several
other membrane proteins other than APP [5], the most phar- Another possible explanation for the detrimental effects
macologically relevant being the Notch receptor. Secretase of semagacestat in AD patients is the higher inhibitory po-
inhibitors block proteolysis of Notch by inhibiting cleavage tency displayed by the drug on A1-40 production compared
at site 3 [69]. Physiological cleavage of Notch leads to re- A1-42 [17], a characteristic shared with other secretase
lease of the Notch intracellular domain (NICD), a protein inhibitors. Recent studies in double transgenic mice have
fragment that is translocated to the nucleus where it regulates shown that A1-40 inhibits amyloid deposition while A1-42
transcription of target genes involved in cell development increases it [82]. In addition, increasing A1-40 levels pro-
and in differentiation of adult self-renewing cells. The in- tected transgenic mice from the premature death. The protec-
hibitory effects of -secretase inhibitors on Notch activation tive properties of A1-40 with respect to amyloid deposition
in embryonic and fetal development may not be of concern suggest that drugs preferentially targeting A1-40 may actu-
for the treatment of AD patients. However, it is known that ally worsen the disease course. Studies in guinea-pigs [17]]
Notch signaling plays an important role in the ongoing dif- and in normal men [26] have indicated that semagacestat
ferentiation processes of the immune system [70]], gastroin- may cause rebound effects on A1-42 levels in the CNS.
testinal tract [71] and epidermis [72]. Treatment of mice with Whatever is the mechanism of the detrimental effects of se-
-secretase inhibitors can cause severe gastrointestinal toxic- magacestat on cognition in AD patients, it has to be pointed
ity and compromise the proper maturation of B- and T- out that no studies have been published showing positive
lymphocytes [73,74]]. Notch signaling is also present in the cognitive or behavioral effects of the drug in animal models
mature brain where its activation influences structural and of AD, neither after acute or chronic administration [20].
functional plasticity including processes involved in learning This is a problem shared by other -secretase inhibitors in
and memories [75]. Mice heterozygous for a null mutation in clinical developemt: there are no studies showing that the
the gene encoding Notch1 display deficits in spatial learning chronic aministration of these drugs produce positive effects
[76] and mice overexpressing Notch1 antisense mRNA have on memory in animal models of AD.
about 50% of the normal levels of Notch protein in the hip-
pocampus and do not display long-term potentiation (LTP) -Secretase Modulators
in response to high-frequency stimulation [77]. Thus, the Modulation of -secretase appears to be a logical strategy
detrimental cognitive and functional effects of semagacestat to selectively decrease A142 accumulation in AD patients
observed in the interrupted Phase 3 clinical trials may be without causing an accumulation of the potentially neuro-
ascribed to its interference activity on Notch signaling. The toxic CTF, interfering with Notch signaling and AICD re-
increased rate of skin cancer observed with semagacestat in lease. In the last two years, significant progresses have been
AD patients could be also linked to the drug inhibitory activ- made in designing and developing -secretase modulators
ity on Notch1 signaling that may have a role as tumor sup- with improved potency for lowering the production of A1-42.
pressor in certain type of non-melanoma skin cancer [71]. However, the increase in A1-42 inhibitory potency may im-
Another possible reason for the faster clinical decline of ply the loss of selectivity versus other -secretase substrates.
semagacestat-treated AD patients could be linked to the abil- It has been shown that at high concentrations -secretase
ity of the drug to accumulate the neurotoxic C-terminal modulating NSAIDs may inhibit the S3 cleavage site of
fragment of APP (CTF or C99) in response to the block of Notch-1, an effect reminiscent of classical -secretase inhibi-
1566 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

tion [83]. It appears that the separation of the inhibitory ef- normal synaptic function and for normal learning and mem-
fects on the various cleavage sites is concentration dependent ory. Thus, it may be plausible that indiscriminate and com-
leaving a “window of modulation”. The positive neuropa- plete inhibition (with -secretase inhibitors) or removal (with
thological and behavioral effects observed for some NSAIDs anti-A antibodies) of endogenous A could be detrimental
(the best documented being ibuprofen) in different trans- in AD patients rather than beneficial. A significant propor-
genic mouse models of AD, could be not linked to their abil- tion (20-30%) of cognitively intact individuals shows a sig-
ity to modulate -secretase (quite unlikely at the drug con- nificant amount of pre- or post-mortem amyloid [97, 98].
centrations reached into the brain) but rather to a plethora of Collectively, these obvservations question the hypothesis
other mechanisms that include inhibition of microglia- that A is the key pathologic factor affecting AD process. It
mediated release of cytokines [84], inhibition of microglia has been recently proposed that a decline in brain metabolic
peroxidase [85], inhibition of neuronal cell cycle events [86], activity or synaptic activity is the underlying cause of the
stimulation of neurotrophin synthesis or expression [87], disease [99]. Decreased metabolic activity, which can be
inhibition of mitochondrial Ca2+ overload [88], effects on consequent to decreased synaptic activity, increases -
astrocyte motility [89]. secretase expression or activity which, in turn, increases A
deposition as a secondary response [99]. If this is true we
The major issue for NSAIDs-like -secretase modulators
should assist to other failures of -secretase inhibitors or
is brain penetration. NSAIDs per se show good blood-brain
barrier permeability, but their high plasma protein binding other A-lowering agents, especially those for which no
proofs of neuronal rescue and attenuation of memory deficit
reduces the unbound fraction to less than 3-5% of the total
have been documented in preclinical model of AD.
concentration, thus limiting the amount of drug available to
reach CNS. -Secretase modulators with non-NSAID struc-
ture tend to have high molecular weight and lipophilicity and CONFLICT OF INTEREST
this may represent an issue since mounting evidence in the Bruno P. Imbimbo is an employee at Chiesi Farmaceutici
literature indicate that large, lipophilic compounds have a and is involved in the development of CHF5074. He is listed
higher incidence of off-target and non-selective pharmacolo- between the inventors of a number of Chiesi Framaceutici’s
gies. Finally, it will be important to gain a better understand- patents of anti-AD drugs, including -secretase modulators.
ing of the specific target(s) that these -secretase modulators
interact with in order to facilitate future drug design efforts. Giuseppe Giardina has no conflict of interest to declare.

CONCLUSIONS REFERENCES
[1] Hardy, J.; Allsop, D. Amyloid deposition as the central event in the
The recent failure of semagacestat has fueled the con- aetiology of Alzheimer's disease. Trends Pharmacol. Sci., 1991,
cerns on the safety and the efficacy of -secretase inhibitors, 12, 383-388.
not to mention questions about the A hypothesis for AD in [2] Lesne, S.; Koh, M. T.; Kotilinek, L.; Kayed, R.; Glabe, C. G.;
general. The semagacestat failure echoes a previous observa- Yang, A.; Gallagher, M.; Ashe, KH. A specific amyloid- protein
assembly in the brain impairs memory. Nature, 2006, 440, 352-
tion that immunization with pre-aggregated A1-42 (AN1792) 357.
resulted in almost complete clearance of amyloid plaques [3] Shankar, G. M.; Li S.; Mehta, T.; Garcia-Munoz A.; Shepardson,
from the brain of patients with AD but did not alter disease N. E.; Smith, I.; Brett, F. M.; Farrell, M. A.; Rowan, M. J.; Lemere,
progression [90]. It has been argued that the accumulation of C. A.; Regan, C. M.; Walsh, D. M.; Sabatini, B. L.; Selkoe, D.
A in the brain of AD patients is simply a downstream mani- J.Amyloid- protein dimers isolated directly from Alzheimer's
brains impair synaptic plasticity and memory. Nat. Med., 2008, 14,
festation of the disease rather then its cause [91]. Brain 837-842.
plaques may represent a defensive mechanism in response to [4] Imbimbo, B. P. Therapeutic potential of -secretase inhibitors and
a neuronal damage process [92]. The reason for the presence modulators. Curr. Top. Med. Chem., 2008, 8, 54-61.
of A in the normal brain and its physiological role is not [5] Lleo, A. Activity of -secretase on substrates other than APP. Curr.
Top. Med. Chem., 2008, 8, 9-16.
fully understood but it may regulate neuroplasticity [93]. [6] Weggen, S.; Eriksen, J. L.; Das, P.; Sagi, S. A.; Wang, R.; Pietrzik,
Electrophysiological studies in rodent hippocampal prepara- C. U.; Findlay, K. A.; Smith, T. E.; Murphy, M. P.; Bulter, T.;
tions have shown that endogenously released A peptides Kang, D. E.; Marquez-Sterling, N.; Golde, T. E.; Koo, E. H. A sub-
positively regulate the release ability of synapses without set of NSAIDs lower amyloidogenic A42 independently of cy-
altering postsynaptic function or intrinsic neuronal excitabil- clooxygenase activity. Nature, 2001, 414, 212-216.
[7] Peretto, I.; La Porta, E. -Secretase modulation and its promise for
ity [94]. Other in vivo studies have shown that intra- Alzheimer’s disease: a medicinal chemistry perspective. Curr. Top.
hippocampal injections of picomolar concentrations of A1-42 Med. Chem., 2008, 8, 38-46.
monomers and oligomers to normal mice cause a marked [8] Kreft, A. F.; Martone, R.; Porte, A. Recent advances in the identifi-
increase of hippocampal LTP and enhancement of reference cation of -secretase inhibitors to clinically test the A oligomer
hypothesis of Alzheimer's disease. J. Med. Chem., 2009, 52, 6169-
and contextual memory [95]. More recently, it has been 6188.
shown that low doses of A enhance memory retention and [9] Serneels, L.; Van Biervliet, J.; Craessaerts, K.; Dejaegere, T.;
acetylcholine production in the hippocampus of normal mice Horre, K.; Van Houtvin, T.; Esselmann, H.; Paul, S.; Schafer, M.
[96]. Blocking endogenous A with antibodies or decreasing K.; Berezovska, O.; Hyman, B. T.; Sprangers, B.; Sciot, R.; Moons,
A expression with antisense directed at APP, all resulted in L.; Jucker, M.; Yang, Z.; May, P. C.; Karran, E.; Wiltfang, J.;
D’hooge, R.; De Strooper, B. -Secretase heterogeneity in the
impaired learning in a spatial memory test [96]. Interest- Aph1 subunit: relevance for Alzheimer’s disease. Science, 2009,
ingly, A1-42 facilitated induction and maintenance of LTP in 324, 639-642.
hippocampal slices, whereas antibodies to A inhibited hip- [10] Osenkowski, P.; Li, H.; Ye, W.; Li, D.; Aeschbach, L.; Fraering, P.
pocampal LTP [96]. All these studies indicate that in normal C.; Wolfe, M. S.; Selkoe, D. J.; Li, H. Cryoelectron microscopy
healthy young animals the presence of A is important for
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1567

structure of purified -secretase at 12 Å resolution. J. Mol. Biol., [24] Siemers, E. R.; Quinn, J. F.; Kaye, J.; Farlow, M. R.; Porsteinsson,
2009, 385, 642-652. A.; Tariot, P.; Zoulnouni, P.; Galvin, J.E.; Holtzman, D. M.;
[11] Dovey, H. F.; John, V.; Anderson, J. P.; Chen, L. Z.; de Saint An- Knopman, D. S.; Satterwhite, J.; Gonzales, C.; Dean, R. A.; May,
drieu, P.; Fang, L. Y.; Freedman, S. B.; Folmer, B.; Goldbach, E.; P. C. Effects of a -secretase inhibitor in a randomized study of pa-
Holsztynska, E.J.; Hu, K.L.; Johnson-Wood, K. L.; Kennedy, S. L.; tients with Alzheimer disease. Neurology 2006, 66, 602-604.
Kholodenko, D.; Knops, J. E.; Latimer, L. H.; Lee, M.; Liao, Z.; [25] Fleisher, A. S.; Raman, R.; Siemers, E. R.; Becerra, L.; Clark, C.
Lieberburg, I. M.; Motter, R. N.; Mutter, L. C.; Nietz, J.; Quinn, K. M.; Dean, R. A.; Farlow, M. R.; Galvin, J. E.; Peskind, E. R.;
P.; Sacchi, K. L.; Seubert, P. A.; Shopp, G.M.; Thorsett, Quinn, J. F.; Sherzai, A. Sowell, B. B.; Aisen P. S.; Thal, L. J.
E.D.;Tung, J. S.; Wu, J.; Yang, S.; Yin, C. T.; Schenk, D. B.; May, Phase 2 safety trial targeting amyloid beta production with a -
P. C.; Altstiel, L. D.; Bender, M. H.; Boggs, L. N.; Britton, T. C.; secretase inhibitor in Alzheimer disease. Arch. Neurol., 2008, 65,
Clemens, J. C.; Czilli, D. L.; Dieckman-McGinty, D. K.; Droste, J. 1031-1038.
J. Fuson, K. S.; Gitter, B. D.; Hyslop, P. A.; Johnstone, E. M.; Li, [26] Bateman, R. J.; Siemers, E. R.; Mawuenyega, K. G.; Wen, G.;
W. Y.; Little, S. P.; Mabry, T. E.; Miller, F.D.; Audia, J. E. Func- Browning, K. R.; Sigurdson, W. C.; Yarasheski, K. E.; Friedrich, S.
tional -secretase inhibitors reduce -amyloid peptide levels in W.; Demattos, R. B.; May, P. C.; Paul, S. M.; Holtzman, D. M. A
brain. J. Neurochem., 2001, 76, 173-181. -secretase inhibitor decreases amyloid- production in the central
[12] Olson, R. E.; Albright, C. F. Recent progress in the medicinal nervous system. Ann. Neurol., 2009, 66, 48-54.
chemistry of -secretase inhibitors. Curr. Top. Med. Chem., 2008, [27] Siemers, E. R. Update on the semagacestat program. 3rd Conference
8, 17-33. - Clinical Trials on Alzheimer’s Disease. Toulouse, November 3-5,
[13] Comery, T. A.; Martone, R. L.; Aschmies, S.; Atchison, K. P.; 2010.
Diamantidis, G.; Gong, X.; Zhou, H.; Kreft, A.F.; Pangalos, M. N.; [28] Morimoto B. Drug Development for Neurodegenerative Diseases -
Sonnenberg-Reines, J.; Jacobsen, J.S.; Marquis, K.L. Acute - a Marcus Evans Conference. IDdb Meeting Report, Boston, MA,
secretase inhibition improves contextual fear conditioning in the April 7-8, 2009.
Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 2005, [29] Rosen, L. B.; Stone, J. A.; Plump, A.; Yuan, J.; Harrison, T.; Flynn,
25, 8898-8902. M.; Dallob, A.; Matthews, C.; Stevenson, D.; Schmidt, D.; Palm-
[14] Gitter, B. D.; Czilli, D. L.; Li, W.; Dieckman, D. K.; Bender, M. ieri, T.; Leibowitz, M.; Jhee, S.; Ereshefsky, L.; Salomon, R.; Win-
H.; Nissen, J. S.; Mabry, T. E.; Yin, T.; Boggs, L. N.; McClure, D. chell, G.; Shearman, M. S.; Murphy, M. G.; Gottesdiener, K. M.
B.; Little, S. P.; Johnstone, E. M.; Audia, J. E.; May, P. C., Hyslop, The -secretase inhibitor MK-0752 acutely and significantly re-
P. A. Stereoselective inhibition of amyloid  peptide secretion by duces CSF A40 concentrations in humans. Alzheimers Dement.,
LY450139 dihydrate, a novel functional gamma secretase inhibitor. 2006, 2 (Suppl 1), S79 (Abstract O4-03-02).
Neurobiol. Aging, 2004, 25 (Suppl. 2), S571 (Abstract P4-339). [30] Deangelo, D. J.; Stone, R. M.; Silverman, L. B.; Stock, W.; Attar,
[15] Boggs, L. N.; Fuson, S. F.; Gitter, B. D.; Czilli, D. L.; Hyslop, P. E. C.; Fearen, I.; Dallob, A.; Matthews, C.; Stone, J.; Freedman, S.
A.; Bender, M. H.; Li, W.; Audia, J. E.; Nissen, J. S.; Mabry, T. E.; J.; Aster, J. A phase I clinical trial of the notch inhibitor MK-0752
Ni, B.; May, P. C. In vivo characterization of LY450139, a novel, in patients with T-cell acute lymphoblastic leukemia/lymphoma (T-
stereoselective, functional -secretase inhibitor. Neurobiol. Aging, ALL) and other leukemias. J. Clin. Oncol., 2006, 24 (Suppl 18),
2004, 25 (Suppl. 2), S 218 (Abstract P1-419). 6585.
[16] May, P. C.; Yang, Z.; Li, W.; Hyslop, P. A.; Siemers, E.; Boggs, L. [31] Krop, I. E.; Kosh, M.; Fearen, I.; Savoie, J.; Dallob, A.; Matthews,
N. Multi-compartmental pharmacodynamic assessment of the func- C.; Stone, J.; Winer, E.; Freedman, S.J.; Lorusso, P. Phase I
tional -secretase inhibitor LY450139 in PDAPP transgenic mice pharmacokinetic (PK), and pharmacodynamic (PD) trial of the
and non-transgenic mice. Neuobiol. Aging, 2004, 25 (Suppl. 25), 65 novel oral Notch inhibitor MK-0752 in patients (pts) with advanced
(Abstract O3-06-07). breast cancer (BC) and other solid tumors. J. Clin. Oncol., 2006, 24
[17] Lanz, T. A.; Karmilowicz, M. J.; Wood, K. M.; Pozdnyakov, N.; (Suppl 18), 10574.
Du, P.; Piotrowski, M. A.; Brown, T. M.; Nolan, C. E.; Richter, K. [32] Gillman, K. W.; Starrett, J. E.; Parker, M. F.; Xie, K.; Bronson, J.
E.; Finley, J. E.; Fei, Q.; Ebbinghaus, C. F.; Chen, Y. L.; Spracklin, J.; Marcin, L. R; McElhone, K. E.; Bergstrom, C. P.; Mate, R. A.;
D. K.; Tate, B.; Geoghegan, K. F.; Lau, L. F.; Auperin, D. D.; Williams, R.; Meredith Jr., J. E.; Burton, C. R.; Barten, D. M.;
Schachter, J. B. Concentration-dependent modulation of A in vivo Toyn, J. H.; Roberts, S. B.; Lents, K. A.; Houston, J. G., Zaczek,
and in vitro using the -secretase inhibitor, LY- 450139. J. Phar- R.; Albright, C. F.; Decicco, C. P.; Macor, J. E., Olson, R. E. Dis-
macol. Exp. Ther., 2006, 319, 924-933. covery and evaluation of BMS-708163, a potent, selective and
[18] Cocke, P. J.; Boggs, L. N.; Ness, D. K.; May, P. C. Comparison of orally bioavailable -secretase inhibitor. ACS Med. Chem. Lett.,
cisternal and lumbar cerebrospinal fluid amyloid beta concentra- 2010, 1, 120-124.
tions following oral administration of LY450139 dihydrate to Bea- [33] Albright, C. F.; Dockens, R.; Olson, R. E.; Meredith, J.; Slemmon,
gle dogs. Alzheimers Dement., 2008, 4 (Suppl 2), T210 (Abstract R.; Lentz, K.; Wang, J. S.; Denton, R. R.; Oilcher, G. P.; Zacaek,
P1-017). R.; Macor, J.E.; Wong, O.; Gu, H., Berman, R.; Tong, G. BMS-
[19] Ness, D.K.; Boggs, L. N.; Hepburn, D. L.; Gitter, B.; Long, G. G.; 708163, a potent and selective -secretase inhibitor, decreases CSF
May, P. C.; Piroozi, K. S.; Schafer, K. A., Yang, Z. Reduced - A at safe and tolerable doses in animals and humans. 11th Interna-
amyloid burden, increased C-99 concentrations and evaluation of tional Conference on Alzheimer’s Disease (ICAD), 26-31 July
neuropathology in the brains of PDAPP mice given LY450139 di- 2008, Chicago, IL, USA (Abstract HT-01-05).
hydrate daily by gavage for 5 months. Neurobiol. Aging, 2004, 25 [34] Tong, G.; Castaneda, L.; Wang, J. S.; Sverdlov, A.; Huamg, S.;
(Suppl. 2), S238 (Abstract P2-053). Slemmon, R.; Gu, H.; Wong, O.; Li, H.; Berman, R. M.; Smith, C.;
[20] Imbimbo, B. P.; Peretto, I. Semagacestat, a -secretase inhibitor for Albright, C. F. A study to evaluate the effects of single oral doses
the potential treatment of Alzheimer's disease. Curr. Opin. Investig. of BMS-708163 in the cerebrospinal fluid of healthy young men.
Drugs, 2009, 10, 721-730. Alzheimers Dement., 2010, 6 (Suppl 1), S143 (Abstract O3-07-07).
[21] Bittner, T.; Fuhrmann, M.; Burgold, S.; Jung, C. K.; Volbracht, C.; [35] Wang, J. S.; Catenada, L.; Sverdlov, A.; Huang, S. P.; Slemmon,
Steiner, H.; Mitteregger, G.; Kretzschmar, H. A.; Haass, C.; Herms, R.; Gu, H.; Wong, O.; Li, H.; Berman, R. M.; Smith, C.; Albright,
J. -Secretase inhibition reduces spine density in vivo via an amy- C. F. A placebo-controlled, ascending, multiple-dose study to
loid precursor protein-dependent pathway. J. Neurosci., 2009, 29, evaluate the safety, pharmacokinetics (P) and pharmacodynamics
10405-10409. (PD) of BMS-708163 in healthy young and elderly subjects. Alz-
[22] Siemers, E. R.; Dean, R. A.; Friedrich, S.; Ferguson-Sells, L.; heimers Dement., 2010, 6 (Suppl 1), S540 (Abstract P3-303).
Gonzales, C.; Farlow, M. R.; May, P. C. Safety, tolerability, and ef- [36] Lanz, T. A.; Wood, K. M.; Richter, K. E.; Nolan, C. E.; Becker, S.
fects on plasma and cerebrospinal fluid amyloid- after inhibition L.; Pozdnyakov, N.; Martin, B. A.; Du P.; Oborski, C. E.; Wood,
of -secretase. Clin. Neuropharmacol., 2007, 30, 317-325. D. E.; Brown, T. M.; Finley, J. E.; Sokolowski, S. A.; Hicks, C. D.;
[23] Siemers, E.; Skinner, M.; Dean, R. A.; Gonzales, C.; Satterwhite, Coffman, K. J.; Geoghegan, K. F.; Brodney, M. A.; Liston, D.;
J.; Farlow, M.; Ness, D.; May, P. C. Safety, tolerability, and Tate, B. Pharmacodynamics and pharmacokinetics of the -
changes in amyloid- concentrations after administration of a - secretase inhibitor PF-3084014. J. Pharmacol. Exp. Ther., 2010,
secretase inhibitor in volunteers. Clin. Neuropharmacol. 2005, 28, 334, 269-277.
126-132.
1568 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

[37] Brodney, M. Discovery of PF-3084014: A novel -secretase inhibi- Riccardi, B.; Biscaioli, M.; Marchetti, S.; Puccini, P.; Catinella, S.;
tor for the treatment of Alzheimer's disease. 238th ACS National Rondelli, I.; Cenacchi, V.; Bolzoni, P. T.; Caruso, P.; Villetti, G.;
Meeting, Washington, DC, August 16-20, 2009. Abstract MEDI Facchinetti, F.; Del Giudice, E.; Moretto, N.; Imbimbo, B. P. Syn-
233. thesis and biological activity of flurbiprofen analogues as selective
[38] Qui, R.; Willavize, S.; Fullerion, T.; Gastonguay, M. R. Modeling inhibitors of amyloid1-42 secretion. J. Med. Chem., 2005, 48,
and simulation of plasma A in humans after multiple oral doses of 5705-5720.
Pf-3084014, a potent -secretase inhibitor. Alzheimers Dement., [50] Eriksen, J. L.; Sagi, S. A.; Smith, T. E.; Weggen, S.; Das, P.;
2009, 5 (Suppl 1), P253 (Abstract P1-261). McLendon, D. C.; Ozols, V. V.; Jessing, K. W.; Zavitz, K. H.;
[39] Martone, R. L.; Zhou, H.; Atchison, K.; Comery, T.; Xu, J. Z.; Koo, E. H.; Golde, T. E. NSAIDs and enantiomers of flurbiprofen
Huang, X.; Gong, X.; Jin M.; Kreft, A.; Harrison, B.; Mayer, S. C.; target -secretase and lower A42 in vivo. J. Clin. Invest., 2003,
Aschmies, S.; Gonzales, C.; Zaleska, M. M.; Riddell, D. R.; 112, 440-449.
Wagner, E.; Lu, P.; Sun, S. C. ; Sonnenberg-Reines, J.; Oganesian, [51] Kukar, T.; Prescott, S.; Eriksen, J. L.; Holloway, V.; Murphy, M.
A.; Adkins, K.; Leach, M. W.; Clarke, D. W.; Huryn, D.; Abou- P.; Koo, E. H.; Golde, T. E.; Nicolle, M. M. Chronic administration
Gharbia, M.; Magolda, R.; Bard, J.; Frick, G.; Raje, S.; Forlow, S. of R-flurbiprofen attenuates learning impairments in transgenic
B.; Balliet, C.; Burczynski, M. E.; Reinhart, P. H.; Wan, H. I.; amyloid precursor protein mice. BMC Neurosci., 2007, 8, 54.
Pangalos, M. N.; Jacobsen, J. S. Begacestat (GSI-953): a novel, se- [52] Lanz, T. A.; Fici, G. J.; Merchant, K. M. Lack of specific amyloid-
lective thiophene sulfonamide inhibitor of amyloid precursor pro- 1-42 suppression by nonsteroidal anti-inflammatory drugs in young,
tein -secretase for the treatment of Alzheimer's disease. J. plaque-free Tg2576 mice and in guinea pig neuronal cultures. J.
Pharmacol. Exp. Ther., 2009, 331, 598-608. Pharmacol. Exp. Ther., 2005, 312, 399-406.
[40] Frick, G.; Raje, S.; Wan, H.; Forlow; S. B.; Balliet, C.; Pastore, A.; [53] Galasko, D. R.; Graff-Radford, N.; May, S.; Hendrix, S.; Cottrell,
Burczynski, M. E.; Jhee, S.; Ereshefsky, L.; Paul, J. GSI-953, a po- B. A.; Sagi, S. A.; Mather, G.; Laughlin, M.; Zavitz, K. H.; Swabb,
tent and selective -secretase inhibitor - Modulation of -amyloid E.; Golde, T. E.; Murphy, M. P. Koo, E. H. Safety, tolerability,
peptides and plasma and cerebrospinal fluid pharmacokinetic/ pharmacokinetics, and Ab levels after short-term administration of
pharmacodynamic relationships in humans. Alzheimers Dement., R-flurbiprofen in healthy elderly individuals. Alzheimer. Dis. As-
2008, 4 (Suppl. 2), T781 (Abstract P4-366). soc. Disord., 2007, 21, 292-299.
[41] Wan, H.; Bard, J.; Martone, R.; Rage, S.; Forlow, S.; Kreft, A.; [54] Wilcock, G. K.; Black, S. E.; Hendrix, S. B.; Zavitz, K. H.; Swabb,
Jacobsen, S.; Silver, P.; Paul, J.; Frick, G. GSI-953, a potent and E. A.; Laughlin, M. A. Efficacy and safety of tarenflurbil in mild to
selective -secretase inhibitor, modulates A peptides in mice and moderate Alzheimer's disease: a randomised phase II trial. Lancet
humans: Translating the PK/PD biomarker relationships in differ- Neurol., 2008, 7, 483-493.
ent biological compartments between rodent and human. Alzheim- [55] Green, R. C.; Schneider, L. S.; Amato, D. A.; Beelen, A. P.;
ers Dement., 2008, 4 (Suppl. 2), T548 (Abstract P2-101). Wilcock, G.; Swabb, E. A.; Zavitz, K. H.; +Tarenflurbil Phase 3
[42] Brigham, E.; Quinn, K.; Kwong, G.; Willits, C.; Goldbach, E.; Study Group. Effect of tarenflurbil on cognitive decline and activi-
Motter, R.; Lee, M.; Wallace, W.; Kholodenko, D.; Tang-Tanake, ties of daily living in patients with mild Alzheimer disease: a ran-
P.; Ni, H.; Hemphill, S.; Chen, X. H.; Eichenbaum, T.; Ruslim, L.; domized controlled trial. JAMA, 2009, 302, 2557-2564.
Nguyen, L.; Santiago, P.; Liao, A.; Bova, M.; Probst, G.; Dappen, [56] Wilcock, G. K.; Black, S. E.; Balch, A. H.; Amato, D. A.; Beelen,
M.; Jagodzinski, J.; Basi, G.; Ness, D. Pharmacokinetic and phar- A. P.; Schneider, L. S.; Green, R. C.; Swabb, E. A.; Zavitz, K. H.
macodynamic investigation of ELND006, a novel APP-selective - Safety and efficacy of tarenflurbil in subjects with mild Alz-
secretase inhibitor, on amyloid- concentrations in the brain, CSF heimer’s disease: results from an 18-month international multi-
and plasma of multiple nonclinical species following oral admini- center Phase 3 trial. Alzheimers Dement., 2009, 5 (Suppl 1), P86
stration.. Alzheimers Dement., 2010a, 6 (Suppl 1), S546 (Abstract (Abstract O1-04-07).
P3-320). [57] Amino, H.; Hagiwara, H.; Murata, Y.; Watanabe, H.; Sasaki, T.;
[43] Brigham, E.; Quinn, K.; Motter, R.; Hoffman, W.; Goldbach, E.; Miyagawa, T. E2012: a novel -secretase modulator - mechanism
Kholodenko, D.; Kwong, G.; Willits, C.; Probst, G.; Gunther, J.; of action. Alzheimers Dement., 2010, 6 (Suppl 1): S54-S542 (Ab-
Admas, E.; Sauer, J.M.; Kinney, G.; Ness, D. Effects of single and stract P3-307).
multiple dose oral administration of ELND006, a novel APP- [58] Hashimoto, T.; Ishibashi, A.; Hagiwara, H.; Murata, Y.; Takenaka,
selective -secretase inhibitor, on amyloid- concentrations in the O.; Miyagawa, T. E2012: A novel -secretase modulator - pharma-
brain and CSF of cynomolgus monkeys. Alzheimers Dement., cology part. 13th International Conference on Alzheimer's Disease
2010b, 6 (Suppl 1), S546 (Abstract P3-322). and Related Disorders (ICAD), Honolulu, July 10-15, 2010. Alz-
[44] Schroeter, A.; Brigham, E.; Motter, R.; Nishioka, C.; Guido, T.; heimers Dement., 2010, 6 (Suppl 1), S242 (Abstract P1-236).
Khan, K.; Kholodenko, D.; Tanaka, P.; Soriano, F.; Quinn, K.; [59] Portelius, E.; Van Broeck, B.; Andreasson, U.; Gustavsson, M. K.;
Goldbach, E.; Games, D.; Ness, D. APP-selective -secretase in- Mercken, M.; Zetterberg, H.; Borghys, H.; Blennow, K. Acute ef-
hibitor ELND006 effects on brain parenchymal and vascular amy- fect on the A isoform pattern in CSF in response to -secretase
loid- in the PDAPP mouse model of Alzheimer’s disease. Alz- modulator and inhibitor treatment in dogs. J. Alzheimers Dis.,
heimers Dement., 2010, 6 (Suppl 1): S546 (Abstract P3-321). 2010, 21, 1005-1012.
[45] Kukar, T. L.; Ladd, T. B.; Bann,M. A.; Fraering, P. C.; Narlawar, [60] Nagy, C.; Schuck, E.; Ishibashi, A.; Nakatani, Y.; Rege, B.; Lago-
R.; Maharvi, G. M.; Healy, B.; Chapman, R.; Welzel, A. T.; Price, vinsky, V. E2012, a novel -secretase modulator, decreases plasma
R. W.; Moore, B.; Rangachari, V.; Cusack, B.; Eriksen, J.; Jansen- amyloid beta (A) levels in humans. Alzheimers Dement. 2010, 6
West, K.; Verbeeck, C.; Yager, D.; Eckman, C.; Ye, W.; Sagi, S.; (Suppl 1), S574 (Abstract P3-415).
Cottrell, B. A.; Torpey, J.; Rosenberry, T. L.; Fauq, A.; Wolfe, M. [61] Imbimbo, B. P.; Del Giudice, E.; Colavito, D.; D'Arrigo, A.; Dalle
S.; Schmidt, B.; Walsh, D. M.; Koo, E. H.; Golde, T. E. Sub- Carbonare, M.; Villetti, G.; Facchinetti, F.; Volta, R., Pietrini, V.;
stratetargeting -secretase modulators. Nature, 2008, 453, 925-929. Baroc, M. F.; Serneels, L.; De Strooper, B.; Leon, A. CHF5074, a
[46] Beher, D.; Clarke, E. E.; Wrigley, J. D.; Martin, A. C.; Nadin, A.; novel -secretase modulator, reduces brain -amyloid pathology in
Churcher, I.; Shearman, M. S. Selected non-steroidal anti- a transgenic mouse model of Alzheimer's disease without causing
inflammatory drugs and their derivatives target -secretase at a peripheral toxicity. J. Pharmacol. Exp. Ther., 2007, 323, 822-830.
novel site-evidence for an allosteric mechanism. J. Biol. Chem., [62] Imbimbo, B. P.; Hutter-Paier, B.; Villetti, V.; Facchinetti, F.;
2004, 279, 43419-43426. Cenacchi, V.; Volta, R. Lanzillotta, A. Pizzi, M.; Windisch, M.
[47] Sato, T.; Tang, T.-C.; Reubins, G.; Fei, J. Z.; Fujimoto, T.; Kien- CHF5074, a novel -secretase modulator, attenuates brain
len-Campard, P.; Constantinescu, S. N.; Octave, J.-N.; Aimoto, S.; -amyloid pathology and learning deficit in a mouse model of Alz-
Smith, S. O. A helix-to-coil transition at the -cut site in the trans- heimer’s disease. Br. J. Pharmacol., 2009, 156, 982-993.
membrane dimer of the amyloid precursor protein is required for [63] Imbimbo, B. P.; Giardino, L.; Sivilia, S.; Giuliani, A.; Gusciglio,
proteolysis. Proc. Natl. Acad. Sci. USA, 2009, 106, 1421-1426. M.; Pietrini, V.; Del Giudice, E.; D'Arrigo, A.; Leon, A.; Villetti,
[48] Morihara, T.; Chu, T.; Ubeda, O.; Beech, W.; Cole, G.M. Selective G.; Calzà, C. CHF5074, a novel -secretase modulator, restores
inhibition of A42 production by NSAID R-enantiomers. J. Neuro- hippocampal neurogenesis potential and reverses contextual mem-
chem., 2002, 83, 1009-1012. ory deficit in a transgenic mouse model of Alzheimer’s disease. J.
[49] Peretto, I.; Radaelli, S.; Parini, C.; Zandi, M.; Raveglia, L. F.; Don- Alzheimers Dis., 2010a, 20, 159-173.
dio, G.; Fontanella, L.; Misiano, P.; Bigogno, C.; Rizzi, A.;
 -Secretase Inhibitors and Modulators for the Treatment of Alzheimer’s Disease Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 1569

[64] Imbimbo, B. P.; Mackintosh, D.; Aponte, T.; Frigerio, E.; Breda, orphan G protein-coupled receptor 3 modulates amyloid- peptide
M.; Fernandez, M.; Giardino, L.; Calzà, L.; Norris, D.;Wagner, K.; generation in neurons. Science, 2009, 323, 946-951.
Ross, J. Pharmacokinetics and pharmacodynamics of CHF5074, a [81] Müller, T.; Meyer, H. E.; Egensperger, R.; Marcus, K. The amyloid
novel -secretase modulator, after single oral administration to precursor protein intracellular domain (AICD) as modulator of
healthy male subjects. Alzheimers Dement., 2010b, 6 (Suppl 1), gene expression, apoptosis, and cytoskeletal dynamics-relevance
S573 (Abstract P3-411). for Alzheimer's disease. Prog. Neurobiol., 2008, 85, 393-406.
[65] Eisai Co., Ltd. FY2009 Product Creation Meeting, December 18, [82] Kim, J.; Onstead, L.; Randle, S.; Price, R.; Smithson, L.; Zwizin-
2009. www.eisai.co.jp/pdf/eir/emat/material20091218e.pdf. ski, C.; Dickson, D. W.; Golde, T.; McGowan, E. A40 inhibits
[66] Felsenstein, K. M.; Spaulding, D.; Yang, Z.; Hodgon, H.; Costa, amyloid deposition in vivo. J. Neurosci., 2007, 27, 627-633.
D.; Nolan, S.; Wen, M.; Lee, W.; Hrdlicka, L.; Catana, F.; Albaya, [83] Wrigley, J. D. J.; Martin, A. C. L.; Clarke, E. E.; Lewis, H. D.;
F.; Tu, Z.; Patzke, H.; Chesworth, R.; Shapiro, G.; Zaninovic, I.; Pollack, S. J.; Shearman, M. S.; Beher, D. Modulation versus inhi-
Ahlijanian, M.; Koenig, G.; Rogers, K. Pharmacokinetic and phar- bition - selected NSAIDs can act as -secretase inhibitors in vitro.
macodynamic analysis of the -secretase modulator (GSM) EVP- 33rd Annual Meeting of Society of Neuroscience. New Orleans,
0015962. Alzheimers Dement., 2010, 6 (Suppl 1), S538 (Abstract November 8-12, 2003. Abstract 295.2.
P3-295). [84] Terrando, N.; Monaco, C.; Ma, D.; Foxwell, B. M.; Feldmann, M.;
[67] Rogers, K.; Leventhal, L.; Hopp, S.; Miller, M. J.; Yang, Z.; Maze, M. Tumor necrosis factor- triggers a cytokine cascade
Hrdlicka, L.; Lee, W.; Hodgon, H.; Nolan, S.; Wen, M.; Koperniak, yielding postoperative cognitive decline. Proc. Natl. Acad. Sci.
T.; Spaulding, D.; Catana, F.; Chesworth, R.; Shapiro, G.; Costa, USA, 2010, 107, 20518-20522.
D.; Ahlijanian, M.; Koenig, G.; Felsenstein, K.; The -secretase [85] Wilkinson, B. L.; Cramer, P. E.; Varvel, N. H.; Reed-Geaghan, E.;
modulator EVP-0015962 improves cognitive deficits in Tg2576 Jiang, Q.; Szabo, A.; Herrup, K.; Lamb, B. T.; Landreth, G. E. Ibu-
mice concomitantly with decreases in A42. Alzheimers Dement., profen attenuates oxidative damage through NOX2 inhibition in
2010, 6 (Suppl 1), Abstract P4-022. Alzheimer's disease. Neurobiol. Aging., 2010, [Epub ahead of
[68] Cheng, S.; Comer, D.; Mao, L.; Pleynet, D.; Yu, C.; Kounnas, M. print].
Z.; Tyree, C.; Danks, A. D.; Wagner, S. L.; Comer, W. T. 2- [86] Varvel, N. H.; Bhaskar, K.; Kounnas, M. Z.; Wagner, S. L.; Yang,
Aminothiazoles derivatives as potent -secretase modulators. 240th Y.; Lamb, B. T.; Herrup, K. NSAIDs prevent, but do not reverse,
ACS National Meeting, Boston, August 22-26, 2010, Abstract neuronal cell cycle reentry in a mouse model of Alzheimer disease.
MEDI 1. J. Clin. Invest., 2009, 119, 3692-3702.
[69] Lewis, H. D.; Perez Revuelta, B. I.; Nadin, A.; Neduvelil, J. G.; [87] Zhao, X.; Rebeck, G. W.; Hoe, H. S.; Andrews, P. M. Tarenflurbil
Harrison, T.; Pollack, S. J.; Shearman, M. S. Catalytic site-directed protection from cytotoxicity is associated with an upregulation of
-secretase complex inhibitors do not discriminate pharmacologi- neurotrophins. J. Alzheimers Dis., 2008, 15, 397-407.
cally between Notch S3 and beta-APP cleavages, Biochemistry, [88] Sanz-Blasco, S.; Valero, R.A.; Rodríguez-Crespo, I.; Villalobos,
2003, 42, 7580-7586. C.; Núñez, L. Mitochondrial Ca2+ overload underlies A oli-
[70] Maillard, I.; Adler, S. H.; Pear, W. S. Notch and the immune sys- gomers neurotoxicity providing an unexpected mechanism of neu-
tem. Immunity, 2003, 19, 781-.791. roprotection by NSAIDs. PLoS One, 2008, 3, e2718.
[71] Stanger, B. Z.; Datar, R.; Murtaugh, L. C.; Melton, D. A. Direct [89] Lichtenstein, M.; Carriba, P.; Wojciak-Stothard, B.; Baltrons, M.
regulation of intestinal fate by Notch. Proc. Natl. Acad. Sci. USA, A.; Petersen, J.; García, A.; Galea, E. Secretase-independent and
2005, 102, 12443-12448. RhoGTPase/ERK/PAK regulation of cytoskeleton dynamics and
[72] Panelos, J.; Massi, D. Emerging role of Notch signaling in epider- motility in astrocytes by NSAIDs and derivatives. J. Alzheimers
mal differentiation and skin cancer. Cancer Biol Ther., 2009, 8, Dis., 2010, 22(4),1135-1155
1986-1993. [90] Holmes, C.; Boche, D.; Wilkinson, D.; Yadegarfar, G.; Hopkins,
[73] Searfoss, G. H.; Jordan, W. H.; Calligaro, D. O.; Galbreath, E. J.; V.; Bayer, A.; Jones, R. W.; Bullock, R.; Love, S.; Neal, J. W.;
Schirtzinger, L. M.; Berridge, B. R.; Gao, H.; Higgins, M. A.; May, Zotova, E.; Nicoll, J. A. Long-term effects of A42 immunisation
P. C.; Ryan, T. P. Adipsin, a biomarker of gastrointestinal toxicity in Alzheimer's disease: follow-up of a randomised, placebo-
mediated by a functional -secretase inhibitor. J. Biol. Chem., 2003, controlled phase I trial. Lancet, 2008, 372, 216-223.
278, 46107-46116. [91] Castellani, R. J.; Lee, H.G.; Zhu, X.; Perry, G.; Smith, M. A. Alz-
[74] Wong, G. T.; Manfra, D.; Poulet, F. M.; Zhang, Q.; Josien, H.; heimer disease pathology as a host response. J. Neuropathol. Exp.
Bara, T.; Engstrom, L.; Pinzon-Ortiz, M.; Fine, J. S.; Lee, H. J.; Neurol., 2008, 67, 523-531.
Zhang, L.; Higgins, G. A.; Parker, E. M. Chronic treatment with [92] Castellani, R. J.; Lee, H. G.; Siedlak, S. L.; Nunomura, A.; Haya-
the -secretase inhibitor LY-411,575 inhibits -amyloid peptide shi, T.; Nakamura, M.; Zhu, X.; Perry, G.; Smith, M. A. Reexamin-
production and alters lymphopoiesis and intestinal cell differentia- ing Alzheimer's disease: evidence for a protective role for amyloid-
tion. J. Biol. Chem., 2004, 279, 2876-2882. beta protein precursor and amyloid-beta. J. Alzheimers Dis., 2009,
[75] Lathia, J. D.; Mattson, M. P.; Cheng, A. Notch: from neural devel- 18, 447-452.
opment to neurological disorders. J. Neurochem., 2008, 107, 1471- [93] Tampellini, D.; Rahman, N.; Gallo, E. F.; Huang, Z.; Dumont, M.;
1481. Capetillo-Zarate, E.; Ma, T.; Zheng, R.; Lu, B.; Nanus, D. M.; Lin,
[76] Costa, R. M.; Honjo, T.; Silva, A. J. Learning and memory deficits M. T.; Gouras, G. K. Synaptic activity reduces intraneuronal A,
in Notch mutant mice. Curr. Biol. 2003, 13, 1348-1354. promotes APP transport to synapses, and protects against A re-
[77] Wang, Y.; Chan, S. L.; Miele, L.; Yao, P. J.; Mackes, J.; Ingram, lated synaptic alterations. J. Neurosci. 2009, 29, 9704-9713.
D. K.; Mattson, M. P.; Furukawa, K. Involvement of Notch signal- [94] Abramov, E.; Dolev, I.; Fogel, H.; Ciccotosto, G.D.; Ruff, E.;
ing in hippocampal synaptic plasticity. Proc. Natl. Acad. Sci. USA, Slutsky, I. Amyloid- as a positive endogenous regulator of release
2004, 101, 9458-9462. probability at hippocampal synapses. Nat. Neurosci., 2009, 12,
[78] He, G.; Luo, W.; Li, P.; Remmers, C.; Netzer, W.J.; Hendrick, J.; 1567-1576.
Bettayeb, K.; Flajolet, M.; Gorelick, F.; Wennogle, L. P.; Green- [95] Puzzo, D.; Privitera, L.; Leznik, E.; Fà, M.; Staniszewski, A;
gard, P. -Secretase activating protein is a therapeutic target for Palmeri, A.; Arancio, O. Picomolar amyloid- positively modulates
Alzheimer's disease. Nature, 2010, 467, 95-98. synaptic plasticity and memory in hippocampus. J. Neurosci.,
[79] Chen, F.; Hasegawa, H.; Schmitt-Ulms, G.; Kawarai, T.; Bohm, C.; 2008, 28, 14537-14545.
Katayama, T.; Gu, Y.; Sanjo, N.; Glista, M.; Rogaeva, E.; [96] Morley, J. E.; Farr, S. A.; Banks, W. A.; Johnson, S. N.; Yamada,
Wakutani, Y.; Pardossi-Piquard, R.; Ruan, X.; Tandon, A.; K. A.; Xu, L. A physiological role for amyloid- protein: en-
Checler, F.; Marambaud, P.; Hansen, K.; Westaway, D.; St George- hancement of learning and memory. J. Alzheimers Dis., 2010, 19,
Hyslop, P.; Fraser P. TMP21 is a presenilin complex component 441-449.
that modulates -secretase but not -secretase activity. Nature, [97] Bennett, D. A.; Schneider, J. A.; Arvanitakis, Z.; Kelly, J. F.; Ag-
2006, 440, 1208-1212. garwal, N. T.; Shah, R. C.; Wilson, R. S. Neuropathology of older
[80] Thathiah, A.; Spittaels, K.; Hoffmann, M.; Staes, M.; Cohen, A.; persons without cognitive impairment from two communitybased
Horré, K.; Vanbrabant, M.; Coun, F.; Baekelandt, V.; Delacourte, studies. Neurology, 2006, 66, 1837-1844.
A.; Fischer, D. F.; Pollet, D.; De Strooper, B.; Merchiers. P. The [98] Aizenstein, H. J.; Nebes, R. D.; Saxton, J. A.; Price, J. C.; Mathis,
C. A.; Tsopelas, N. D.; Ziolko, S. K.; James, J. A.; Snitz, B. E.;
1570 Current Topics in Medicinal Chemistry, 2011, Vol. 11, No. 12 Imbimbo and Giardina

Houck, P. R.; Bi, W.; Cohen, A. D.; Lopresti, B. J.; DeKosky, S. [99] Struble, R. G.; Ala, T.; Patrylo, P. R.; Brewer, G. J., Yan, X. X. Is
T.; Halligan, E. M.; Klunk, W. E. Frequent amyloid deposition brain amyloid production a cause or a result of dementia of the Alz-
without significant cognitive impairment among the elderly. Arch. heimer's type? J. Alzheimers Dis., 2010, 22, 393-399.
Neurol., 2008, 65, 1509-1517.

Received: November 28, 2010 Accepted: March 22, 2011

View publication stats

You might also like