Anda di halaman 1dari 12

Hindawi Publishing Corporation

International Journal of Alzheimers Disease


Volume 2012, Article ID 369808, 11 pages
doi:10.1155/2012/369808

Review Article
Alzheimers Disease and the Amyloid Cascade Hypothesis:
A Critical Review

Christiane Reitz1, 2, 3
1 Taub Institute for Research on Alzheimers Disease and the Aging Brain, College of Physicians and Surgeons,
Columbia University, New York, NY 10032, USA
2 Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
3 Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA

Correspondence should be addressed to Christiane Reitz, cr2101@columbia.edu

Received 28 November 2011; Accepted 3 January 2012

Academic Editor: Laura Morelli

Copyright 2012 Christiane Reitz. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Since 1992, the amyloid cascade hypothesis has played the prominent role in explaining the etiology and pathogenesis of
Alzheimers disease (AD). It proposes that the deposition of -amyloid (A) is the initial pathological event in AD leading to the
formation of senile plaques (SPs) and then to neurofibrillary tangles (NFTs), neuronal cell death, and ultimately dementia. While
there is substantial evidence supporting the hypothesis, there are also limitations: (1) SP and NFT may develop independently, and
(2) SPs and NFTs may be the products rather than the causes of neurodegeneration in AD. In addition, randomized clinical trials
that tested drugs or antibodies targeting components of the amyloid pathway have been inconclusive. This paper provides a critical
overview of the evidence for and against the amyloid cascade hypothesis in AD and provides suggestions for future directions.

1. Introduction the deposition of A is the initial pathological trigger in


the disease, which subsequently leads to the formation
Alzheimers disease (AD), which is characterized by pro- of NFTs, neuronal cell death and dementia. While there
gressive deterioration in cognition, function, and behavior, is considerable evidence supporting this hypothesis, there
places a considerable burden on western societies. It is the are observations that seem to be inconsistent. This paper
sixth leading cause of all deaths and the fifth leading cause summarizes the current evidence for and against the amyloid
of death in persons aged 65 years. To date, an estimated cascade in AD.
5.4 million Americans have AD, but due to the baby boom
generation, the incidence in 2050 is expected to reach a
million persons per year, resulting in a total estimated 2. Amyloid Cascade Hypothesis
prevalence of 11 to 16 million aected persons.
Since the first description of presenile dementia by Alois As described above, two key observations resulted in the
Alzheimer in 1907 [1], senile plaques (SPs) and neurofib- original formulation of the ACH (Figure 1). First, the
rillary tangles (NFTs) are considered the key pathological detection of A as a main constituent of the SPs [3] and
hallmarks of AD [2]. The identification of -amyloid (A) second mutations of the APP [4], PSEN1, and PSEN2 genes
in SPs [3] and genetic studies that identified mutations [5, 6], which were found in families with early-onset AD
in the amyloid precursor protein (APP) [4], presenilin 1 (FAD, disease onset < 60 years). As a consequence of these
(PSEN1), and presenilin 2 (PSEN2) genes [5, 6] leading to observations, the presence of A within SPs was interpreted
the accumulation of A and early-onset familial dementia as an eect of these mutations that subsequently leads to
[4, 5, 7], resulted in the formulation of the Amyloid Cascade cell death and dementia. Since FAD hasexcept the earlier
Hypothesis (ACH; Figure 1) [8, 9]. According to the ACH, onseta similar phenotype to late-onset AD, it was assumed
2 International Journal of Alzheimers Disease

APP
APP, PSEN1, PSEN2 APP FAD mutations,
FAD mutations trisomy 21
A42 aggregation

?
Soluble forms of oligomeric A Deposited A peptide
Aggregate stress

Tau + NFT

Neuronal dysfunction and death

Dementia

Figure 1: Amyloid cascade hypothesis.

that this amyloid deposition could explain the pathogenesis hippocampal neurons, suggesting that the generation of APP
of all types of AD. could be a specific response to loss of functional innervation
of the cortex [16, 17]. Denervation of the dopamine pathways
and septal lesions aecting both the cholinergic system
3. Evidence from Studies on the Formation and -aminobutyric acid (GABA) neurons projecting to the
of A and Tau dentate gyrus can result in a loss of dendritic microtubule-
associated protein 2 (MAP2) and the appearance of tau-
There are two major objections regarding the ACH as
immunoreactive dentate gyrus granule cells [18]. Thus, den-
originally formulated. First, SPs and NFTs may be reactive
ervation can cause transsynaptic changes in dentate gyrus
products resulting from neurodegeneration in AD rather
neurons, and these alterations may represent an intermediate
than being its cause, and, second, it remains unclear whether
step to NFTs formation.
and how the deposition of A leads to the formation of NFTs.

3.1. A and Tau as Reactive Processes. In persons who suf- 3.2. Relation of the Formation of NFT to A. SPs and NFTs
fered from head trauma, APP is found with pathological cluster in a significant proportion of cortical areas but they
features similar to AD in neuronal perikarya and in dys- seem to be distributed independently of each other [19]. SP
trophic neurites surrounding A deposits [10]. In addition, and NFTs also seem to occur temporally separated; in the
there is evidence that neurons in the medial temporal lobe entorhinal cortex the occurrence of NFTs may in fact precede
secrete APP and display increased APP immunoreactivity the occurrence of SPs [20]. This spatial and temporal separa-
[11]. These findings suggest that increased expression of APP tion may suggest that they are pathogenically disconnected.
in head trauma cases may be an acute-phase response to However, evidence for an eect of A on the formation
neuronal injury [12], which in turn leads to increased A of NFT comes from transgenic experiments. The presence of
deposition. This notion is supported by the observation APP mutations alone or in combination with PSEN1 muta-
that the dierent morphological forms of A deposits, tions seems to induce A deposits in normal brain and some
including diuse, primitive, and classic deposits, contain degree of hyperphosphorylated tau in neurites [21] although
acute phase proteins such as complement factors and -anti- it does not appear to induce tau pathology or a significant
chymotrypsin [13]. Consequently, it has been proposed that, inflammatory response. These findings are consistent with
in AD, APP may be a reaction to the disease process in order studies in which fetal rat hippocampal neurons and human
to help maintain cell function, neuronal growth, and survival cortical neurons treated with fibrielar A display an increased
[14]. The putative neurotrophic action of APP is supported degree of tau phosphorylation [22] providing additional
by the observation that it shares structural features with the evidence that amyloid fibril formation might alter the
precursor for epidermal growth factor [14]. Finally, there is phosphorylation state of tau, which in turn results in the loss
also evidence that NFTs may form as a neuronal response to of microtubule-binding capacity. Other studies showed that
injury [15]. A2535 can induce the aggregation of tau proteins and that a
There are also findings from animal studies suggesting decrease in aggregation of A was induced by tau peptides
that the formation of A and NFT may be reactive. In rats, [23]. Thus, aggregation of tau may be associated with
both experimental damage or chemically induced lesions of disassembly of A, which could explain the lack of spatial
the nucleus basalis can elevate cortical APP, and intrathecal correlation of the SPs and NFTs [19]. Finally, the notion of
or intraparenchymal injections of toxins can induce APP in an impact of A on NFT formation is supported by studies in
International Journal of Alzheimers Disease 3

APP-transgenic mice reporting that a reduction in endoge- contribute to A clearance by complement activation [45].
nous levels of tau can ameliorate some of the behavioral CD2AP, CD33, BIN1, and PICALM are involved in endo-
and other deficits that are mediated by A [24, 25] and by cytosis (CME), and a recent study [46] showed that several
the discovery that mutations in the tau gene cause autosomal of these factors involved in endocytosis modify A toxicity
dominant frontotemporal lobe dementia with a tau pathol- in glutamatergic neurons of Caenorhabditis elegans and in
ogy similar to the tau pathology seen in AD but without the primary rat cortical neurons. In yeast, A impaired the
appearance of A plaques [26]. Both these observations seem endocytic tracking of a plasma membrane receptor, which
to place tau pathology downstream of amyloid- pathology. was ameliorated by endocytic pathway factors identified in
the yeast screen also providing substantial evidence for a link
between A, endocytosis, and human AD [46]. In summary,
4. Evidence from Genetic Studies convincing evidence for an A-related mechanism exists for
all of these identified LOAD genes, providing a substantial
In particular the genes identified in the late-onset form of amount of support for the ACH in AD.
the disease provide support for the ACH. In general, these
genes are not inherited in a Mendelian but a sporadic fashion.
However, first-degree relatives of patients with late-onset AD 5. Evidence from Clinical Trials Targeting
have twice the expected life time risk of this disease compared A and Tau
to persons without an aected first-degree relative, and late-
The drugs currently used to treat AD (i.e., cholinesterase
onset AD is more frequent among monozygotic than dizy-
inhibitors, NMDA receptor antagonists, and antipsychotic
gotic cotwins, suggesting a substantial genetic contribution
drugs) have limited therapeutic value. New, potentially
to this form of the disease.
disease-modifying, therapeutic approaches are targeting A
The apolipoprotein E (APOE) gene, which was identified
and tau protein. Driven by the ACH, there are currently four
as the first susceptibility gene for late-onset AD, is the major
main therapeutic approaches: (a) reducing the generation of
genetic risk factor (population attributable risk: 20%) [27,
A, (b) facilitating the clearance of A, (c) preventing the
28]. Each APOE-4 allele lowers the age at onset in a dose-
aggregation of A and destabilizing A oligomers, and (d)
dependent fashion [27]. How the dierent APOE proteins
drugs targeting tau [47]. Drugs classes include active and
mediate their eects in AD is not fully clarified, but there
passive immunization directed against A, compounds that
is compelling evidence by PDAPP transgenic mice models
interfere with the secretases regulating A generation from
indicating that APOE mediates the clearance of amyloid-
APP, drugs to prevent A aggregation and destabilize A
[29], with the APOE2, APOE3, and APOE4 isoforms being
oligomers, and drugs targeting tau protein.
increasingly less eective [30]. Consistent with this notion,
the presence of an APOE-4 allele is associated with a higher
A burden in the brains of LOAD patients [31, 32], sug- 5.1. Active and Passive Immunization. Active and passive
gesting that APOE interacts with A by enhancing its depo- immunizations were developed to inhibit generation of toxic
sition in plaques. In various ethnic groups, two haplotypes A aggregates and to remove soluble and aggregated A.
in the sortilin-related receptor (SORL1) gene associated with At least three dierent immune-mediated mechanisms can
LOAD were identified [3337]. SORL1 is involved in track- promote A removal: solubilization by antibody binding to
ing of APP from the cell surface to the golgi-endoplasmic A, phagocytosis of A by microglia, and A extraction from
reticulum complex and -secretase processing of APP [34, the brain by plasma antibodies.
38, 39], also in line with the ACH. Recent large-scale GWA In phase II randomized controlled trials (RCTs) of active
studies performed primarily in samples and populations of immunization of patients with mild-to-moderate AD with
European ancestry detected genetic variants associated the anti-A vaccine AN-1792 (QS-21) most but not all
with AD in complement component (3b/4b) receptor 1 participants developed significant A-antibody titres [48, 49]
(CR1), clusterin (CLU, APOJ), bridging integrator 1 and there was evidence of memory and function improve-
(BIN1), phosphatidylinositol-binding clathrin assembly pro- ment and reduced CSF tau concentrations in patients with
tein (PICALM), EPH receptor A1 (EPHA1), CD33 molecule increased IgG titres [48, 49]. However, in the first trial
(CD33), membrane-spanning 4-domains, subfamily A, patients immunized with AN-1792 had a greater brain
members 4 and 6E (MS4A4/MS4A6E), CD2-associated pro- atrophy rate on MRI than did patients given placebo possibly
tein (CD2AP), and ATP-binding cassette, subfamily A, mem- because of amyloid removal and cerebral fluid shifts. In
ber 7 (ABCA7) [4042]. While these genes remain to addition, several patients developed meningoencephalitis
undergo functional validation, they are functionally plausi- due to a T-cell response. In the follow-up trial, brain volume
ble and also largely consistent with the ACH. Similar and loss in antibody responders was not dierent from that in
additive to APOE, CLU encodes an apolipoprotein and acts patients receiving placebo, and no further cases of menin-
as an A chaperone, regulating the conversion of A to goencephalitis were found [49]. Responders maintained low,
insoluble forms and A toxicity thereby promoting amyloid but detectable, anti-AN-1792 antibody titres at about 4.6
plaque formation [43]. ABCA7 is involved in the eux of years after immunization and had significantly reduced func-
lipids from cells to lipoprotein particles, such as APOE and tional decline compared with placebo-treated patients [49].
CLU, and in addition regulates APP processing and inhibits In addition, immunization with anti-AN-1792 antibody
-amyloid secretion [44]. There is evidence that CR1 may could completely remove amyloid plaques as determined by
4 International Journal of Alzheimers Disease

postmortem assessment although patients still had end-stage a monoclonal antibody that targets specifically soluble A,
dementia symptoms before death. promotes A clearance from the brain through the blood. In
In order to avoid neuroinflammation and neurotoxicity, a phase II RCT, there was a correlation between total plasma
new vaccines that selectively target B-cell epitopes have been A142 after treatment (dose-dependent increase), baseline
developed. CAD-106, which consists of the immunodrug amyloid plaque burden shown by single-photon emission
carrier Qb coupled with a fragment of the A16 peptide, CT scanning, and a dose-dependent increase in unbound
could in animal studies induce A-specific antibodies and CSF A142 , suggesting that solanezumab might mobilize
reduce amyloid accumulation without stimulating T cells. A142 from plaques and might normalize soluble CSF
In patients with mild-to-moderate AD, CAD-106 induced A142 in patients with AD [56]. Consequently, two phase
a substantial anti-A IgG response and was well tolerated III RCTs have been initiated (NCT00905372, NCT00904683,
[50], confirmatory phase II RCTs are ongoing (NCT01097 NCT01127633). PF-04360365 is a modified IgG2 antibody
096, NCT01023685, NCT00795418, NCT00956410, and that binds to the C terminus of A140 . Preliminary results
NCT00733863). ACC-001 is an A16 fragment derived from on a single-dose regimen indicate that this antibody is well
the N-terminal B cell epitope of A and conjugated to tolerated in patients with AD [57]. Currently, two phase
the mutated diphtheria toxin protein CRM19. It is being II RCTs of multiple doses are ongoing (NCT00722046 and
studied in phase II RCTs (NCT00479557, NCT01284387, NCT00945672). GSK-933776, R-1450 (RO-4909832), and
NCT01227564, NCT00498602, NCT00752232, NCT00955 MABT-5102A are monoclonal antibodies that target A and
409, NCT01238991, NCT00960531, NCT00959192). ACI- have been tested in patients with AD in phase I and phase
24 is a vaccine that contains A115 closely apposed to the II trials (NCT01424436, NCT00459550, NCT01224106,
surface of the liposome. It reduced brain amyloid load and NCT00531804, NCT00736775, NCT00997919, NCT0134
restored memory deficits in mice [51] and is entering a phase 3966, and NCT01397578).
II RCT. Vaccines that are currently being tested in phase I Passive immunization [58] can also be achieved by
RCTs are V-950 (NCT00464334; an aluminium-containing intravenous infusion of immunoglobulins (IVIg), from
adjuvant with or without ISCOMATRIX (CSL Behring, PA, healthy donors, which include naturally occurring polyclonal
USA, a biological adjuvant of saponin, cholesterol, and anti-A antibodies. IVIg is already approved as therapy
phospholipids) and UB-311 (NCT00965588), a vaccine in for immune deficiency, with good safety and tolerability
which the immunogen A114 is associated with the UBITh evidence. In two small studies, short-term immunoglobulin
peptide (United Biomedical, NY, USA) and a mineral salt administration in patients with AD was well tolerated,
suspension adjuvant [52]. promoted a decrease of total A CSF concentrations, and
Atopes, which are short peptides mimicking parts of increased plasma total A concentrations [59, 60], with
native A142 , represent an alternative active immunization evidence of improvement or stabilization of cognitive func-
strategy. The atopes AD-01 and AD-02 target the N- tions. Preliminary data from a phase II RCT confirmed
terminal A fragment and both had disease-modifying prop- the positive eects on cognition [61], a phase III study is
erties in animal models of AD [53]. Results of recent phase ongoing (NCT00818662). In summary, the RCTs on active
I RCTs indicate that both are safe and well tolerated and passive immunization agents consistently show an eect
(NCT00495417, NCT00633841, and NCT00711139) [53]. on amyloid clearance, and several but not all phase II RCTs
Atope AD-02 recently progressed to phase II clinical testing show promising eects on cognition.
(NCT01117818).
Passive immunotherapy is based on monoclonal anti- 5.2. Drugs to Reduce A Generation from APP. BACE1 (-
bodies or polyclonal immunoglobulins targeting A to secretase) initiates the amyloidogenic pathway. Pioglitazone
promote its clearance. Animal studies have shown that and rosiglitazone are thiazolidinediones and drugs com-
anti-A antibodies can prevent oligomer formation and monly used to treat type II diabetes. They happen to act as
reduce brain amyloid load with improvement in cognitive BACE1 inhibitors through stimulating the nuclear peroxi-
functions [54]. Several monoclonal antibodies are cur- some proliferator-activated receptor (PPAR). Activation
rently being tested: bapineuzumab (AAB-001), solanezumab of PPAR receptors, in turn, can suppress expression of
(LY-2062430), PF-04360365, GSK-933776, R-1450 (RO- BACE1 and APP and can promote APP degradation by
4909832), and MABT-5102A. A phase II RCT of bapin- increasing its ubiquitination [62]. In addition to their eects
euzumab in patients with mild-to-moderate AD that had on BACE1, therapeutic eects of PPAR agonists in AD could
a follow-up period of longer than 18 months reported no be caused by their eect on insulin action. Both rosiglitazone
significant eects on the primary measures of cognition and pioglitazone increase peripheral insulin sensitivity and
or activities of daily living, as measured in prespecified reduce concentrations of insulin. Insulin, in turn, competes
within-dose cohort analyses. However, post hoc anal- yses with A for degradation by the insulin-degrading enzyme
of clinical and neuroimaging data from all dose cohorts [62].
showed nonsignificant improvements in cognitive end- There are only few phase III RCTs, which likely reflects
points and signs of ecacy in APOE 4 noncarriers [55]. the diculty in development of BACE1 targeting agents.
Phase III studies are ongoing, including separate RCTs BACE1 has many substrates including several with phys-
for APOE 4 carriers and non-carriers (NCT00574132, iologically important functions such as neuregulin-1 that
NCT00996918, NCT00998764, NCT00667810, NCT005 is involved in myelination, and drugs must cross the
75055, NCT00676143, and NCT00937352). Solanezumab, blood-brain barrier in order to modulate BACE1 function.
International Journal of Alzheimers Disease 5

Pioglitazone can cross the blood-brain barrier although in phase III RCTs in patients with mild AD but did not
whether rosiglitazone can reach the CNS in human beings is show clinical eects [71] possibly due to low -secretase
unclear [62]. Out of the RCTs that have explored the eects of modulator potency, poor CNS penetration, or inhibition
pioglitazone and rosiglitazone on cognition in patients with of microglia-mediated A clearance by residual NSAID
AD or MCI (NCT00982202, NCT00736996, NCT00550420, activity. Another -secretase modulator, CHF-5074, reduced
NCT00428090, NCT00348309, NCT00242593, NCT00265 A brain load and improved behavioral deficits in animals
148, NCT00348140, NCT00334568, and NCT00490568), [72] and has reached phase II clinical testing (NCT01303744
only three (NCT00982202, NCT00428090, and NCT002651 and NCT01421056).
48) have reported results to date, and these were negative Upregulation of -secretase activity, leading to non-
[63]. Currently, several new -secretase inhibitors are under amyloidogenic cleavage of APP, can decrease A formation
investigation. Of these, CTS-21166, an orally administered and increase production of a potentially neuroprotective
compound, was well tolerated and reduced plasma A soluble domain (sAPP) [73]. Several drugs can stimulate -
concentrations in mice [64] and has proceeded to phase I secretase (agonists of muscarinic, glutamate, and serotonin
clinical testing [65]. receptors; statins; oestrogens; testosterone; protein kinase
Development of drugs targeting -secretase, the enzyme C activators) and have been tested in clinical trials, but
responsible for the final step in A generation, presents no conclusive results are available yet [74]. These -
challenges similar to those for -secretase inhibitors as - secretase modulators include Exebryl-1, which modulates
secretase is one of the main complexes involved in intram- - and -secretase activity causing substantial reduction
embranous cleavage of several proteins, including APP, of A formation and accumulation in the mouse brain
Notch receptor, and various neuronal substrates [66]. As a with memory improvements (a phase I RCT was approved
consequence, adverse eects of -secretase inhibitors include in 2008) [75], Etazolate (EHT-0202), a selective GABAA
hematological and gastrointestinal toxicity, skin reactions, receptor modulator that stimulates neuronal -secretase and
and changes to hair color, mainly caused by inhibition increases sAPP production [76] and has been recently
of the Notch signaling pathway, which is involved in cell tested in a phase II RCT in patients with mild-to-moderate
dierentiation. AD (NCT00880412) [77], and Bryostatin-1, a macrocyclic
Phase III trials for the Notch-inhibiting drug semagace- lactone that can stimulate -secretase by activating protein
stat failed. Preliminary findings showed that semagacestat kinase C and promoting sAPP secretion [78] reducing
not only failed to slow disease progression, but also was brain A140 and A142 and improving behavioral outcomes
associated with worsening of clinical measures of cognition in mouse models of AD [78] (phase II study in process
and the ability to perform activities of daily living and a (NCT00606164)).
higher incidence of skin cancer in the treatment group
than the placebo group. However, several Notch-sparing -
secretase inhibitors (second-generation inhibitors) are cur- 5.3. Drugs to Prevent A Aggregation and Destabilize A
rently under development: begacestat was tested in a phase Oligomers. Compounds that inhibit A aggregation or
I RCT (NCT00959881) and BMS-708163 in two phase II destabilize A oligomeric species can act twofold: (a) either
RCTs in patients with prodromal or mild-to-moderate AD they bind to A monomers thereby preventing oligomeriza-
(NCT00810147 and NCT00890890). Begacestat reduced A tion and allowing A elimination, or (b) they react with A
concentrations in the plasma (with delayed rebound) [67] oligomers thereby neutralizing their toxicity and promoting
but did not substantially aect CSF A140 , whereas BMS- their clearance. They are chemically heterogeneous and also
708163 promoted a dose-dependent decrease of A140 in here the challenge is to develop agents that can cross the
the CSF [68]. Results from animal studies testing PF-3084014 blood brain barrier and have low toxicity.
showed decreases in A in the plasma, CSF, and brain, The first generation of nonpeptidic antiaggregates failed
without a rebound eect on plasma A [69]. In a subsequent to fulfill these criteria. Tramiprosate (3APS), which main-
small phase I study, PF-3084014 promoted a dose-dependent tains A in the nonfibrillar state by binding to soluble
reduction in plasma A concentrations although eects on form, showed negative results in the Alphase study, a phase
CSF concentrations were small [70]. NIC515, a naturally III RCT [79] although previous experimental and phase
occurring monosaccharide found in many foods, can act as II trials had been promising [80]. Although there are
a Notch-sparing -secretase inhibitor and insulin sensitizer several possible reasons for this failure, including variability
(i.e., it increases the sensitivity of the tissue to insulin). It is among study sites, dierences in the treatment and control
currently being tested in patients with AD in a phase II study groups because of the concomitant treatment with cognitive-
(NCT00470418). enhancing drugs, and low CNS bioavailability of the drug, a
-secretase modulators can selectively block APP pro- European phase III RCT with tramiprosate was terminated
teolysis without Notch-based adverse eects. A subset of as a consequence of the negative findings.
nonsteroidal anti-inflammatory drugs (NSAIDs), including Clioquinol (PBT1) inhibits A aggregation through
ibuprofen, indomethacin, and sulindac sulfide, bind to APP interfering with interactions between A, copper, and zinc.
and act as -secretase modulators, decreasing A140 and Studies in Tg2576 mice and human volunteers showed that
A142 production, with increased generation of A138 CQ entry into the brain is limited although upon brain
fragments. Among these compounds, known as selective entry it binds to amyloid plaques [81]. PBT1 showed positive
-amyloid-lowering agents (SALAs), tarenflurbil was tested results in phase II RCTs but further phase II/III studies
6 International Journal of Alzheimers Disease

were halted due to manufacturing toxicity issues [82]. The neurotrophic factors, and hindering A toxicity [89]. How-
second-generation inhibitor, PBT2, has a greater blood- ever, a small RCT with lithium (10 weeks, including a 6-week
brain barrier permeability than does clioquinol, and animal titration phase) in patients with mild AD did not show any
experiments showed that PBT2 prevents A oligomerization, cognitive benefit or any change in CSF biomarkers, including
promotes A oligomer clearance, reduces soluble and insol- phosphorylated tau, total tau, and A142 [90].
uble brain A, decreases plaque burden, and has positive The AD Cooperative Study (ADCS) of valproate was
eects on cognition [82]. A 12-week, phase II RCT in designed to determine whether chronic valproate treatment
patients with mild AD, was consistent with these findings, could delay the onset of behavioral symptoms in outpatients
PBT2 reduced A142 CSF concentrations and improved with mild-to-moderate AD; a secondary aim was to test
executive function [83]. Scyllo-inositol (ELND-005) is an whether valproate can delay cognitive and functional decline.
orally administered stereoisomer of inositol that can cross the No eects on cognition and functional status were reported,
blood-brain barrier using inositol transporters. By binding but incidence of agitation and psychosis seemed to be
to A, it modulates its misfolding, inhibits its aggregation reduced [89].
and stimulates dissociation of aggregates. It was successful in Several GSK3 inhibitors are under development. NP-
animal studies, reducing brain concentrations of soluble and 031112 (NP-12) is a thiadiazolidinone-derived compound, a
insoluble A140 and A142 , plaque burden, synaptic loss, non-ATP competitive inhibitor of GSK3, which can reduce
and glial inflammatory reaction and significantly improving brain concentrations of phosphorylated tau and amyloid
spatial memory function [84]. It is currently being tested in deposition and prevent neuronal death and cognitive deficits
phase II RCTs (NCT00568776 and NCT00934050). However, in animals [91]. This drug has been tested in patients with
because of serious adverse events among patients in the AD in a phase II RCT (NCT00948259); no results have yet
two high-dose groups (1000 mg and 2000 mg twice daily), been published.
these doses have been removed from the RCT, and the study Methylthioninium chloride (methylene blue), a widely
continues restricted to patients who are assigned the lower used histology dye, acts as a tau antiaggregate [92]. This
dose (250 mg twice daily) and placebo. Epigallocatechin- compound also has antioxidant properties, enhances mito-
3-gallate (EGCg), a polyphenol from green tea, induces chondrial function [93], and was eective, alone and in
-secretase and prevents A aggregation in animals by combination with rivastigmine, in reversing learning deficits
directly binding to the unfolded peptide [85]. In addition, it and hyoscine-induced memory impairments in animals [94].
modulates signal transduction pathways, expression of genes Dierent doses of methylthioninium chloride (up to 100 mg)
regulating cell survival and apoptosis, and mitochondrial were tested in a phase II study in patients with moderate
function [85]. It is currently being tested in a phase II/III AD. The group given the 60 mg dose had improved cognitive
RCT in patients with early AD. function and, after 1 year, evidence of slower disease
progression compared with placebo [95]. The ineectiveness
5.4. Drugs to Target Tau Protein. Tau is a cytoplasmatic pro- in the group on the 100 mg dose was attributed to drug
tein that binds to tubulin during its polymerisation, stabilis- formulation defects, limiting release. A new formulation
ing microtubules. In AD, tau is abnormally phosphorylated, (leuco-methylthioninium), with a higher bioavailability, was
resulting in the generation of aggregates (neurofibrillary recently announced [96], and phase III RCTs are needed to
tangles) toxic to neurons. The hypothesis that tau pathology confirm its safety and clinical ecacy.
causes AD has been the main competitor of the amyloid Davunetide (AL-108, NAP), an intranasally adminis-
hypothesis [86]. However, only one tau-directed compound tered, eight-aminoacid peptide fragment derived from the
(valproate; valproic acid) has so far reached phase III RCT, activity-dependent neuroprotective protein, and AL-208, an
with disappointing results because there were no eects on intravenous formulation of Davunetide, are being developed.
cognition and functional status [87]. Davunetide has been tested in animal models of AD and
There are two main therapeutic approaches to target tauopathy, and its neuroprotective activity includes regula-
the tau protein: modulation of tau phosphorylation with tion of microtubule dynamics, as well as inhibition of tau
inhibitors of tau-phosphorylating kinases and compounds hyperphosphorylation and protection against A toxicity
that inhibit tau aggregation and/or promoting aggregate [97, 98]. Davunetide was studied in patients with amnestic
disassembly. The first approach is based on the observation mild cognitive impairment in a 12-week, phase II RCT
that tau hyperphosphorylation and neurofibrillary tangle and was safe and well tolerated and had positive eects on
formation can be promoted by imbalanced activity of protein cognition [99], although confirmatory studies are needed.
kinases (glycogen-synthase-kinase-3 (GSK3) and p70-S6- Nicotinamide is the biologically active form of niacin
kinase) and the phosphatase PP2A [88]. GSK3 deregulation (vitamin B3) and the precursor of coenzyme NAD+. Orally
might have a role in AD pathogenesis because GSK3 is administered nicotinamide can prevent cognitive deficits in a
involved in tau and amyloid processing, cellular signaling, mouse model of AD and can reduce brain concentrations of a
and gene transcription [88]. species of phosphorylated tau (Thr231) that inhibits micro-
Both lithium and valproate, well known for the treatment tubule polymerization [100]. Furthermore, nicotinamide
of psychiatric disorders, inhibit GSK3,to reduce tau phos- inhibits brain sirtuin deacetylase and upregulates acetyl--
phorylation and prevent or reverse aspects of tauopathy in tubulin, protein p25, and MAP2c; all these interactions are
animal models [89]. Both drugs can also be neuroprotective associated with increased microtubule stabilization [100].
by upregulating the antiapoptotic factor BCL2, inducing Nicotinamide has been used in several clinical studies,
International Journal of Alzheimers Disease 7

Table 1: Issues of RCTs of AD.


Issue Possible solution
Subjects
Target group selection: patients with AD have various types of
Criteria for identifying subgroups with more homogeneous biomarker
neuropathology (i.e., amyloid plaques, NFTs, infarcts, and Lewy
evidence of AD pathology are needed to facilitate RCTs
bodies)
In patients with mild-to-moderate AD, the disease could be too RCTs that include patients with early AD might enable detection of
advanced for a disease-modifying eect of a specific drug (e.g., disease-modifying eects; investigation into which stage of the AD
immunotherapy) process a therapeutic strategy is more eective is warranted
Agents
Choosing the right drug: compounds with positive results in
Robust proof-of-concept studies should be mandatory
preclinical and early clinical testing failed in large phase III
Investigators should take into account class ecacy
RCTs, with costly losses (e.g., tramiprosate)
Use of drug-related biomarkers in preclinical and early clinical stages
can help to confirm the target engagement and to assure early
withdrawal of ineective drugs
Some RCTs were likely hindered by the inability to reach a
Optimization of drug dosage and treatment duration based on
therapeutic dosage (e.g., tarenflurbil) or short treatment
pharmacokinetics
duration
Personalized therapeutic approach: considering genetic polymorphisms
Genetics: polymorphisms (e.g., APOE,) might aect drug
that aect drug response can help to optimize drug dosage (e.g.,
response
increased doses for individuals with a rapid metabolism)
Outcome measurements
Development and use of relevant, reliable, multidimensional measures
for clinical (cognitive and functional) endpoints are key factors, as well
Measuring eects: many RCTs are developed according to the
the use of biomarkers (neuroimaging, CSF, or blood molecules) that
design of AChEI RCTs, an approach that has indicated the
reliably and quantitatively correlate with disease progression; collection
AChEI symptomatic eect but is not sensitive in detecting the
of baseline data (clinical, biomarkers) that can be used as reference to
ecacy of disease-modifying drugs, rating scales used may have
interpret later findings is advisable; for early AD (i.e., mild cognitive
low sensitivity for changes and/or the drug type assessed and
impairment), self-rated and observer-rated assessments of activities of
these tools have a subjective component
daily living, instrumental activities of daily living, and quality of life are
recommended
Adequate training and monitoring of RCT raters to maximize
homogeneous recruitment of patients, reduce variance, and guarantee a
Unreliable evaluation of patients by RCT raters
more accurate rating; eective implementation of quality control on
data at research sites
Optimization of resources
Consistency: multicenter RCTs done in several countries can
have cultural and linguistic issues with assessment scales (e.g., Multicenter trials should use centers of excellence that are already
translation, validation), as well as infrastructure problems experienced in RCTs to minimize intersite and intercountry variability
(technological disparities between centers)
More collaboration between pharmaceutical companies and clinical
Unsuccessful preclinical and clinical studies are often not
researchers, with information sharing, can lead to more standardized
published leading to repetition of unsuccessful trials or errors
RCT protocols, reduction of errors, and decreased costs

including RCTs in patients with neurodegenerative disor- 6. Conclusions


ders, and is generally safe and well tolerated; a phase II
RCT is ongoing in patients with mild-to-moderate AD Overall, there is substantial evidence supporting a role of the
(NCT00580931). ACH in AD. However, the available results from RCTs are
What do these trials tell us? Sadly, they leave little not in line with previous optimistic predictions of an immi-
certainty. Amyloid immunization teaches us that we can nent breakthrough in development of a disease-modifying
massively reduce amyloid burden, but when administered therapy. To explain the disappointing results of several RCTs,
late in the disease, it is not a miracle cure. It may have researchers have highlighted various potential issues, both in
clinically relevant benefits and it may lead to better outcomes drug choice and development programs. Table 1 summarizes
if it is given early in the disease or presymptomatically but we these and provides possible solutions. Clinical trials need to
simply do not have data to address these issues. be organized in those in the very earliest stages of the disease.
Whether this can be carried out genetically (e.g., by using
E4 homozygotes) or by PIB imaging or some combination
8 International Journal of Alzheimers Disease

of both is not clear. Of course, it could be argued that even amyloid fibril protein, Biochemical and Biophysical Research
persons who show PIB signals are already too far down Communications, vol. 122, no. 3, pp. 11311135, 1984.
the disease progression for disease-modifying therapy and [4] A. Goate, M. C. Chartier-Harlin, M. Mullan et al., Segrega-
that treatment needs to be initiated even before this stage. tion of a missense mutation in the amyloid precursor protein
Certainly, even those with mild AD have profound cell loss. gene with familial Alzheimers disease, Nature, vol. 349, no.
6311, pp. 704706, 1991.
In addition, it would be helpful to perform antiamyloid
[5] E. Levy-Lahad, W. Wasco, P. Poorkaj et al., Candidate gene
trials in individuals with APP and PSEN mutations or those
for the chromosome 1 familial Alzheimers disease locus,
with Downs syndrome as they provide the best test of the Science, vol. 269, no. 5226, pp. 973977, 1995.
ACH hypothesis. Biomarker studies should be included in [6] R. Sherrington, E. I. Rogaev, Y. Liang et al., Cloning of
trial designs so that the researchers can form, as clearly as a gene bearing missense mutations in early-onset familial
possible, informed opinions as to whether the drug has hit Alzheimers disease, Nature, vol. 375, no. 6534, pp. 754760,
the proposed target. 1995.
However, in addition to implementing new guidelines in [7] B. I. Giasson, V. M. Y. Lee, and J. Q. Trojanowski, Interac-
preclinical and clinical phases of drug development, several tions of amyloidogenic proteins, Neuromolecular Medicine,
additional issues are key to validate the ACH and successfully vol. 4, no. 1-2, pp. 4958, 2003.
develop therapeutic targets. From a molecular point of view, [8] J. A. Hardy and G. A. Higgins, Alzheimers disease: the
we need a focused eort to fully understand the functions amyloid cascade hypothesis, Science, vol. 256, no. 5054, pp.
184185, 1992.
of APP and A and to answer the two key questions:
[9] D. J. Selkoe, The molecular pathology of Alzheimers dis-
does A in fact influence tau phosphorylation and, if yes, ease, Neuron, vol. 6, no. 4, pp. 487498, 1991.
does tau phosphorylation in fact lead to dementia? Second, [10] S. M. Gentleman, M. J. Nash, C. J. Sweeting, D. I. Graham,
we need to understand the nature of disease propagation: and G. W. Roberts, -Amyloid precursor protein (APP) as
is permissive templating of A [101, 102] and tau [103] a marker for axonal injury after head injury, Neuroscience
the reason for both the characteristic neuroanatomy of Letters, vol. 160, no. 2, pp. 139144, 1993.
the disease [104] and the reason that the disease seems [11] J. E. McKenzie, S. M. Gentleman, G. W. Roberts, D. I.
to become self-propagating once it has started [105, 106]?. Graham, and M. C. Royston, Increased numbers of APP-
Finally, it makes sense to pursue other targets beyond A immunoreactive neurones in the entorhinal cortex after head
as there is substantial evidence for additional potential injury, NeuroReport, vol. 6, no. 1, pp. 161164, 1994.
pathways increasing disease susceptibility, among these lipid [12] G. W. Roberts, S. M. Gentleman, A. Lynch, L. Murray, M.
metabolism and inflammatory processes [107]. Landon, and D. I. Graham, Amyloid protein deposition
in the brain after severe head injury: implications for the
pathogenesis of Alzheimers disease, Journal of Neurology
Conflict of Interests Neurosurgery and Psychiatry, vol. 57, no. 4, pp. 419425,
1994.
The author does not have any actual or potential conflict of [13] R. N. Kalaria and G. Perry, Amyloid P component and other
interests. acute-phase proteins associated with cerebellar A -deposits
in Alzheimers disease, Brain Research, vol. 631, no. 1, pp.
151155, 1993.
Acknowledgments [14] B. Regland and C. G. Gottfries, The role of amyloid -
protein in Alzheimers disease, The Lancet, vol. 340, no.
This work was supported by grants from the National 8817, pp. 467469, 1992.
Institute of Health and the National Institute on Aging, R37- [15] K. Renkawek, W. W. de Jong, K. B. Merck, C. W. G. M.
AG15473, P01-AG07232, the Blanchette Hooker Rockefeller Frenken, F. P. A. van Workum, and G. J. Bosman, Alpha B-
Foundation, the Charles S. Robertson Gift from the Banbury crystallin is present in reactive glia in Creutzfeldt-Jakob
Fund, and the Merrill Lynch Foundation. Dr. Reitz was disease, Acta Neuropathologica, vol. 83, no. 3, pp. 324327,
further supported by a Paul B. Beeson Career Development 1992.
[16] W. C. Wallace, V. Bragin, N. K. Robakis et al., Increased
Award (K23AG034550).
biosynthesis of Alzheimer amyloid precursor protein the in
cerebral cortex of rats with lesions of the nucleus basalis of
References Meynert, Molecular Brain Research, vol. 10, no. 2, pp. 173
178, 1991.
[1] About a peculiar disease of the cerebral cortex. By Alois [17] R. Yan, M. J. Blenkowski, M. E. Shuck et al., Membrane-
Alzheimer, 1907 (Translated by L. Jarvik and H. Greenson), anchored aspartyl protease with Alzheimers disease -
Alzheimer Disease and Associated Disorders, vol. 1, no. 1, pp. secretase activity, Nature, vol. 402, no. 6761, pp. 533537,
38, 1987. 1999.
[2] K. L. Newell, B. T. Hyman, J. H. Growdon, and E. T. Hedley- [18] R. M. Torack and J. W. Miller, Immunoreactive changes
Whyte, Application of the National Institute on Aging resulting from dopaminergic denervation of the dentate
(NIA)-Reagan Institute criteria for the neuropathological gyrus of the rat hippocampal formation, Neuroscience
diagnosis of Alzheimer disease, Journal of Neuropathology Letters, vol. 169, no. 1-2, pp. 912, 1994.
and Experimental Neurology, vol. 58, no. 11, pp. 11471155, [19] R. A. Armstrong, D. Myers, and C. U. M. Smith, The spatial
1999. patterns of plaques and tangles in Alzheimers disease do not
[3] G. G. Glenner and C. W. Wong, Alzheimers disease and support the cascade hypothesis, Dementia, vol. 4, no. 1, pp.
Downs syndrome: sharing of a unique cerebrovascular 1620, 1993.
International Journal of Alzheimers Disease 9

[20] C. Duyckaerts, Looking for the link between plaques and ease, Current Neurology and Neuroscience Reports, vol. 8, no.
tangles, Neurobiology of Aging, vol. 25, no. 6, pp. 735739, 5, pp. 384391, 2008.
2004. [36] J. H. Lee, R. Cheng, N. Schupf et al., The association be-
[21] R. A. Armstrong, Plaques and tangles and the pathogenesis tween genetic variants in SORL1 and Alzheimer disease in an
of Alzheimers disease, Folia Neuropathologica, vol. 44, no. 1, urban, multiethnic, community-based cohort, Archives of
pp. 111, 2006. Neurology, vol. 64, no. 4, pp. 501506, 2007.
[22] J. Busciglio, A. Lorenzo, J. Yeh, and B. A. Yankner, - [37] C. Reitz, R. Cheng, E. Rogaeva et al., Meta-analysis of
Amyloid fibrils induce tau phosphorylation and loss of the association between variants in SORL1 and Alzheimer
microtubule binding, Neuron, vol. 14, no. 4, pp. 879888, disease, Archives of Neurology, vol. 68, no. 1, pp. 99106,
1995. 2011.
[23] M. Perez, R. Cuadros, M. J. Benitez, and J. S. Jimenez, Inter- [38] R. F. Lane, S. M. Raines, J. W. Steele et al., Diabetes-
action of Alzheimers disease amyloid peptide fragment 25 associated SorCS1 regulates Alzheimers amyloid- me-
35 with tau protein, and with a tau peptide containing the tabolism: evidence for involvement of SorL1 and the re-
microtubule binding domain, Journal of Alzheimers Disease, tromer complex, Journal of Neuroscience, vol. 30, no. 39, pp.
vol. 6, no. 5, pp. 461467, 2004. 1311013115, 2010.
[24] E. D. Roberson, B. Halabisky, J. W. Yoo et al., Amyloid- [39] C. Reitz, S. Tokuhiro, L. N. Clark et al., SORCS1 alters amy-
/fyn-induced synaptic, network, and cognitive impair- loid precursor protein processing and variants may increase
ments depend on tau levels in multiple mouse models of Alzheimers disease risk, Annals of Neurology, vol. 69, no. 1,
Alzheimers disease, Journal of Neuroscience, vol. 31, no. 2, pp. 4764, 2011.
pp. 700711, 2011. [40] D. Harold, R. Abraham, P. Hollingworth et al., Genome-
[25] E. D. Roberson, K. Scearce-Levie, J. J. Palop et al., Reducing wide association study identifies variants at CLU and
endogenous tau ameliorates amyloid -induced deficits in PICALM associated with Alzheimers disease, Nature Genet-
an Alzheimers disease mouse model, Science, vol. 316, no. ics, vol. 41, no. 10, pp. 10881093, 2009.
5825, pp. 750754, 2007. [41] J. C. Lambert, S. Heath, G. Even et al., Genome-wide asso-
[26] M. Hutton, C. L. Lendon, P. Rizzu et al., Association of mis- ciation study identifies variants at CLU and CR1 associated
sense and 5-splice-site mutations in tau with the inherited with Alzheimers disease, Nature Genetics, vol. 41, no. 10, pp.
dementia FTDP-17, Nature, vol. 393, no. 6686, pp. 702705, 10941099, 2009.
1998. [42] S. Seshadri, A. L. Fitzpatrick, M. A. Ikram et al., Genome-
[27] E. H. Corder, A. M. Saunders, W. J. Strittmatter et al., Gene wide analysis of genetic loci associated with Alzheimer
dose of apolipoprotein E type 4 allele and the risk of Alz- disease, Journal of the American Medical Association, vol. 303,
heimers disease in late onset families, Science, vol. 261, no. no. 18, pp. 18321840, 2010.
5123, pp. 921923, 1993. [43] R. B. DeMattos, J. R. Cirrito, M. Parsadanian et al., ApoE and
[28] A. J. C. Slooter, M. Cruts, S. Kalmijn et al., Risk estimates of clusterin cooperatively suppress A levels and deposition:
dementia by apolipoprotein E genotypes from a population- evidence that ApoE regulates extracellular A metabolism in
based incidence study: the rotterdam study, Archives of vivo, Neuron, vol. 41, no. 2, pp. 193202, 2004.
Neurology, vol. 55, no. 7, pp. 964968, 1998. [44] P. Hollingworth, D. Harold, R. Sims et al., Common variants
[29] D. M. Holtzman, K. R. Bales, T. Tenkova et al., Apolipopro- at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP
tein E isoform-dependent amyloid deposition and neuritic are associated with Alzheimers disease, Nature Genetics, vol.
degeneration in a mouse model of Alzheimers disease, 43, no. 5, pp. 429436, 2011.
Proceedings of the National Academy of Sciences of the United [45] T. Wyss-Coray, F. Yan, A. H. T. Lin et al., Prominent
States of America, vol. 97, no. 6, pp. 28922897, 2000. neurodegeneration and increased plaque formation in
[30] J. Kim, J. M. Basak, and D. M. Holtzman, The role of apoli- complement-inhibited Alzheimers mice, Proceedings of the
poprotein E in Alzheimers disease, Neuron, vol. 63, no. 3, National Academy of Sciences of the United States of America,
pp. 287303, 2009. vol. 99, no. 16, pp. 1083710842, 2002.
[31] G. W. Rebeck, J. S. Reiter, D. K. Strickland, and B. T. Hyman, [46] S. Treusch, S. Hamamichi, J. L. Goodman et al., Func-
Apolipoprotein E in sporadic Alzheimers disease: allelic tional links between A troxicity, endocytic tracking, and
variation and receptor interactions, Neuron, vol. 11, no. 4, Alzheimers disease risk factors in yeast, Science, vol. 334, no.
pp. 575580, 1993. 6060, pp. 12411245, 2011.
[32] D. E. Schmechel, A. M. Saunders, W. J. Strittmatter et al., [47] F. Mangialasche, A. Solomon, B. Winblad, P. Mecocci, and
Increased amyloid -peptide deposition in cerebral cortex M. Kivipelto, Alzheimers disease: clinical trials and drug
as a consequence of apolipoprotein E genotype in late-onset development, The Lancet Neurology, vol. 9, no. 7, pp. 702
Alzheimer disease, Proceedings of the National Academy of 716, 2010.
Sciences of the United States of America, vol. 90, no. 20, pp. [48] S. Gilman, M. Koller, R. S. Black et al., Clinical eects
96499653, 1993. of A immunization (AN1792) in patients with AD in an
[33] Y. Meng, J. H. Lee, R. Cheng, P. St George-Hyslop, R. Mayeux, interrupted trial, Neurology, vol. 64, no. 9, pp. 15531562,
and L. A. Farrer, Association between SORL1 and Alz- 2005.
heimers disease in a genome-wide study, NeuroReport, vol. [49] B. Vellas, R. Black, L. J. Thal et al., Long-term follow-up
18, no. 17, pp. 17611764, 2007. of patients immunized with AN1792: reduced functional
[34] E. Rogaeva, Y. Meng, J. H. Lee et al., The neuronal sortilin- decline in antibody responders, Current Alzheimer Research,
related receptor SORL1 is genetically associated with Alz- vol. 6, no. 2, pp. 144151, 2009.
heimer disease, Nature Genetics, vol. 39, no. 2, pp. 168177, [50] B. Winblad, L. Minthon, A. Floesser et al., Results of
2007. the first-in-man study with the active A immunotherapy
[35] J. H. Lee, S. Barral, and C. Reitz, The neuronal sortilin- CAD106 in Alzheimer patients, Alzheimers & Dementia, vol.
related receptor gene SORL1 and late-onset Alzheimers dis- 5, no. 4, supplement 1, pp. P113P114, 2009.
10 International Journal of Alzheimers Disease

[51] A. Muhs, D. T. Hickman, M. Pihlgren et al., Liposomal [65] J. J. N. Tang, Beta-secretase as target for amyloid-reduction
vaccines with conformation-specific amyloid peptide anti- therapy, Alzheimers Dement, vol. 5, no. 4, supplement 1, p.
gens define immune response and ecacy in APP transgenic P74, 2009.
mice, Proceedings of the National Academy of Sciences of the [66] T. Tomita, Secretase inhibitors and modulators for
United States of America, vol. 104, no. 23, pp. 98109815, Alzheimers disease treatment, Expert Review of
2007. Neurotherapeutics, vol. 9, no. 5, pp. 661679, 2009.
[52] C. Y. Wang, C. L. Finstad, A. M. Walfield et al., Site-specific [67] S. Jacobsen, T. A. Comery, A. Kreft et al., GSI-953 is a
UBITh amyloid- vaccine for immunotherapy of Alzheimers potent APP-selective -secretase inhibitor for the treatment
disease, Vaccine, vol. 25, no. 16, pp. 30413052, 2007. of Alzheimers disease, Alzheimers & Dementia, vol. 5, no. 4,
[53] A. Schneeberger, M. Mandler, O. Otava, W. Zauner, F. supplement 1, p. P139, 2009.
Mattner, and W. Schmidt, Development of AFFITOPE [68] B. P. Imbimbo, Alzheimers disease: -secretase inhibitors,
vaccines for Alzheimers Disease (AD)from concept to Drug Discovery Today, vol. 5, no. 3, pp. 169175, 2008.
clinical testing, Journal of Nutrition, Health and Aging, vol. [69] T. A. Lanz, K. M. Wood, K. E. G. Richter et al., Pharmaco-
13, no. 3, pp. 264267, 2009. dynamics and pharmacokinetics of the -secretase inhibitor
[54] D. M. Wilcock and C. A. Colton, Anti-amyloid- im- PF-3084014, Journal of Pharmacology and Experimental
munotherapy in Alzheimers disease: relevance of transgenic Therapeutics, vol. 334, no. 1, pp. 269277, 2010.
mouse studies to clinical trials, Journal of Alzheimers [70] H. Soares, N. Raha, M. Sikpi et al., A variability and eect
Disease, vol. 15, no. 4, pp. 555569, 2008. of secretase inhibition on cerebrospinal fluid levels of A
[55] S. Salloway, R. Sperling, S. Gilman et al., A phase 2 multiple in healthy volunteers, Alzheimers & Dementia, vol. 5, no. 4,
ascending dose trial of bapineuzumab in mild to moderate supplement 1, pp. P252P253, 2009.
Alzheimer disease, Neurology, vol. 73, no. 24, pp. 20612070, [71] R. C. Green, L. S. Schneider, D. A. Amato et al., Eect
2009. of tarenflurbil on cognitive decline and activities of daily
[56] E. R. Siemers, S. Friedrich, R. A. Dean et al., Safety, tolera- living in patients with mild Alzheimer disease: a randomized
bility and biomarker e ects of an A monoclonal antibody controlled trial, Journal of the American Medical Association,
administered to patients with Alzheimers disease, Alz- vol. 302, no. 23, pp. 25572564, 2009.
heimers & Dementia, vol. 4, no. 4, supplement 2, p. T774, [72] B. P. Imbimbo, B. Hutter-Paier, G. Villetti et al., CHF5074,
2008. a novel -secretase modulator, attenuates brain -amyloid
[57] M. Bednar, Q. Zhao, J. W. Landen, C. B. Billing, K. pathology and learning deficit in a mouse model of
Rohrbacher, and J. W. Kupiec, Safety and pharmacokinetics Alzheimers disease, British Journal of Pharmacology, vol.
of the anti-amyloid monoclonal antibody PF-04360365 156, no. 6, pp. 982993, 2009.
following a single infusion in patients with mild-to-modertae [73] R. J. van Marum, Current and future therapy in Alzheimers
Alzheimers disease: preliminary results, Alzheimers & disease, Fundamental and Clinical Pharmacology, vol. 22, no.
Dementia, vol. 5, no. 4, supplement 1, p. P157, 2009. 3, pp. 265274, 2008.
[58] W. Thies and L. Bleiler, 2011 Alzheimers disease facts and [74] H. H. Griths, I. J. Morten, and N. M. Hooper, Emerging
figures, Alzheimers & Dementia, vol. 7, no. 2, pp. 208244, and potential therapies for Alzheimers disease, Expert
2011. Opinion on Therapeutic Targets, vol. 12, no. 6, pp. 693704,
[59] N. R. Relkin, P. Szabo, B. Adamiak et al., 18-Month study 2008.
of intravenous immunoglobulin for treatment of mild Alz- [75] A. D. Snow, J. Cummings, T. Lake et al., Exebryl-1: a novel
heimer disease, Neurobiology of Aging, vol. 30, no. 11, pp. small molecule currently in human clinical trials as a disease-
17281736, 2009. modifying drug for the treatment of Alzheimers disease,
[60] R. C. Dodel, Y. Du, C. Depboylu et al., Intravenous immun- Alzheimers & Dementia, vol. 5, no. 4, supplement 1, p. P418,
oglobulins containing antibodies against -amyloid for the 2009.
treatment of Alzheimers disease, Journal of Neurology, [76] M. Marcade, J. Bourdin, N. Loiseau et al., Etazolate, a neu-
Neurosurgery and Psychiatry, vol. 75, no. 10, pp. 14721474, roprotective drug linking GABAA receptor pharmacology
2004. to amyloid precursor protein processing, Journal of Neuro-
[61] D. Tsakanikas, K. Shah, C. Flores, S. Assuras, and N. R. chemistry, vol. 106, no. 1, pp. 392404, 2008.
Relkin, Eects of uninterrupetd intravenous immunoglob- [77] L. Desire, M. Marcade, H. Peillon, D. Drouin, O. Sol, and
ulin treatment of Alzheimers disease for nine months, Alz- M. Pando, Clinical trials of EHT 0202, a neuroprotective
heimers & Dementia, vol. 4, no. 4, supplement 2, p. T776, and procognitive alpha-secretase stimulator for Alzheimers
2008. disease, Alzheimers & Dementia, vol. 5, no. 4, supplement 1,
[62] G. Landreth, Q. Jiang, S. Mandrekar, and M. Heneka, PP- pp. P255P256, 2009.
AR agonists as therapeutics for the treatment of Alz- [78] R. Etcheberrigaray, M. Tan, I. Dewachtert et al., Therapeutic
heimers disease, Neurotherapeutics, vol. 5, no. 3, pp. 481 eects of PKC activators in Alzheimers disease transgenic
489, 2008. mice, Proceedings of the National Academy of Sciences of the
[63] M. Gold, C. Alderton, M. Zvartau-Hind et al., Rosiglita- United States of America, vol. 101, no. 30, pp. 1114111146,
zone monotherapy in mild-to-moderate alzheimers disease: 2004.
results from a randomized, double-blind, placebo-controlled [79] P. S. Aisen, S. Gauthier, and S. Ferris, A phase III, placebo-
phase III study, Dementia and Geriatric Cognitive Disorders, controlled, double-blind, randomized trial of tramiprosate in
vol. 30, no. 2, pp. 131146, 2010. the clinical management of patients with mild-to-moderate
[64] J. Tang and A. Ghosh, Treating transgenic Alzheimer mice Alzheimers disease (the Alphase study), in Proceedings of the
with a -secretase inhibitor, what have we learned? Aging, 61st American Academy of Neurology Annual Meeting, Seattle,
vol. 3, no. 1, pp. 1416, 2011. Wash, USA, May 2009, S32003.
International Journal of Alzheimers Disease 11

[80] P. S. Aisen, D. Saumier, R. Briand et al., A phase II study tar- in Alzheimers disease, Alzheimers & Dementia, vol. 5, no. 4,
geting amyloid- with 3APS in mild-to-moderate Alzheimer supplement 1, p. P74, 2009.
disease, Neurology, vol. 67, no. 10, pp. 17571763, 2006. [97] I. Gozes, I. Divinski, and I. Piltzer, NAP and D-SAL: neu-
[81] C. Opazo, S. Luza, V. L. Villemagne et al., Radioiodinated roprotection against the amyloid peptide (142), BMC
clioquinol as a biomarker for -amyloid: Zn2+ complexes in Neuroscience, vol. 9, supplement 3, article S3, 2008.
Alzheimers disease, Aging Cell, vol. 5, no. 1, pp. 6979, 2006. [98] N. Shiryaev, Y. Jouroukhin, E. Giladi et al., NAP protects
[82] P. A. Adlard, R. A. Cherny, D. I. Finkelstein et al., Rapid memory, increases soluble tau and reduces tau hyperphos-
restoration of cognition in Alzheimers transgenic mice with phorylation in a tauopathy model, Neurobiology of Disease,
8-hydroxy quinoline analogs is associated with decreased vol. 34, no. 2, pp. 381388, 2009.
interstitial A, Neuron, vol. 59, no. 1, pp. 4355, 2008. [99] D. E. Schmechel, G. Gerard, N. G. Vatakis et al., A phase 2,
[83] L. Lannfelt, K. Blennow, H. Zetterberg et al., Safety, ecacy, double-blind, placebo-controlled study to evaluate the safety,
and biomarker findings of PBT2 in targeting A as a modify- tolerability, and eect on cognitive function of AL-108 after
ing therapy for Alzheimers disease: a phase IIa, double-blind, 12 weeks of intranasal administration in subjects with mild
randomised, placebo-controlled trial, The Lancet Neurology, cognitive impairment, Alzheimers & Dementia, vol. 4, no. 4,
vol. 7, no. 9, pp. 779786, 2008. supplement 2, p. T483, 2008.
[84] J. McLaurin, M. E. Kierstead, M. E. Brown et al., Cyclohex- [100] K. N. Green, J. S. Stean, H. Martinez-Coria et al., Nicoti-
anehexol inhibitors of A aggregation prevent and reverse namide restores cognition in Alzheimers disease transgenic
Alzheimer phenotype in a mouse model, Nature Medicine, mice via a mechanism involving sirtuin inhibition and se-
vol. 12, no. 7, pp. 801808, 2006. lective reduction of Thr231-phosphotau, Journal of Neuro-
[85] S. A. Mandel, T. Amit, L. Kalfon, L. Reznichenko, O. science, vol. 28, no. 45, pp. 1150011510, 2008.
Weinreb, and M. B. H. Youdim, Cell signaling pathways [101] J. Hardy, Expression of normal sequence pathogenic pro-
and iron chelation in the neurorestorative activity of green teins for neurodegenerative disease contributes to disease
tea polyphenols: special reference to epigallocatechin gallate risk: permissive templating as a general mechanism under-
(EGCG), Journal of Alzheimers Disease, vol. 15, no. 2, pp. lying neurodegeneration, Biochemical Society Transactions,
211222, 2008. vol. 33, no. 4, pp. 578581, 2005.
[86] A. Mudher and S. Lovestone, Alzheimers diseasedo [102] M. Meyer-Luehmann, J. Coomaraswamy, T. Bolmont et al.,
tauists and baptists finally shake hands? Trends in Neurosci- Exogenous induction of cerebral -amyloidogenesis is gov-
ences, vol. 25, no. 1, pp. 2226, 2002. erned by agent and host, Science, vol. 313, no. 5794, pp.
[87] P. Tarriot, P. S. Aisen, J. Cummings et al., The ADCS 17811784, 2006.
valproate neuroprotection trial: primary ecacy and safety [103] B. Frost, J. Ollesch, H. Wille, and M. I. Diamond, Confor-
results, Alzheimers & Dementia, vol. 5, no. 4, supplement 1, mational diversity of wild-type tau fibrils specified by tem-
pp. P84P85, 2009. plated conformation change, Journal of Biological Chemistry,
[88] J. J. Pei, M. Sjogren, and B. Winblad, Neurofibrillary degen- vol. 284, no. 6, pp. 35463551, 2009.
eration in Alzheimers disease: from molecular mechanisms [104] H. Braak and E. Braak, Demonstration of amyloid deposits
to identification of drug targets, Current Opinion in Psychi- and neurofibrillary changes in whole brain sections, Brain
atry, vol. 21, no. 6, pp. 555561, 2008. Pathology, vol. 1, no. 3, pp. 213216, 1991.
[89] P. N. Tariot and P. S. Aisen, Can lithium or valproate untie [105] S. Oddo, L. Billings, J. P. Kesslak, D. H. Cribbs, and F. M.
tangles in Alzheimers disease? Journal of Clinical Psychiatry, LaFerla, A immunotherapy leads to clearance of early,
vol. 70, no. 6, pp. 919921, 2009. but not late, hyperphosphorylated tau aggregates via the
[90] H. Hampel, M. Ewers, K. Burger et al., Lithium trial in proteasome, Neuron, vol. 43, no. 3, pp. 321332, 2004.
Alzheimers disease: a randomized, single-blind, placebo- [106] K. Santacruz, J. Lewis, T. Spires et al., Medicine: tau sup-
controlled, multicenter 10-week study, Journal of Clinical pression in a neurodegenerative mouse model improves
Psychiatry, vol. 70, no. 6, pp. 922931, 2009. memory function, Science, vol. 309, no. 5733, pp. 476481,
[91] L. Sereno, M. Coma, M. Rodriguez et al., A novel GSK- 2005.
3 inhibitor reduces Alzheimers pathology and rescues [107] W. Noble, E. Planel, C. Zehr et al., Inhibition of glycogen
neuronal loss in vivo, Neurobiology of Disease, vol. 35, no. synthase kinase-3 by lithium correlates with reduced tauopa-
3, pp. 359367, 2009. thy and degeneration in vivo, Proceedings of the National
[92] C. M. Wischik, P. C. Edwards, R. Y. K. Lai, M. Roth, and Academy of Sciences of the United States of America, vol. 102,
C. R. Harrington, Selective inhibition of Alzheimer disease- no. 19, pp. 69906995, 2005.
like tau aggregation by phenothiazines, Proceedings of the
National Academy of Sciences of the United States of America,
vol. 93, no. 20, pp. 1121311218, 1996.
[93] H. Atamna, A. Nguyen, C. Schultz et al., Methylene blue
delays cellular senescence and enhances key mitochondrial
biochemical pathways, FASEB Journal, vol. 22, no. 3, pp.
703712, 2008.
[94] S. Deiana, C. R. Harrington, C. M. Wischik, and G. Riedel,
Methylthioninium chloride reverses cognitive deficits in-
duced by scopolamine: comparison with rivastigmine, Psy-
chopharmacology, vol. 202, no. 1-3, pp. 5365, 2009.
[95] T. Gura, Hope in Alzheimers fight emerges from unex-
pected places, Nature Medicine, vol. 14, no. 9, p. 894, 2008.
[96] C. Wischik, Rember: issues in design of a phase 3 disease
modifying clinical trial of tau aggregation inhibitor therapy
MEDIATORS of

INFLAMMATION

The Scientific Gastroenterology Journal of


World Journal
Hindawi Publishing Corporation
Research and Practice
Hindawi Publishing Corporation
Hindawi Publishing Corporation
Diabetes Research
Hindawi Publishing Corporation
Disease Markers
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of International Journal of


Immunology Research
Hindawi Publishing Corporation
Endocrinology
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Submit your manuscripts at


http://www.hindawi.com

BioMed
PPAR Research
Hindawi Publishing Corporation
Research International
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of
Obesity

Evidence-Based
Journal of Stem Cells Complementary and Journal of
Ophthalmology
Hindawi Publishing Corporation
International
Hindawi Publishing Corporation
Alternative Medicine
Hindawi Publishing Corporation Hindawi Publishing Corporation
Oncology
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Parkinsons
Disease

Computational and
Mathematical Methods
in Medicine
Behavioural
Neurology
AIDS
Research and Treatment
Oxidative Medicine and
Cellular Longevity
Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Anda mungkin juga menyukai