Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 13

Received: 2 October 2018Revised: 21 November 2018Accepted: 3 December 2018

DOI: 10.1111/bph.14581

Themed Section: Therapeutics for Dementia and Alzheimer's Disease: BJP


New Directions for Precision Medicine

REV I EW A R T I C L E

A short perspective on the long road to effective treatments


for Alzheimer's disease

David S. Reynolds

Research & Policy, Alzheimer's Research UK,


Cambridge, UK
Globally, there are approximately 47 million people living with dementia, and about two thirds of
Correspondence those have Alzheimer's disease (AD). Age is the single biggest risk factor for the vast majority
David S. Reynolds, Research & Policy,
of sporadic AD cases, and because the world's population is aging, the number of people living
Alzheimer's Research UK, Riverside 3, Granta Park,
Great Abington, Cambridge CB21 6AD, UK. with AD is set to rise dramatically over the coming decades. There are currently no disease‐
Email: david.reynolds@alzheimersresearchuk. org
modifying treatments for AD, and the few symptomatic agents available have limited impact on
the disease. Perhaps surprisingly, there is relatively little activity in the AD research and
development field compared with other diseases with a high mortality burden, such as cancer.
There is enormous economic incentive to discover and develop the first disease‐modifying
treatment, but previous failure has significantly reduced further industrial investment in this
field. The short review looks at the historical path trodden to develop treatments and reflects
on the journey down the road to truly effective treatments for people living with AD.
LINKED ARTICLES: This article is part of a themed section on Therapeutics for
Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other
articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/ bph.v176.18/issuetoc

1 | INTRODUCTION globally living with dementia (World Alzheimer Report, 2015). Given that age
is the single biggest risk factor for developing most forms of dementia, including
Alzheimer's disease (AD) is the most common form of dementia accounting
AD, and the world's population is aging, that num- ber is set to rise to
for approximately two thirds of all cases, with vascular dementia, fronto‐
approximately 131 million by 2050 (World Alzheimer Report, 2015). AD, like
temporal dementia, dementia with Lewy bodies, and a host of rarer dementias
most forms of dementia, is progres- sive, slowly robbing the patient of their
making up the rest. Different countries have different ways of recording disease
cognitive abilities, which then impacts on daily activities and their ability to
and its impact on mortality and morbidity, and AD is not always broken out
live independently. Not surprisingly, this places a huge burden on health and
separately. The World Health Organization lists dementia as the fifth largest
social care, with the estimated global cost hitting $1 trillion in 2018 and set to
cause of death across the globe (World Health Organization, 2018). Dementia
rise to
has recently become the single biggest cause of death in the United
$2 trillion by 2030 (World Alzheimer Report, 2015). Unlike many major diseases,
Kingdom responsible for approximately 13% of all deaths (UK Office of
the direct health care cost makes up a relatively small propor- tion of these costs,
National Statistics, 2018). There are approximately 47 million people
social care and unpaid care (e.g., a family member giving up work to look after
someone) being a much larger component
(Prince, Comas‐Herrera, Knapp, Guerchet, & Karagiannidou, 2016). AD
Abbreviations: AD, Alzheimer's disease; ApoE, apolipoprotein E; APP, amyloid precursor protein; Aβ,
and dementia are frequently referred to as the most pressing health care
amyloid peptide; Aβx, amyloid peptide of a specific length (i.e., x amino acids); BACE 1/2, β‐secretase
1/2; FAD, familial Alzheimer's disease challenge of our time, and the numbers above demonstrate why.
3636© 2019 The British Pharmacological Society Br J Pharmacol. 2019;176:3636–3648.
REYNOLDS
3
years before the onset of overt cognitive decline (Jack et al., 2010), such that
AD is now usually described as having preclinical (i.e., pre- symptomatic),
prodromal, mild, moderate, and severe stages. Defini- tions vary in the literature,
but preclinical AD is typically defined as someone with positive AD
biomarkers but not yet showing cognitive impairment. This progresses to the
prodromal stage once mild cognitive impairment is observed, and this may
progress on to mild AD and sub- sequently to moderate and severe AD
followed by death (Amieva et al., 2008; Ballard et al., 2011; Weintraub et al.,
2018). The annual conver- sion rate from mild cognitive impairment to mild AD
varies considerably across studies because of differences in definitions of clinical
symptoms and use of biomarker data, but meta‐analyses suggest that it is approx-
imately 10% (Ward, Tardiff, Dye, & Arrighi, 2013).
The pathological hallmarks of AD were first described by Alois
Alzheimer in 1906 consisting of extracellular plaques, intracellular tan- gles, and
widespread neurodegeneration (Figure 2), although it was not until many decades
FIGURE 1 Comparison of the number of publications in different disease
later that amyloid and tau were identified as the main constituents of these
areas. The graph shows the total number of publications in PubMed in 3‐year
time bins using the search terms “cancer,” “heart disease,” “HIV,” “dementia,” aggregates (Brion, Passareiro, Nunez, & Flament‐Durand, 1985; Glenner,
and “Alzheimer.” All diseases show that the number of publications has Wong, Quaranta, & Eanes, 1984). In the 1970s, the relative vulnerability of the
consistently increased over the last 30 years, with cancer showing the steepest septal cholinergic system was first identified (Davies & Maloney, 1976;
rise. It is also clear that the volume of research activity is far higher in other Whitehouse et al., 1982), and this led to the first effective drugs for treating the
diseases that have effective drug therapies than it is for Alzheimer's disease
cognitive symptoms of AD that were launched around the turn of the last
(AD) or dementia. Whilst the number of publications is not a measure of the
amount of drug discovery or development activity for a given disease, it does century (Francis, Palmer, Snape, & Wilcock, 1999). Donepezil,
act as a surrogate for how much is understood about a given disease, which is rivastigmine, and galantamine are all AChE inhibitors approved for the
very important for successful drug discovery symptomatic treatment of mild‐to‐moderate AD patients (Birks, 2006). AChE
inhibitors prevent the breakdown of synaptically released ACh and thereby
The large, and growing, patient population coupled with high impact on quality of
prolong postsynaptic activation of nicotinic and mus- carinic receptors. It is
life for patients and their families clearly makes the treat- ment of dementia an
thought that the therapeutic benefit largely derives from potentiation of
attractive opportunity for pharmaceutical compa- nies. Not surprisingly, most drug
septo‐hippocampal cholinergic activity improving attention and thus enabling
discovery efforts have been focussed on AD because the size of patient
better cognitive performance (Muir, Everitt, & Robbins, 1994; Sahakian &
population and this article will dis- cuss these primarily. Therefore, it is
Coull, 1993). However, the cholinergic system is also involved in many other,
perhaps surprising that there is such a dearth of approved medications and
notably auto- nomic, functions, which is the reason these drugs have a range
comparatively little R&D activity in this area compared with cancer or heart
of adverse effects (e.g., nausea, diarrhoea, and vomiting). The clinical dose of these
disease. Figure 1 indicates that scientific productivity, as measured by number of
drugs has been empirically selected to balance their tolerabil- ity against
publi- cations, is 28‐fold and eightfold lower for AD, compared with cancer or
therapeutic benefit (Gauthier, 2001; Thompson, Lanctôt, & Herrmann, 2004),
heart disease. Whilst publications alone do not lead to new therapies, they do
although it is likely that greater efficacy could be achieved if tolerability could be
act as a good surrogate of the level of investment and research activity for a
improved (Felder et al., 2018). The only other approved symptomatic treatment
given disease. The dearth of AD literature compared with cancer or heart
for AD is memantine, which is a non‐competitive NMDA glutamate
disease is likely why these latter diseases have many effective treatments and
receptor blocker, although its mechanism of action was only discovered
AD does not. A look back at the efforts, activ- ities, and subsequent failures in
during its clinical develop- ment, which was driven by empirical observation of
AD research is informative for where the field stands today and how to
symptomatic ben- efit (Lipton, 2006; McShane, Areosa Sastre, & Minakaran,
achieve success in the future.
2006). None of these drugs have any significant impact on the progression of
AD, instead just offering modest amelioration of some symptoms.

2 | EXISTING TREATMENTS FOR AD

In common medical practice, clinical AD diagnosis has typically been made


3 | THE AMYLOID CASCADE HYPOTHESIS
based on symptoms, rather than via pathophysiological mea- surements such as
The search for disease‐modifying treatments for AD is the primary goal of most
MRI, PET, or biomarker analyses of blood or CSF samples. It should be noted
therapeutic research efforts, and it was a field that grew rapidly in the 1990s but
that the situation is rapidly changing with increased use of biomarker assessment.
then began to wane over the next 10 years (Figure 2).
Availability of new treatments will likely accelerate that change further.
Biomarker studies and amy- loid PET scans have shown that disease
pathology is evident many
36
B

FIGURE 2 The long road to anti‐amyloid therapies. Starting with the first description of Alzheimer's disease (AD) in 1906 through to ongoing clinical trials with
amyloid‐lowering therapies, this timeline indicates some of the key discoveries and clinical trial outcomes. Relatively little progress was made through the 20th
century until genetic studies clearly linked amyloid processing to disease causation. Since the 1990s, therapeutic research and development has explored several
different approaches to lowering amyloid in the brain with the aim of providing therapeutic benefit in the form of slowing, or stopping, disease progression. Most of
these approaches have been stopped due to either safety issues or lack of robust efficacy. Several anti‐amyloid antibodies are in late stage clinical trials and will
hopefully prove to be effective in patients. FAD: familial Alzheimer's disease; MCI: mild cognitive impairment; APP: amyloid precursor protein

Two key factors drove the large‐scale pharmaceutical investment; the first was
functional changes, and empirical observations. At that time, it appeared that
economic and the second scientific advances. The 1990s saw enormous
genetics had paved a clear path to effective medicines.
economic success of neuroscience drugs such as the atypical antipsychotics for
In 1991, the first Alzheimer's autosomal dominant mutation (V717I) was
schizophrenia (e.g., olanzapine and risperidone), selective 5‐HT (serotonin)
discovered in a family in London in the gene that codes for the amyloid
reuptake inhibitors for depression (e.g., flu- oxetine and paroxetine), α2δ
precursor protein (APP; Goate et al., 1991). It was quickly followed by a
ligands for neuropathic pain (gabapentin and pregabalin), and a range of whole series of other disease‐causative mutations in this gene (Tcw & Goate,
new mechanisms to treat epilepsy (e.g., lamotrigine and topiramate). These 2017). Biochemical studies determined that APP was preferentially cleaved by
successes instilled the (not unreasonable) belief in senior pharmaceutical
enzymes α‐secretase and β‐secretase (more typically referred to as
executives that psychiat- ric and neurological conditions were lucrative
BACE) as well as the γ‐secretase com- plex, to produce peptide fragments of
opportunities for R&D investment given the large patient populations and
varying sequences (De Strooper, Vassar, & Golde, 2010), the most important of
significant unmet patient need, along with advancing scientific understanding of
which was β‐amyloid (Aβ), which was already known to be the primary
the molecular underpinnings of these diseases. AD was top of the neurosci- ence
constituent of amy- loid plaques (Glenner et al., 1984). The cleavage of APP
priority list for many companies. The key scientific advance that increased
into various fragments with a range of biological functions is complex and has been
confidence that effective disease‐modifying drugs could be developed was the
reviewed in detail elsewhere (Gralle & Ferreira, 2007; Ludewig & Korte,
identification of causative genes for human disease. Discovery of the genes
2017). Sequential BACE and γ‐secretase cleavage are required to produce Aβ
responsible for autosomal dominant diseases for the first time directly linked
peptides (De Strooper et al., 2010), whereas α‐secretase cleaves in the middle of
potential drug targets with human disease bypassing the need for less direct linkage
the Aβ sequence and yields nonamyloidogenic fragments (Jorissen et al., 2010;
provided by animal models,
Kuhn et al., 2010). Intriguingly, nearly
REYNOLDS
3
all of the disease‐causing mutations on APP cluster around the BACE and γ‐
et al., 1999; Lin et al., 2000; Sinha et al., 1999; Vassar et al., 1999; Yan et al.,
secretase cleavage sites, implicating inappropriate processing as the 1999). There are two BACE isoforms (BACE1 and BACE2), which initially
pathophysiological mechanism in AD. Two further genes linked to familial AD
appeared to have fewer alternative substrates than γ‐secretase. It was therefore
(FAD), presenilin 1 and 2 (Bertram, Lill, & Tanzi, 2010; Lendon, Ashall,
reasoned that BACE inhibitors may be safer than γ‐ secretase inhibitors; however,
& Goate, 1997; Sherrington et al., 1995), were a few years later shown to be
subsequent investigation demonstrated a range of important substrates for both
the critical aspartyl protease components in the γ‐secretase complex (De
BACE1 and BACE2 (Dislich & Lichtenthaler, 2012). The active site of BACE
Strooper et al., 2010; Sisodia & St George‐Hyslop, 2002). FAD cases make
is very open compared with other aspartyl proteases (Hong et al., 2000), which
up <1% of all AD cases, but the clinical symptoms, disease progression, and
made it very dif- ficult to find potent and selective inhibitors that also possessed
biochemical and neu- ropathological changes are all remarkably similar to typical
drug‐ like properties (Ghosh, Brindisi, & Tang, 2012; Vassar et al., 2014). Many of
late‐onset sporadic AD, except that symptom onset is usually in the third to fifth
the early inhibitors were peptide mimetic molecules with poor absorption,
decades of life rather than in old age (Ryman et al., 2014).
stability, and brain or cell penetration making them unsuit- able even for rodent in
The amyloid cascade hypothesis of AD was proposed by Hardy in the early
vivo studies let alone for clinical development (Ghosh et al., 2012). Concerted
1990s, in which build‐up of longer forms of Aβ, particularly the aggregation‐
medicinal chemistry efforts eventually yielded appropriate molecules, and Merck
prone Aβ42, led sequentially to the formation of neu- rotoxic oligomers, Inc. led the way into large‐ scale clinical trials with verubecestat (MK‐8931)
insoluble amyloid fibrils, and ultimately amyloid plaques (Hardy & Allsop, in 2012 (Scott et al., 2016). BACE inhibitors have demonstrated an acceptable
1991; Hardy & Selkoe, 2002). Most FAD mutations shift the ratio of Aβ safety profile for large‐scale clinical trials in AD patients at doses that robustly
peptides towards the longer, more aggregation‐prone forms. In sporadic reduce Aβ levels in plasma and CSF (Kennedy et al., 2016). Verubecestat has
forms, it was not clear if there was over production of Aβ generally, an been tested in prodromal, mild, and moderate AD patient populations, but despite
increase in the ratio Aβ42/ Aβ40, or alternatively, insufficient clearance from lowering CSF Aβ levels (Kennedy et al., 2016), no cognitive or functional
the brain of Aβ42, any of which could lead to accumulation and in turn benefit was observed in patients (Alzforum, 2018c; Egan et al., 2018). Since the
aggregation. The critical involvement of BACE and γ‐secretase in disease negative Phase III results of verubecestat, other BACE inhibitor studies have also
pathology presented two obvious drug targets, especially as enzyme inhibitors been terminated (Alzforum, 2018b,d). The third major approach to therapeutically
are a well‐precedented class of drugs (Vassar et al., 2014). target Aβ directly is active or passive immunization. Encouraging efficacy was
γ‐Secretase was the target most heavily pursued initially, largely because first shown by Schenk et al. (1999) at Elan Corporation using the PDAPP
assays of γ‐secretase activity existed before the complete make‐up of the Alzheimer's mouse model. PDAPP mice overexpress a mutant form of human
enzymatic complex was elucidated and presenilins identified as the catalytic APP (APP V717F) and show age‐dependent deposition of amyloid plaques
subunit (Sisodia & St George‐Hyslop, 2002). γ‐Secretase inhibitors, such as (Masliah et al., 1996). Active immunization of these mice with Aβ42 peptide and
semagacestat from Eli Lilly, were relatively quickly identified and advanced QS‐21 adjuvant blocked the development of amyloid plaque pathology when
into large clinical trials on AD patients. Unfortunately, γ‐secretase has around 40 given to young mice (6 weeks of age) and markedly reduced established
cellular substrates, which resulted in significant on‐target toxicity. Notch, a key pathology in 11‐month‐old mice (Schenk et al., 1999). Elan then progressed this
compo- nent of the Wnt signalling pathway, is a substrate of γ‐secretase agent, known as AN‐1792, into a clinical trial in mild‐to‐moderate AD patients.
(Geling, Steiner, Willem, Bally‐Cuif, & Haass, 2002; Micchelli et al., 2003), Unfortu- nately, a small number of patients developed serious brain inflamma- tion
and blocking Notch cleavage is likely the reason increases in skin cancers and that was later shown to be aseptic meningoencephalitis (Nicoll et al., 2003).
infections were observed in these clinical trials (Chávez‐Gutiérrez et al., Given the serious nature of this adverse event and no means of
2012; Doody et al., 2013). These adverse effects, plus the fact that the predicting/excluding patients at greatest risk, development of AN‐1792 was
semagacestat‐treated patients actually had worse cognitive decline than those in terminated in 2002 (Check, 2002). Only a subset of patients in the trial mounted
the placebo group, resulted in its discontinuation in 2011 (Doody et al., 2013). a significant antibody response to the treatment, mostly directed to the N‐
Several companies pursued other γ‐secretase inhibitors into the clinic and terminus portion of Aβ, and this was not correlated with those experiencing aseptic
experienced similar challenges, eventually resulting in them all being stopped. Two meningoencephali- tis (Lee et al., 2005). Importantly, post‐mortem analysis of the
alternative approaches to γ‐secretase have been proposed and to some extent patients who mounted a response showed that some clearance of amyloid
persued: (a) γ‐secretase modulators (Bursavich, Harrison, & Blain, 2016; Hall & plaques from their brains had occurred (Nicoll et al., 2003). However, there
Patel, 2014) that reduce APP cleavage but have reduced effect on the cleavage were conflicting results as to whether the patients experienced
of other substrates and (b) γ‐secretase stabilizers that shift cleavage to shorter, clinical benefit or not (Holmes et al., 2008; Vellas et al., 2009).
less toxic Aβ peptides such as Aβ38 (Szaruga et al., 2017). γ‐Secretase The mechanistic success of AN‐1792 (i.e., clearance of amyloid plaques in
modulators have reached clin- ical trials, but clinical benefit has not yet been humans) encouraged the field to find similar approaches with an improved safety
demonstrated, whereas γ‐secretase stabilizers are still at the preclinical profile. Several companies developed passive immunotherapies where humanized
discovery stage. antibodies targeting Aβ peptides were administered to patients. The major safety
BACE inhibitor development has also been an area of intense focus once advantage to this
BACE was cloned by five independent groups in 1999 (Hussain
REYNOLDS 36

approach is that the host immune system is not directly stimulated, thereby
4 | Aβ‐ LOWERING THERAPIES: HAVE WE
avoiding the inflammatory response seen with AN‐1792. Additionally, the
REACHED A DEAD END?
epitope of the therapeutic antibody is defined so that therapies aimed at soluble
monomers, oligomers, or fibrillar Aβ could be designed. The disadvantage of
The lack of reproducible success of amyloid‐targeting therapeutics to date is
the passive immunotherapy response is that the antibodies administered will
clearly disappointing for researchers, pharmaceutical compa- nies, and most of
be cleared from the body necessitating regular dosing (usually every 4 weeks).
all to patients who are still without an effective treatment to slow down or
Antibodies are also very poorly brain penetrant with only approximately 0.1%
stop the course of their disease. However, does this mean that the amyloid
of the dose getting into the brain (Banks et al., 2002; Levites et al., 2006),
hypothesis of AD is incorrect? Cer- tainly, there are voices saying that it is, but
resulting in a high peripheral load of antibody and consequently a higher dose
is this really true? A careful look at the research data and clinical trial findings
needing to be given, which greatly increases the cost of manufacturing the
is required to deter- mine how thoroughly the hypothesis has actually been
therapy. In the decade following AN‐1792 termina- tion, a number of Aβ‐
tested. Three key questions need to be answered to understand how well we
targeting antibodies, including solanezumab (Eli Lilly), crenezumab
have tested the amyloid hypothesis. These are (a) have we been treating the
(Roche), gantenerumab (Roche), and bapineuzumab (Johnson &
right patients, (b) when do we need to treat with Aβ‐lowering therapies to
Johnson/Pfizer), entered clinical trials and demonstrated an improved safety
be effective, and (c) how long do we need to treat for? Over the last 20 years
profile. Some instances of amyloid‐related imaging abnormalities associated with
of clinical studies in AD, we have learnt a huge amount about the disease
either microhaemorrhage or vasogenic oedema were observed in subsets of
course of AD, improved the tools available for studying the disease in
patients, but these have proved manageable for the running of large clinical trials
patients (Blennow & Zetterberg, 2018; Pietrzak, Czarnecka, Mikiciuk‐Olasik,
aimed at establishing efficacy in AD patients (Ostrowitzki et al., 2012; Salloway
& Szymanski, 2018; Rice & Bisdas, 2017), and increased our understanding of
et al., 2014; Sperling et al., 2012). Sadly, the therapeutic benefit in terms of rate
which endpoints are the most informative (Aisen et al., 2017; Reiman et al.,
of cognitive or functional decline has been minimal in prodromal, mild, and
2016). It is therefore much more accurate to say that these large clinical devel-
moderate patient AD populations for many of these antibodies (Cummings,
opment programmes have been negative (i.e., failed to meet their pri- mary
Cohen, et al., 2018; Doody, Farlow, et al., 2014; Doody, Thomas, et al., 2014;
endpoint), rather than failed (i.e., the trial design or execution was flawed such
Ostrowitzki et al., 2017; Siemers et al., 2016). Subgroup analyses, usually
that the results are uninterpretable).
performed post hoc, have indicated some cognitive benefits, but these have failed
The question of whether we have been treating the right patients
to be robustly reproduced.
may seem a simple one to answer, but for dementias, it is actually far from
On a more positive note, two other amyloid antibodies, aducanumab
trivial. In fact, in the earlier antibody trials where an amyloid PET scan was not
and more recently BAN2401, have reported positive effects (Alzforum,
available and therefore was not an entry criterion, it is estimated that up to a
2018a; Sevigny et al., 2016). Aducanumab demon- strated dose‐dependent
third of patients enrolled did not have elevated amyloid levels (Siemers et al.,
clearance of amyloid plaques, as determined by amyloid PET, in a 12‐month
2016; Vellas et al., 2013). It is critical to get the inclusion criteria for enrolment
Phase Ib safety study with approxi- mately 25–30 prodromal and mild AD
into a clinical trial correct such that only patients with a high amyloid load
patients. Impressively, it also showed largely dose‐dependent reductions in the
resulting in AD are studied in the trial. From a scientific point of view, these
Clinical Dementia Rating Scale sum of boxes after 12 months of dosing
criteria need to be as stringent as possible to avoid recruiting inappropriate
(Sevigny et al., 2016). Presentation of data from subsequent open‐label
patients, as those not possessing an amyloid pathology will only add to the
extension phases from this study shows that those benefits are sustained over 4
noise and potentially obscure a genuine positive effect. However, that strin-
years of dosing. This is a small study, and therefore, replication of these
gency needs to be balanced against the practical reality that enough patients
findings in large Phase III trials is required to establish robust safety and
need to be identified to make the running of the trial feasible from both a time
efficacy data. Two Phase III studies completed enrolment in mid‐2018, and the
and cost perspective. The gold‐standard method of diagnosis is
outcomes are awaited. BAN2401 reported the results of an 856 patient dose‐
neuropathological confirmation of plaques and tangles post‐mortem, which is
ranging study in prodromal and mild AD (Alzforum, 2018a). Again, there was
obviously not applicable. Cognitive assessment alone determines that the patient
a dose‐dependent reduction in amyloid PET signal, but only the highest dose (10
has cognitive deficits consistent with AD, but given the wide variation in
mg·kg−1 biweekly) showed a statistically significant improvement in cogntive
symptoms for AD and other dementias such as vascular dementia and fronto‐
decline. Interpretation of the results is complicated by the adaptive randomi-
temporal dementia (Pasquier, 1999), those findings alone are insufficient (Fox
zation design of the study and a change in regulatory safety guidance part way
& Rossor, 1999). Assessment of plasma Aβ concentrations, whether Aβ42 or
through the study resulting in an inbalance in the percentage of apolipoprotein
the ratio of Aβ42/Aβ40, generally has a poor correlation to brain Aβ load
E (ApoE) ε4 carriers in the different arms (Alzforum, 2018a). These results
(Beach, Monsell, Phillips, & Kukull, 2012), although recent meth- odological
are encouraging because of the size of the study but will likely require
advances may be starting to overcome that issue (Nakamura et al., 2018;
replication in pivotal Phase III studies before approval. The reasons why
Ovod et al., 2017). CSF Aβ concentrations are much more predictive of the
aducanumab may be effective when several other antibodies have failed have
brain amyloid load (Clark et al., 2003; Galasko et al., 1998), but there are
not been determined.
concerns over patient
36
B
willingness to undergo a lumbar puncture, which has limited its usage. A key
role of amyloid in AD is yet to be fully determined, but accumulating evidence
advance in biomarker tools for accurate diagnosis of AD patients was the
suggests that it may be more likely to act as a trigger for other pathologies
development of amyloid PET ligands over the last 10 years (Chételat et al.,
and may be less important, or even irrelevant, later in the disease course
2013; Morris et al., 2016). Several agents such as florbetapir and
(Karran, Mercken, & De Strooper, 2011).
flutemetamol are available for use in clinical trials and approved for
Our understanding of this sequence of progressive brain pathol- ogy in
diagnostic use in medical practice. These ligands bind to aggregated Aβ
advance of overt clinical symptoms points to the most likely reason that
(Klunk et al., 2001) and allow the clear determination of elevated amyloid
amyloid‐lowering therapies have not been beneficial in most trials run to date:
load. Their use in screening patients for AD trials is now very wide spread
They were administered too late (van Dyck, 2018). Amyloid load plateaus
and ensures that patients are appropriate for amyloid‐lowering therapy trials.
many years before a patient would be given a diagnosis of prodromal or mild
Trials run before these ligands were widely available recruited a high propor- tion
AD, by which time amyloid may not be the primary driver of disease
of amyloid negative subjects (Siemers et al., 2016; Vellas et al., 2013), which
progression (Jack et al., 2010; Weiner et al., 2010). The ability of amyloid
was initially thought to be a contributory reason for the lack of robust
therapies to lower brain amyloid load, even to the point of being classified as
clinical efficacy. However, a more recent solanezumab trial that included a
amyloid negative by PET scanning and yet have little to no benefit on
positive amyloid PET scan as an inclusion criterion also did not show
cogni- tive decline, would certainly be consistent with this hypothesis (Egan et
robust efficacy (Honig et al., 2018).
al., 2018; Panza et al., 2014). Clinical intervention earlier in the dis- ease
The second question of when patients need to be treated with amyloid‐
course is the obvious next step for these therapies, and several studies in
lowering therapies to have a beneficial effect on disease is still somewhat of an
prodromal or preclinical disease are being conducted (Reiman et al., 2016;
open question because there are no approved therapies. Genetic findings from
Sperling, Mormino, & Johnson, 2014). Recom- mended inclusion criteria for
FAD mutations indicate that increased production of Aβ42 is sufficient to
such studies include a positive amyloid PET scan, lack of significant cognitive
cause AD (Tcw & Goate, 2017). Additionally, a protective mutation, A673T
impairment, and one or more risk factors such as family history or being a
which is close to the BACE cleavage site, has also been discovered that
carrier of the ApoEε4 allele (Dubois et al., 2016). The primary outcome of
reduces Aβ production by approximately 40% in cell culture models (Jonsson
these studies is a slowing of the rate of cognitive decline as measured by a
et al., 2012; Maloney et al., 2014) and protects human carriers from developing
composite cognitive score. The challenge with such studies is the length of time
AD or cognitive decline (Jonsson et al., 2012). The protec- tive genetic
that they take to conduct and the number of patients required given the
mutation is carried from birth, suggesting that in the worst‐case scenario,
variability in rates of disease progression. Both of these factors make such trials
life‐long reduction in Aβ production is required for protection. On the flip‐
prohibitively expensive, but thankfully, industrial and philanthropic funding is
side even in FAD patients, the disease is rarely diagnosed before the age of
allowing a small number of such studies to be carried out. An alternative
30 (Ryman et al., 2014) and more usually in their 40s or 50s, suggesting that
approach to test the early interven- tion hypothesis is selection of a much more
the brain is resilient for several decades before the onset of cognitive decline.
defined patient popula- tion, that is, those with FAD mutations. These
The use of amyloid PET ligands allowed the longitudinal mapping of amyloid
patients will definitely progress to AD, and we know that their APP/PS
deposition and, in combination with other scanning techniques (Wei- ner et al.,
mutation is the driving cause of their disease. Additionally, they are often
2010), built a relatively clear picture of the sequence of events leading to
much more motivated to participate in clinical trials than healthy older indi-
cognitive decline and a clinical diagnosis of AD (Jack, Holtzman, &
viduals in the general population who are currently symptom free. Research
Holtzman, 2013; Jack et al., 2010). Amyloid levels start to rise at least 20
networks of FAD carriers have been established, and amyloid‐lowering
years prior and actually plateau about 10–15 years before the onset of overt
therapy preventive trials, such as DIAN‐TU, have been ongoing for the last few
clinical symptoms resulting in a diagnosis of AD (Schott & Petersen, 2015).
years (Mills et al., 2013; Sperling, Rentz, et al., 2014).
Additional scanning techniques such as fluorodeoxyglucose uptake to look at
The third question of how long we need to treat for is also
metabolic activity (O'Brien et al., 2014), volumetric MRI to assess atrophy
something of an unknown and likely to depend on the exact mech- anism of
(Jack et al., 2010), microglial activation (Edison, Donat, & Sastre, 2018;
the therapy. PET studies in mild‐to‐moderate AD patients using antiamyloid
Varley, Brooks, & Edison, 2015), and tau (Okamura et al., 2018) PET ligands
antibodies have shown that amyloid load can be reduced over a period of 6–18
have built up a picture of the sequence of events in the brain leading up to
months in a dose‐dependent manner (Ostrowitzki et al., 2017; Sevigny et al.,
cognitive decline. These can be plotted as a series of sequential but
2016). The relatively slow clearance of already deposited amyloid is probably
overlapping curves first clearly summarized by Jack et al. (2010) and Jack et
a combination of preventing further deposition plus removal of plaques via
al. (2013): Amyloid build up occurs first, followed by metabolic decline,
activa- tion of microglial clearance mechanisms (Ostrowitzki et al., 2012;
then tau accumulation leading to reductions in brain volume. Only at this
Sevigny et al., 2016). Verubecestat has likewise shown a gradual clearance
relatively late stage in terms of brain pathology are obvious symptoms of
of plaques over several months even though a reduction of CSF or plasma Aβ
cognitive decline observed and a clinical diagnosis of AD typically given. The
occurs much more quickly. Understanding the kinetics of amyloid plaque
precise
clearance is interesting from a scientific
3 B
point of view, but evidence to date shows that it offers limited patient
diagnosis, and their accumulation correlates much better with cogni- tive
benefit. Instead, prevention of deposition in the first place may be a more
decline (Arriagada, Growdon, Hedley‐Whyte, & Hyman, 1992; Nelson et al.,
relevant goal, which hopefully, the ongoing second- ary prevention trials will
2012; Tomlinson, Blessed, & Roth, 1970). Interestingly, disease‐causing
tell us. The duration of treatment for sustained beneficial effect is likely to be
mutations in tau are not linked to AD, rather to various other forms of
life long as the longitudinal data suggest that amyloid deposition is probably neurodegenerative diseases such as fronto‐ temporal dementia, progressive
the triggering event for AD rather than necessary throughout the disease supranuclear palsy, Pick's disease, and corticobasal degeneration (Ghetti et
(Karran et al., 2011). As such amyloid levels will need to be kept low for al., 2015; Hutton et al., 1998; Spillantini et al., 1998). Whilst this may appear
long periods to avoid the disease processes reinitiating. This in turn requires to weaken tau as a therapeutic target for AD, it does strengthen the case that
that therapies have a very good safety profile such that the benefit–risk is still aberrant tau and its aggregation into intracellular tangles are sufficient to
favourable even in people who have no overt disease. Any ongoing safety drive neurodegeneration, albeit with varied clinical pre- sentation. Additionally,
monitoring requirements, such as regu- lar MRI scans for signs of amyloid‐ tau is necessary for Aβ‐induced neurotoxicity (Rapoport, Dawson, Binder, Vitek,
related imaging abnormalities, could be detrimental to the real‐world uptake & Ferreira, 2002). The challenge with tau as a drug target has been how to
of these therapies for long‐term secondary prevention. modulate it. The normal function of tau is as an intracellular scaffolding
protein forming microtubules (Weingarten, Lockwood, Hwo, & Kirschner,
1975). Hyperphosphorylation of tau leads to aggregation first in the form of
5 | ALTERNATIVE ROADS TO SUCCESS? paired helical filaments and then into tangles (Grundke‐Iqbal et al., 1986; Iqbal
et al., 1986). As a scaffolding protein, it is much less obvi- ous if there is an
The disproportionate amount of academic research effort and indus- trial amenable binding site for small molecule drugs to interfere with its
investment into the amyloid cascade hypothesis has probably slowed the field pathophysiological function compared with the active site of an enzyme or
down in investigating alternative mechanisms and developing drugs for other ligand binding site on a GPCR. Additionally, being an intracellular protein, it
targets (Figure 3). The most obvious alter- native target is tau: Tau tangles are was not thought to be amenable to antibody therapies that do not readily
a diagnostic criterion for AD penetrate into cells. It is only

FIGURE 3 Alternative routes to effective treatments for Alzheimer's disease (AD). The schematic illustrates nonamyloid mechanisms that have or are being
investigated for therapeutic potential in AD over the last 30 years. The neurochemical analyses of 1970s and 1980s led to the development and launch of
symptomatic treatments for AD around the turn of the century. AChE inhibitors and the glutamate antagonist memantine are the only approved drugs to treat the
cognitive symptoms of AD. Most of the research activity is now focused on disease‐modifying treatments rather than more symptomatic drugs. Tau and apolipoprotein E
were recognized in the 1990s as key players in AD pathophysiology but have yet to be successfully targeted for disease‐modifying therapies. Advances in genetic
technologies led to genome‐wide association studies (GWAS) uncovering small‐effect size risk alleles, which have fuelled most of the avenues of active research
today. These include lipid processing, mitochondrial dysfunction, protein folding, and clearance mechanisms as well as the role of the innate immune system in
neurodegeneration
REYNOLDS
3
in recent years, when it was discovered that tau accumulation follows a specific
alleles are not themselves particularly obvious drug targets, they sit on
pattern of spreading through the brain, which is hypothe- sized to occur via
pathways with tractable targets that are now at the preclinical stages of drug
trans‐synaptic transmission of tau “seeds” (Clavaguera et al., 2009, 2013;
discovery. In the next few years, many of these will move into clinical
Frost & Diamond, 2010), that there has been renewed interest in tau as a target.
testing and hopefully yield positive results on dis- ease progression in
Several anti‐tau antibod- ies have been developed (Golde, Lewis, & McFarland,
patients.
2013) and are moving into Phase II clinical trials (Cummings, Lee, Ritter, & Whilst the main drug development focus is on finding a drug that is
Zhong, 2018). The knowledge gained from negative amyloid trials in terms of
efficacious, it is most likely that polypharmacy will be required for effective
patient selection, appropriate endpoints, and effective use of bio- markers clinical management of AD. The genetic studies indicate that more than one
including tau PET ligands will all hopefully enable much faster testing of the tau
pathophysiological mechanism can cause AD and it is likely that multiple
hypothesis in AD patient trials. The evidence with tau suggests that it remains an
mechanisms will be active in any given patient. Recent post‐mortem studies are
important driver of disease progression later into the disease course than amyloid,
increasingly demonstrating that many patients have overlapping neuropathological
and hence, trials in prodro- mal or mild‐to‐moderate stage AD patients perhaps
features of AD, vascular dementia, dementia with Lewy bodies, and so forth
have a greater chance of success.
(Attems & Jellinger, 2014; Irwin et al., 2017; Spires‐Jones, Attems, &
ApoE function is a second area of great interest, as ApoE is the big- gest
Thal, 2017). These data indicate that mixed pathologies may be relatively
genetic risk factor in the general population (Corder et al., 1993; Holtzman,
common, and therefore, treating the patients' disease with drugs of multiple
Herz, & Bu, 2012; Strittmatter et al., 1993). Unlike APP and PS mutations that
mechanisms will likely be necessary. The use of biomarkers, MRI/PET scans,
are causal for FAD, ApoE just increases the risk of developing late onset AD.
and other means of appropriately stratifying patients will need to develop
There are three common allelic forms ε2, ε3, and ε4 with a frequency of 8%,
significantly from where they are today to be able to correctly identify which
78%, and 14%, respectively (Farrer et al., 1997). Compared with someone with
mechanisms are relevant to a specific patient and therefore start treatment
the ε3/ε3 genotype, the odds ratio of developing AD is 3 and 15 times higher
with the appropriate drugs. Such precision medicine approaches for cancer
for the ε3/ε4 and ε4/ε4 genotypes, respectively (Farrer et al., 1997). The therapy are now common place and hopefully will become so for AD and
precise role ApoEε4 plays in AD pathophysiology has been difficult to deter- other dementias in the future.
mine, but it has been shown to increase amyloid deposition (Bales et al., Improvements in the research landscape and drug target pipeline over the
1997), impair degradation (Jiang et al., 2008), and exacerbate tau‐mediated last decade have also been matched by changes in the political and funding
neurodegeneration (Shi et al., 2017). Finding ways to modulate ApoE landscape. Several governments have woken up to the impact of dementia on
function safely as a drug target for AD has so far proved extremely health and social care systems and developed routes and action plans to address
difficult.
the issue. A prom- inent example was the G8 summit in 2013 focused on
The third major step towards alternative drug targets for AD has again
dementia (Fox & Petersen, 2013) that led to the setting of a key therapeutic
come from the field of genetics. The advance in sequencing technologies over
goal: to identify a cure or disease‐modifying therapy for dementia by 2025
the last 30 years has enabled genome‐wide associ- ation studies so that genes
(Vradenburg, 2015). Many organizations around the world have aligned
with small effect sizes could be identified, rather than just the large effect size
themselves with this goal, which could be achieved by several agents in late
of familial genes first discovered in the 1990s. Several genome wide association
stage clinical trials (Cummings, Lee, et al., 2018), although this is by no means
studies involving hun- dreds of thousands of subjects have now identified
a certainty! The setting of polit- ical goals has also brought increased
approximately
government funding into the dementia research field with big increases in the
30 risk alleles (Guerreiro & Hardy, 2014; Zhu, Tan, Tan, & Yu, 2017) and
U.S.'s National Institute of Health dementia budget in recent years (Alzforum,
two protective variants (ApoEε2 and PLCγ2; Corder et al., 1994; Sims et 2018e) and the establishment of the Dementia Research Institute in the United
al., 2017). Other than ApoEε4, which has been known as a significant risk Kingdom (https://ukdri.ac.uk/). Increased venture cap- ital funding has also
factor for many years (Corder et al., 1993; Farrer et al., 1995), few of the been created through targeted investment mechanisms such the Dementia
newer risk alleles are directly involved in amyloid or tau processing. Instead, Discovery Fund, which now has
pathway and network analyses have shown them to be primarily related to $350 million to invest in dementia drug discovery and development
the innate immune system, energy metabolism, and proteostasis/autophagy (https://theddfund.com/). Finally, philanthropists with global profile such as Bill
(De Strooper & Karran, 2016; Zhu et al., 2017). These discoveries have Gates (Marais, 2018) and the Chan‐Zuckerberg Initiative
also revealed a convergence of the mechanisms involved in
(https://www.chanzuckerberg.com/) are increasingly turning their attention to
neurodegeneration, so that despite different clinical presentations and
dementia research funding, which attracts further public and political interest
neuropathological signatures in other dementia‐causing diseases, the underlying
in the area.
biology shares many commonalities (Brettschneider, Del Tredici, Lee, &
In summary, we are still travelling down the long road to effective disease‐
Trojanowski, 2015; Salter & Stevens, 2017). The above factors have
modifying treatments for dementia, but several alternative routes are hopefully
reinvigorated the dementia research field and industrial drug discovery
increasing our chance of successfully reaching our destination. Experience in
investment. Although many of the risk
how to run clinical trials should help
3 B
avoid some of the inefficiencies of previous trials. Rapidly improving Alzforum. (2018d). Alzforum Scratch Lanabecestat: This BACE inhibitor doesn't
understanding of the mechanisms involved in neurodegenerative dis- eases is work in symptomatic AD, either | ALZFORUM.
opening up new areas of biology and offering a wealth of potential drug Alzforum. (2018e). Alzforum Senate approves $2.34 billion budget for Alzheimer's
targets. Increased political and public awareness and interest in dementia is research | ALZFORUM.
leading to more funding and increased research as well as drug discovery and Amieva, H., Le Goff, M., Millet, X., Orgogozo, J. M., Pérès, K., Barberger‐ Gateau,
development capacity. Hopefully, they will all help us find the urgently P., … Dartigues, J. F. (2008). Prodromal Alzheimer's disease: Successive
needed treatments and better out- comes for people living with dementia. emergence of the clinical symptoms. Annals of Neurology, 64, 492–498.
https://doi.org/10.1002/ana.21509
Arriagada, P. V., Growdon, J. H., Hedley‐Whyte, E. T., & Hyman, B. T. (1992).
5.1 | Nomenclature of targets and ligands Neurofibrillary tangles but not senile plaques parallel duration and severity of
Alzheimer's disease. Neurology, 42, 631–639.
Key protein targets and ligands in this article are hyperlinked to Attems, J., & Jellinger, K. A. (2014). The overlap between vascular disease and
corresponding entries in http://www.guidetopharmacology.org, the common portal Alzheimer's disease—Lessons from pathology. BMC Med- icine, 12, 206.
for data from the IUPHAR/BPS Guide to PHARMA- COLOGY (Harding et Bales, K. R., Verina, T., Dodel, R. C., Du, Y., Altstiel, L., Bender, M., … Paul, S.
al., 2018), and are permanently archived in the Concise Guide to M. (1997). Lack of apolipoprotein E dramatically reduces amyloid β‐ peptide
PHARMACOLOGY 2017/18 (Alexander, Christopoulos et al., 2017; deposition. Nature Genetics, 17, 263–264. https://doi.org/ 10.1038/ng1197‐
263
Alexander, Fabbro et al., 2017; Alexander, Kelly et al., 2017; Alexander, Peters
et al., 2017). Ballard, C., Gauthier, S., Corbett, A., Brayne, C., Aarsland, D., & Jones, E. (2011).
Alzheimer's disease. Lancet, 377, 1019–1031. https://doi.org/ 10.1016/S0140‐
6736(10)61349‐9
CONFLICT OF INTEREST Banks, W. A., Terrell, B., Farr, S. A., Robinson, S. M., Nonaka, N., & Morley,
D.R. holds shares in Pfizer Ltd and was a past employee of Pfizer Inc. and J. E. (2002). Passage of amyloid β protein antibody across the blood‐ brain
barrier in a mouse model of Alzheimer's disease. Peptides, 23, 2223–2226.
Merck Inc.
https://doi.org/10.1016/S0196‐9781(02)00261‐9
Beach, T. G., Monsell, S. E., Phillips, L. E., & Kukull, W. (2012). Accuracy of the
ORCID clinical diagnosis of Alzheimer disease at National Institute on Aging
Alzheimer Disease Centers, 2005–2010. Journal of Neuropathol- ogy and
David S. Reynolds https://orcid.org/0000-0002-5461-3166
Experimental Neurology, 71, 266–273. https://doi.org/
10.1097/NEN.0b013e31824b211b
REFERENCES Bertram, L., Lill, C. M., & Tanzi, R. E. (2010). The genetics of Alzheimer dis- ease:
Aisen, P. S., Cummings, J., Jack, C. R., Morris, J. C., Sperling, R., Frölich, L., … Back to the future. Neuron, 68, 270–281.
Dubois, B. (2017). On the path to 2025: Understanding the Alzheimer's disease
Birks, J. S. (2006). Cholinesterase inhibitors for Alzheimer's disease. In J. S. Birks
continuum. Alzheimers. Res. Ther., 9, 60. https://doi.org/ 10.1186/s13195‐
(Ed.), Cochrane database of systematic reviews. Chichester, UK: John
017‐0283‐5 Wiley & Sons, Ltd.
Alexander, S. P. H., Christopoulos, A., Davenport, A. P., Kelly, E., Marrion, N. V.,
Blennow, K., & Zetterberg, H. (2018). Biomarkers for Alzheimer's disease: Current
Peters, J. A., … CGTP Collaborators. (2017). The Concise Guide to
status and prospects for the future. Journal of Internal Medicine, 284, 643–663.
PHARMACOLOGY 2017/18: G protein‐coupled receptors. British Jour- nal of
https://doi.org/10.1111/joim.12816
Pharmacology, 174(S1), S17–S129. https://doi.org/10.1111/ bph.13878
Brettschneider, J., Del Tredici, K., Lee, V. M.‐Y., & Trojanowski, J. Q. (2015).
Alexander, S. P. H., Fabbro, D., Kelly, E., Marrion, N. V., Peters, J. A., Spreading of pathology in neurodegenerative diseases: A focus on human
Faccenda, E., … CGTP Collaborators. (2017). The Concise Guide to studies. Nature Reviews. Neuroscience, 16, 109–120.
PHARMACOLOGY 2017/18: Enzymes. British Journal of Pharmacology,
174(S1), S272–S359. https://doi.org/10.1111/bph.13877 Brion, J.‐P., Passareiro, H., Nunez, J., & Flament‐Durand, J. (1985). Mise en
évidence immunologique de la protéine tau au niveau des lésions de
Alexander, S. P. H., Kelly, E., Marrion, N. V., Peters, J. A., Faccenda, E., dégénérescence neurofibrillaire de la maladie d'Alzheimer. Archives of Biology
Harding, S. D., … CGTP Collaborators. (2017). The Concise Guide to (Liege), 95, 229–235.
PHARMACOLOGY 2017/18: Overview. British Journal of Pharmacol- ogy,
Bursavich, M. G., Harrison, B. A., & Blain, J.‐F. (2016). Gamma secretase
174(S1), S1–S16. https://doi.org/10.1111/bph.13882
modulators: New Alzheimer's drugs on the horizon? Journal of Medici- nal
Alexander, S. P. H., Peters, J. A., Kelly, E., Marrion, N. V., Faccenda, E., Chemistry, 59, 7389–7409.
Harding, S. D., … CGTP Collaborators. (2017). The Concise Guide to
PHARMACOLOGY 2017/18: Ligand‐gated ion channels. British Journal of Chávez‐Gutiérrez, L., Bammens, L., Benilova, I., Vandersteen, A., Benurwar, M.,
Pharmacology, 174(S1), S130–S159. https://doi.org/10.1111/ bph.13879 Borgers, M., … de Strooper, B. (2012). The mechanism of γ‐ secretase
dysfunction in familial Alzheimer disease. The EMBO Journal, 31, 2261–2274.
Alzforum. (2018a). Alzforum BAN2401 removes brain amyloid, possibly slows https://doi.org/10.1038/emboj.2012.79
cognitive decline | ALZFORUM.
Check, E. (2002). Nerve inflammation halts trial for Alzheimer's drug.
Alzforum. (2018b). Alzforum Liver Tox Ends Janssen BACE program Nature, 415, 462–462. https://doi.org/10.1038/415462a
| ALZFORUM.
Chételat, G., La Joie, R., Villain, N., Perrotin, A., de La Sayette, V., Eustache, F., &
Alzforum. (2018c). Alzforum Merck Axes Verubecestat for Prodromal AD, Vandenberghe, R. (2013). Amyloid imaging in cognitively normal individuals,
Researchers Say ‘Go Earlier’ | ALZFORUM. at‐risk populations and preclinical Alzheimer's disease. NeuroImage: Clinical, 2,
356–365. https://doi.org/10.1016/j. nicl.2013.02.006
REYNOLDS
3
Clark, C. M., Xie, S., Chittams, J., Ewbank, D., Peskind, E., Galasko, D., …
Egan, M. F., Kost, J., Tariot, P. N., Aisen, P. S., Cummings, J. L., Vellas, B., …
Trojanowski, J. Q. (2003). Cerebrospinal fluid tau and β‐amyloid. Archives
Michelson, D. (2018). Randomized trial of verubecestat for mild‐to‐ moderate
of Neurology, 60, 1696. https://doi.org/10.1001/ archneur.60.12.1696
Alzheimer's disease. The New England Journal of Medicine, 378, 1691–1703.
Clavaguera, F., Akatsu, H., Fraser, G., Crowther, R. A., Frank, S., Hench, J., … https://doi.org/10.1056/NEJMoa1706441
Tolnay, M. (2013). Brain homogenates from human tauopathies induce tau Farrer, L. A., Adrienne Cupples, L., Van Duijn, C. M., Kurz, A., Zimmer, R.,
inclusions in mouse brain. Proceedings of the National Academy of Sciences, Müller, U., … Haines, J. L. (1995). Apolipoprotein E genotype in patients with
110, 9535–9540. https://doi.org/10.1073/pnas.1301175110 Alzheimer's disease: Implications for the risk of dementia among relatives.
Clavaguera, F., Bolmont, T., Crowther, R. A., Abramowski, D., Frank, S., Probst, Annals of Neurology, 38, 797–808. https://doi.org/10.1002/ ana.410380515
A., … Tolnay, M. (2009). Transmission and spreading of tauopathy in Farrer, L. A., Cupples, L. A., Haines, J. L., Hyman, B., Kukull, W. A., Mayeux, R.,
transgenic mouse brain. Nature Cell Biology, 11, 909–913. … van Duijn, C. (1997). Effects of age, sex, and ethnicity on the association
https://doi.org/10.1038/ncb1901 between apolipoprotein E genotype and Alzheimer disease. A meta‐analysis.
Corder, E., Saunders, A., Strittmatter, W., Schmechel, D., Gaskell, P., Small, G., … APOE and Alzheimer Disease Meta Analysis Consortium. Jama, 278, 1349–
Pericak‐Vance, M. (1993). Gene dose of apolipoprotein E type 4 allele and the 1356. https://doi.org/10.1001/ jama.1997.03550160069041
risk of Alzheimer's disease in late onset families. Science, 261, 921–923. Felder, C. C., Goldsmith, P. J., Jackson, K., Sanger, H. E., Evans, D. A., Mogg,
https://doi.org/10.1126/science.8346443 A. J., & Broad, L. M. (2018). Current status of muscarinic M1 and M4
Corder, E. H., Saunders, A. M., Risch, N. J., Strittmatter, W. J., Schmechel, receptors as drug targets for neurodegenerative diseases. Neuropharmacology,
D. E., Gaskell, P. C., … Pericak‐Vance, M. A. (1994). Protective effect of 136, 449–458. https://doi.org/10.1016/j. neuropharm.2018.01.028
apolipoprotein E type 2 allele for late onset Alzheimer disease. Nature Fox, N. C., & Petersen, R. C. (2013). The G8 Dementia Research Summit—A starter
Genetics, 7, 180–184. https://doi.org/10.1038/ng0694‐180 for eight? Lancet (London, England), 382, 1968–1969.
Cummings, J., Lee, G., Ritter, A., & Zhong, K. (2018). Alzheimer's disease drug Fox, N. C., & Rossor, M. N. (1999). Diagnosis of early Alzheimer's disease.
development pipeline: 2018. Alzheimer's & Dementia: Translational Research & Revue Neurologique (Paris), 155(Suppl 4), S33–S37.
Clinical Interventions, 4, 195–214. https://doi.org/10.1016/ j.trci.2018.03.009
Francis, P. T., Palmer, A. M., Snape, M., & Wilcock, G. K. (1999). The cholin- ergic
Cummings, J. L., Cohen, S., van Dyck, C. H., Brody, M., Curtis, C., Cho, W., hypothesis of Alzheimer's disease: A review of progress. Journal of Neurology,
… Paul, R. (2018). ABBY. Neurology, 90, e1889–e1897. https://doi.org/ Neurosurgery, and Psychiatry, 66, 137–147. https://doi.org/
10.1212/WNL.0000000000005550 10.1136/jnnp.66.2.137
Davies, P., & Maloney, A. J. (1976). Selective loss of central cholinergic Frost, B., & Diamond, M. I. (2010). Prion‐like mechanisms in neurodegener- ative
neurons in Alzheimer's disease. Lancet (London, England), 2, 1403. diseases. Nature Reviews. Neuroscience, 11, 155–159. https://doi.
De Strooper, B., & Karran, E. (2016). The cellular phase of Alzheimer's dis- ease. org/10.1038/nrn2786
Cell, 164, 603–615. https://doi.org/10.1016/j.cell.2015.12.056 Galasko, D., Chang, L., Motter, R., Clark, C. M., Kaye, J., Knopman, D., … Seubert,
P. (1998). High cerebrospinal fluid tau and low amyloid β42 levels in the clinical
De Strooper, B., Vassar, R., & Golde, T. (2010). The secretases: Enzymes with
therapeutic potential in Alzheimer disease. Nature Reviews. Neurol- ogy, 6, 99– diagnosis of Alzheimer disease and relation to apo- lipoprotein. E genotype.
107. https://doi.org/10.1038/nrneurol.2009.218 Arch. Neurol., 55, 937–945. https://doi.org/ 10.1001/archneur.55.7.937

Dislich, B., & Lichtenthaler, S. F. (2012). The membrane‐bound aspartyl protease Gauthier, S. (2001). Cholinergic adverse effects of cholinesterase inhibitors in
BACE1: Molecular and functional properties in Alzheimer's disease and Alzheimer's disease: Epidemiology and management. Drugs & Aging, 18, 853–
beyond. Frontiers in Physiology, 3, 8. 862. https://doi.org/10.2165/00002512‐200118110‐00006

Doody, R. S., Farlow, M., Aisen, P. S., & Alzheimer's Disease Cooperative Study Geling, A., Steiner, H., Willem, M., Bally‐Cuif, L., & Haass, C. (2002). A
Data Analysis and Publication Committee. (2014). Phase 3 trials of solanezumab gamma‐secretase inhibitor blocks Notch signaling in vivo and causes a severe
and bapineuzumab for Alzheimer's disease. The New England Journal of neurogenic phenotype in zebrafish. EMBO Reports, 3, 688–694.
https://doi.org/10.1093/embo‐reports/kvf124
Medicine, 370, 1459–1460.
Ghetti, B., Oblak, A. L., Boeve, B. F., Johnson, K. A., Dickerson, B. C., &
Doody, R. S., Raman, R., Farlow, M., Iwatsubo, T., Vellas, B., Joffe, S., …
Goedert, M. (2015). Invited review: Frontotemporal dementia caused by
Semagacestat Study Group. (2013). A phase 3 trial of semagacestat for
microtubule‐associated protein tau gene (MAPT) mutations: A chame- leon
treatment of Alzheimer's disease. The New England Journal of Medicine, 369,
for neuropathology and neuroimaging. Neuropathology and Applied
341–350. https://doi.org/10.1056/NEJMoa1210951
Neurobiology, 41, 24–46.
Doody, R. S., Thomas, R. G., Farlow, M., Iwatsubo, T., Vellas, B., Joffe, S., …
Mohs, R. (2014). Phase 3 trials of solanezumab for mild‐to‐moderate Ghosh, A. K., Brindisi, M., & Tang, J. (2012). Developing β‐secretase inhib- itors
Alzheimer's disease. The New England Journal of Medicine, 370, 311–321. for treatment of Alzheimer's disease. Journal of Neurochemistry, 120, 71–83.
https://doi.org/10.1056/NEJMoa1312889 https://doi.org/10.1111/j.1471‐4159.2011.07476.x
Glenner, G. G., Wong, C. W., Quaranta, V., & Eanes, E. D. (1984). The amy- loid
Dubois, B., Hampel, H., Feldman, H. H., Scheltens, P., Aisen, P., Andrieu, S.,
deposits in Alzheimer's disease: Their nature and pathogenesis. Applied
… Jack, C. R. Jr. (2016). Preclinical Alzheimer's disease: Definition, nat- ural
Pathology, 2, 357–369.
history, and diagnostic criteria. Alzheimer's Dement., 12, 292–323.
https://doi.org/10.1016/j.jalz.2016.02.002 Goate, A., Chartier‐Harlin, M.‐C., Mullan, M., Brown, J., Crawford, F., Fidani,
L., … Hardy, J. (1991). Segregation of a missense mutation in the amyloid
van Dyck, C. H. (2018). Anti‐amyloid‐β monoclonal antibodies for Alzheimer's
precursor protein gene with familial Alzheimer's disease. Nature, 349, 704–706.
disease: Pitfalls and promise. Biological Psychiatry, 83, 311–319.
https://doi.org/10.1038/349704a0
Edison, P., Donat, C. K., & Sastre, M. (2018). In vivo imaging of glial activa- tion in
Golde, T. E., Lewis, J., & McFarland, N. R. (2013). Anti‐tau antibodies: Hit- ting
Alzheimer's disease. Frontiers in Neurology, 9, 625. https://doi.
the target. Neuron, 80, 254–256.
org/10.3389/fneur.2018.00625
3 B
Gralle, M., & Ferreira, S. T. (2007). Structure and functions of the human amyloid
Jack, C. R., Wiste, H. J., Vemuri, P., Weigand, S. D., Senjem, M. L., Zeng, G.,
precursor protein: The whole is more than the sum of its parts. Progress in
… the Alzheimer's Disease Neuroimaging Initiative. (2010). Brain beta‐ amyloid
Neurobiology, 82, 11–32. https://doi.org/10.1016/j. pneurobio.2007.02.001
measures and magnetic resonance imaging atrophy both pre- dict time‐to‐
Grundke‐Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y. C., Zaidi, M. S., & progression from mild cognitive impairment to Alzheimer's disease. Brain, 133,
Wisniewski, H. M. (1986). Microtubule‐associated protein tau. A com- ponent 3336–3348. https://doi.org/10.1093/ brain/awq277
of Alzheimer paired helical filaments. The Journal of Biological Chemistry, Jiang, Q., Lee, C. Y. D., Mandrekar, S., Wilkinson, B., Cramer, P., Zelcer, N.,
261, 6084–6089. … Landreth, G. E. (2008). ApoE promotes the proteolytic degradation of Aβ.
Guerreiro, R., & Hardy, J. (2014). Genetics of Alzheimer's disease. Neuron, 58, 681–693. https://doi.org/10.1016/j. neuron.2008.04.010
Neurotherapeutics, 11, 732–737. https://doi.org/10.1007/s13311‐ 014‐0295‐9 Jonsson, T., Atwal, J. K., Steinberg, S., Snaedal, J., Jonsson, P. V., Bjornsson, S., …
Hall, A., & Patel, T. R. (2014). γ‐Secretase modulators. Progress in medicinal Stefansson, K. (2012). A mutation in APP protects against Alzheimer's
chemistry, 53, 101–145. https://doi.org/10.1016/B978‐0‐444‐63380‐ 4.00003‐2 disease and age‐related cognitive decline. Nature, 488, 96–99.
https://doi.org/10.1038/nature11283
Harding, S. D., Sharman, J. L., Faccenda, E., Southan, C., Pawson, A. J., Ire- land,
S., … NC‐IUPHAR. (2018). The IUPHAR/BPS Guide to Jorissen, E., Prox, J., Bernreuther, C., Weber, S., Schwanbeck, R., Serneels, L., …
PHARMACOLOGY in 2018: Updates and expansion to encompass the new Saftig, P. (2010). The disintegrin/metalloproteinase ADAM10 is essential for
guide to IMMUNOPHARMACOLOGY. Nucl Acids Res, 46, D1091–D1106. the establishment of the brain cortex. The Journal of Neu- roscience, 30,
https://doi.org/10.1093/nar/gkx1121 4833–4844. https://doi.org/10.1523/JNEUROSCI.5221‐
09.2010
Hardy, J., & Allsop, D. (1991). Amyloid deposition as the central event in the
aetiology of Alzheimer's disease. Trends in Pharmacological Sciences, 12, 383– Karran, E., Mercken, M., & De Strooper, B. (2011). The amyloid cascade
388. hypothesis for Alzheimer's disease: An appraisal for the development of
therapeutics. Nature Reviews. Drug Discovery, 10, 698–712.
Hardy, J., & Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer's dis- ease:
Progress and problems on the road to therapeutics. Science (80‐.)., 297, 353– Kennedy, M. E., Stamford, A. W., Chen, X., Cox, K., Cumming, J. N.,
356. Dockendorf, M. F., … Forman, M. S. (2016). The BACE1 inhibitor
verubecestat (MK‐8931) reduces CNS β‐amyloid in animal models and in
Holmes, C., Boche, D., Wilkinson, D., Yadegarfar, G., Hopkins, V., Bayer, A.,
Alzheimers disease patients. Science Translational Medicine, 8, 363ra150–
… Nicoll, J. A. R. (2008). Long‐term effects of Aβ42 immunisation in
363ra150. https://doi.org/10.1126/scitranslmed.aad9704
Alzheimer's disease: Follow‐up of a randomised, placebo‐controlled phase I
trial. Lancet, 372, 216–223. https://doi.org/10.1016/S0140‐ 6736(08)61075‐2 Klunk, W. E., Wang, Y., Huang, G. F., Debnath, M. L., Holt, D. P., & Mathis,
C. A. (2001). Uncharged thioflavin‐T derivatives bind to amyloid‐beta protein
Holtzman, D. M., Herz, J., & Bu, G. (2012). Apolipoprotein E and apolipo- protein E
with high affinity and readily enter the brain. Life Sciences, 69, 1471–1484.
receptors: Normal biology and roles in Alzheimer disease. Cold Spring Harbor https://doi.org/10.1016/S0024‐3205(01)01232‐2
Perspectives in Medicine, 2, a006312.
Kuhn, P.‐H., Wang, H., Dislich, B., Colombo, A., Zeitschel, U., Ellwart, J. W.,
Hong, L., Koelsch, G., Lin, X., Wu, S., Terzyan, S., Ghosh, A. K., … Tang, J. … Lichtenthaler, S. F. (2010). ADAM10 is the physiologically relevant,
(2000). Structure of the protease domain of memapsin 2 (β‐secretase) constitutive α‐secretase of the amyloid precursor protein in primary neurons.
complexed with inhibitor. Science, 290, 150–153. https://doi.org/ The EMBO Journal, 29, 3020–3032. https://doi.org/ 10.1038/emboj.2010.167
10.1126/science.290.5489.150
Lee, M., Bard, F., Johnson‐Wood, K., Lee, C., Hu, K., Griffith, S. G., … Seubert,
Honig, L. S., Vellas, B., Woodward, M., Boada, M., Bullock, R., Borrie, M., … P. (2005). Aβ42 immunization in Alzheimer's disease generates Aβ N‐terminal
Siemers, E. (2018). Trial of solanezumab for mild dementia due to antibodies. Annals of Neurology, 58, 430–435. https://
Alzheimer's disease. The New England Journal of Medicine, 378, 321–330. doi.org/10.1002/ana.20592
https://doi.org/10.1056/NEJMoa1705971
Lendon, C. L., Ashall, F., & Goate, A. M. (1997). Exploring the etiology of
Hussain, I., Powell, D., Howlett, D. R., Tew, D. G., Meek, T. D., Chapman, C., Alzheimer disease using molecular genetics. Jama, 277, 825–831.
… Christie, G. (1999). Identification of a novel aspartic protease (Asp 2) as β‐
Levites, Y., Smithson, L. A., Price, R. W., Dakin, R. S., Yuan, B., Sierks, M. R.,
secretase. Molecular and Cellular Neuroscience, 14, 419–427.
… Golde, T. E. (2006). Insights into the mechanisms of action of anti‐Aβ
https://doi.org/10.1006/mcne.1999.0811
antibodies in Alzheimer's disease mouse models. The FASEB Journal, 20, 2576–
Hutton, M., Lendon, C. L., Rizzu, P., Baker, M., Froelich, S., Houlden, H., … 2578. https://doi.org/10.1096/fj.06‐6463fje
Heutink, P. (1998). Association of missense and 5′‐splice‐site mutations in tau with
Lin, X., Koelsch, G., Wu, S., Downs, D., Dashti, A., & Tang, J. (2000). Human
the inherited dementia FTDP‐17. Nature, 393, 702–705.
aspartic protease memapsin 2 cleaves the β‐secretase site of β‐amyloid
https://doi.org/10.1038/31508
precursor protein. Proceedings of the National Academy of Sciences of the
Iqbal, K., Grundke‐Iqbal, I., Zaidi, T., Merz, P. A., Wen, G. Y., Shaikh, S. S., … United States of America, 97, 1456–1460.
Winblad, B. (1986). Defective brain microtubule assembly in Alzheimer's
Lipton, S. A. (2006). Paradigm shift in neuroprotection by NMDA receptor
disease. Lancet (London, England), 2, 421–426.
blockade: Memantine and beyond. Nature Reviews. Drug Discovery, 5, 160–
Irwin, D. J., Grossman, M., Weintraub, D., Hurtig, H. I., Duda, J. E., Xie, S. X., 170. https://doi.org/10.1038/nrd1958
… Trojanowski, J. Q. (2017). Neuropathological and genetic correlates of
Ludewig, S., & Korte, M. (2017). Novel insights into the physiological func- tion of
survival and dementia onset in synucleinopathies: A retrospective analysis.
the APP (gene) family and its proteolytic fragments in synaptic plasticity.
Lancet Neurology, 16, 55–65. https://doi.org/10.1016/ S1474‐
Frontiers in Molecular Neuroscience, 9, 161.
4422(16)30291‐5
Maloney, J. A., Bainbridge, T., Gustafson, A., Zhang, S., Kyauk, R., Steiner,
Jack, C. R., Holtzman, D. M., & Holtzman, D. M. (2013). Biomarker model- ing of
P., … Atwal, J. K. (2014). Molecular mechanisms of Alzheimer disease
Alzheimer's disease. Neuron, 80, 1347–1358. https://doi.org/
protection by the A673T allele of amyloid precursor protein. The Jour- nal of
10.1016/j.neuron.2013.12.003
Biological Chemistry, 289, 30990–31000. https://doi.org/
10.1074/jbc.M114.589069
REYNOLDS
3
Marais, K. (2018). Dementia Discovery Fund attracts $50 million invest- ment from
stable isotope labeling kinetics of human plasma specific to central ner- vous
Bill Gates | Alzheimer's Research UK.
system amyloidosis. Alzheimer's Dement., 13, 841–849. https://
Masliah, E., Sisk, A., Mallory, M., Mucke, L., Schenk, D., & Games, D. (1996). doi.org/10.1016/j.jalz.2017.06.2266
Comparison of neurodegenerative pathology in transgenic mice over- expressing
Panza, F., Solfrizzi, V., Imbimbo, B. P., Giannini, M., Santamato, A., Seripa, D.,
V717F β‐amyloid precursor protein and Alzheimer's disease. The Journal of
& Logroscino, G. (2014). Efficacy and safety studies of gantenerumab in
Neuroscience, 16, 5795–5811. https://doi.org/ 10.1523/JNEUROSCI.16‐18‐
patients with Alzheimer's disease. Expert Review of Neurotherapeutics, 14,
05795.1996
973–986. https://doi.org/10.1586/
McShane, R., Areosa Sastre, A., & Minakaran, N. (2006). Memantine for dementia. 14737175.2014.945522
Cochrane Database of Systematic Reviews, CD003154.
Pasquier, F. (1999). Early diagnosis of dementia: Neuropsychology. Journal of
Micchelli, C. A., Esler, W. P., Kimberely, W. T., Jack, C., Berezovska, O., Neurology, 246, 6–15. https://doi.org/10.1007/s004150050299
Kornilova, A., … Wolfe, M. S. (2003). γ‐Secretase/presenilin inhibitors for
Pietrzak, K., Czarnecka, K., Mikiciuk‐Olasik, E., & Szymanski, P. (2018). New
Alzheimer's disease phenocopy Notch mutations in Drosophila. The FASEB
perspectives of Alzheimer disease diagnosis—The most popular and future
Journal, 17, 79–81. https://doi.org/10.1096/fj.02‐0394fje
methods. Medicinal Chemistry (Los Angeles), 14, 34–43.
Mills, S. M., Mallmann, J., Santacruz, A. M., Fuqua, A., Carril, M., Aisen, P. S.,
Prince, M., Comas‐Herrera, A., Knapp, M., Guerchet, M., & Karagiannidou,
… Bateman, R. J. (2013). Preclinical trials in autosomal dominant AD:
M. (2016). World Alzheimer report 2016: Improving healthcare for people living
Implementation of the DIAN‐TU trial. Revue Neurologique (Paris), 169,
with dementia. Coverage, quality and costs now and in the future. Alzheimer's
737–743. https://doi.org/10.1016/j.neurol.2013.07.017
Dis. Int. 1–140.
Morris, E., Chalkidou, A., Hammers, A., Peacock, J., Summers, J., & Keevil, S.
Rapoport, M., Dawson, H. N., Binder, L. I., Vitek, M. P., & Ferreira, A. (2002).
(2016). Diagnostic accuracy of 18F amyloid PET tracers for the diagno- sis of
Tau is essential to β‐amyloid‐induced neurotoxicity. Proceedings of the National
Alzheimer's disease: A systematic review and meta‐analysis. European Journal
Academy of Sciences, 99, 6364–6369.
of Nuclear Medicine and Molecular Imaging, 43, 374–385.
https://doi.org/10.1007/s00259‐015‐3228‐x Reiman, E. M., Langbaum, J. B., Tariot, P. N., Lopera, F., Bateman, R. J.,
Morris, J. C., … Weninger, S. (2016). CAP—Advancing the evaluation of
Muir, J. L., Everitt, B. J., & Robbins, T. W. (1994). AMPA‐induced excitotoxic
lesions of the basal forebrain: A significant role for the cortical choliner- gic system preclinical Alzheimer disease treatments. Nature Reviews Neurology, 12, 56–61.
https://doi.org/10.1038/nrneurol.2015.177
in attentional function. The Journal of Neuroscience, 14, 2313–2326.
https://doi.org/10.1523/JNEUROSCI.14‐04‐02313.1994 Rice, L., & Bisdas, S. (2017). The diagnostic value of FDG and amyloid PET in
Alzheimer's disease—A systematic review. European Journal of Radiol- ogy, 94,
Nakamura, A., Kaneko, N., Villemagne, V. L., Kato, T., Doecke, J., Doré, V.,
16–24. https://doi.org/10.1016/j.ejrad.2017.07.014
… Yanagisawa, K. (2018). High performance plasma amyloid‐β bio- markers for
Alzheimer's disease. Nature, 554, 249–254. https://doi. Ryman, D. C., Acosta‐Baena, N., Aisen, P. S., Bird, T., Danek, A., Fox, N. C.,
org/10.1038/nature25456 … and the Dominantly Inherited Alzheimer Network. (2014). Symptom onset in
autosomal dominant Alzheimer disease: A systematic review and meta‐analysis.
Nelson, P. T., Alafuzoff, I., Bigio, E. H., Bouras, C., Braak, H., Cairns, N. J., …
Neurology, 83, 253–260. https://doi.org/10.1212/ WNL.0000000000000596
Beach, T. G. (2012). Correlation of Alzheimer disease neuropathologic changes
with cognitive status: A review of the literature. Journal of Neuropathology Sahakian, B. J., & Coull, J. T. (1993). Tetrahydroaminoacridine (THA) in
and Experimental Neurology, 71, 362–381. https://doi. Alzheimer's disease: An assessment of attentional and mnemonic function using
org/10.1097/NEN.0b013e31825018f7 CANTAB. Acta Neurologica Scandinavica. Supplementum, 149, 29–35.
Nicoll, J. A. R., Wilkinson, D., Holmes, C., Steart, P., Markham, H., & Weller, R. Salloway, S., Sperling, R., Fox, N. C., Blennow, K., Klunk, W., Raskind, M., …
O. (2003). Neuropathology of human Alzheimer disease after immuniza- tion with Brashear, H. R. (2014). Two phase 3 trials of bapineuzumab in mild‐to‐
amyloid‐β peptide: A case report. Nature Medicine, 9, 448–452. moderate Alzheimer's disease. The New England Journal of Medicine, 370,
O'Brien, J. T., Firbank, M. J., Davison, C., Barnett, N., Bamford, C., Donaldson, C., … 322–333. https://doi.org/10.1056/NEJMoa1304839
Lloyd, J. (2014). 18F‐FDG PET and perfusion SPECT in the diagnosis of Salter, M. W., & Stevens, B. (2017). Microglia emerge as central players in brain
Alzheimer and Lewy body dementias. Journal of Nuclear Medicine, 55, 1959– disease. Nature Medicine, 23, 1018–1027. https://doi.org/ 10.1038/nm.4397
1965. https://doi.org/10.2967/jnumed.114.143347
Schenk, D., Barbour, R., Dunn, W., Gordon, G., Grajeda, H., Guido, T., …
Office of National Statistics. (2018). https://www.ons.gov.uk/ Seubert, P. (1999). Immunization with amyloid‐β attenuates Alzheimer‐
peoplepopulationandcommunity/birthsdeathsandmarriages/deaths/ disease‐like pathology in the PDAPP mouse. Nature, 400, 173–177.
bulletins/deathsregisteredinenglandandwalesseriesdr/2017. Accessed 26/02/19. https://doi.org/10.1038/22124
Okamura, N., Harada, R., Ishiki, A., Kikuchi, A., Nakamura, T., & Kudo, Y. (2018). Schott, J. M., & Petersen, R. C. (2015). New criteria for Alzheimer's disease: Which,
The development and validation of tau PET tracers: Current status and future when and why? Brain, 138, 1134–1137.
directions. Clin. Transl. Imaging, 6, 305–316. https:// doi.org/10.1007/s40336‐
Scott, J. D., Li, S. W., Brunskill, A. P. J., Chen, X., Cox, K., Cumming, J. N., …
018‐0290‐y
Stamford, A. W. (2016). Discovery of the 3‐imino‐1,2,4‐thiadiazinane 1,1‐
Ostrowitzki, S., Deptula, D., Thurfjell, L., Barkhof, F., Bohrmann, B., Brooks, dioxide derivative verubecestat (MK‐8931)—A β‐site amyloid pre- cursor
D. J., … Santarelli, L. (2012). Mechanism of amyloid removal in patients with protein cleaving enzyme 1 inhibitor for the treatment of Alzheimer's disease.
Alzheimer disease treated with gantenerumab. Archives of Neurol- ogy, 69, 198– Journal of Medicinal Chemistry, 59, 10435–10450.
207. https://doi.org/10.1001/archneurol.2011.1538 https://doi.org/10.1021/acs.jmedchem.6b00307
Ostrowitzki, S., Lasser, R. A., Dorflinger, E., Scheltens, P., Barkhof, F., Sevigny, J., Chiao, P., Bussière, T., Weinreb, P. H., Williams, L., Maier, M., …
Nikolcheva, T., … and for the SCarlet RoAD Investigators. (2017). A phase
Sandrock, A. (2016). The antibody aducanumab reduces Aβ plaques in
III randomized trial of gantenerumab in prodromal Alzheimer's disease.
Alzheimer's disease. Nature, 537, 50–56. https://doi.org/10.1038/ nature19323
Alzheimer's Research & Therapy, 9, 95. https://doi.org/ 10.1186/s13195‐
017‐0318‐y Sherrington, R., Rogaev, E. I., Liang, Y., Rogaeva, E. A., Levesque, G., Ikeda, M.,
… St George‐Hyslop, P. H. (1995). Cloning of a gene bearing
Ovod, V., Ramsey, K. N., Mawuenyega, K. G., Bollinger, J. G., Hicks, T.,
Schneider, T., … Bateman, R. J. (2017). Amyloid β concentrations and
3 B
missense mutations in early‐onset familial Alzheimer's disease. Nature, 375, 754–
Varley, J., Brooks, D. J., & Edison, P. (2015). Imaging neuroinflammation in
760. https://doi.org/10.1038/375754a0
Alzheimer's disease and other dementias: Recent advances and future directions.
Shi, Y., Yamada, K., Liddelow, S. A., Smith, S. T., Zhao, L., Luo, W., … Alzheimer's & Dementia, 11, 1110–1120.
Holtzman, D. M. (2017). ApoE4 markedly exacerbates tau‐mediated Vassar, R., Bennett, B. D., Babu‐Khan, S., Kahn, S., Mendiaz, E. A., Denis, P.,
neurodegeneration in a mouse model of tauopathy. Nature, 549, 523–527.
… Citron, M. (1999). β‐Secretase cleavage of Alzheimer's amyloid pre- cursor
https://doi.org/10.1038/nature24016
protein by the transmembrane aspartic protease BACE. Science, 286, 735–741.
Siemers, E. R., Sundell, K. L., Carlson, C., Case, M., Sethuraman, G., Liu‐Sei- fert, https://doi.org/10.1126/science.286.5440.735
H., … Demattos, R. (2016). Phase 3 solanezumab trials: Secondary outcomes in Vassar, R., Kuhn, P.‐H., Haass, C., Kennedy, M. E., Rajendran, L., Wong, P. C., &
mild Alzheimer's disease patients. Alzheimer's Dement., 12, 110–120. Lichtenthaler, S. F. (2014). Function, therapeutic potential and cell biology of
https://doi.org/10.1016/j.jalz.2015.06.1893 BACE proteases: Current status and future prospects. Journal of
Sims, R., van der Lee, S. J., Naj, A. C., Bellenguez, C., Badarinarayan, N., Neurochemistry, 130, 4–28. https://doi.org/10.1111/jnc.12715
Jakobsdottir, J., … Schellenberg, G. D. (2017). Rare coding variants in PLCG2, Vellas, B., Black, R., Thal, L. J., Fox, N. C., Daniels, M., McLennan, G., …
ABI3, and TREM2 implicate microglial‐mediated innate immu- nity in AN1792 (QS‐21)‐251 Study Team. (2009). Long‐term follow‐up of patients
Alzheimer's disease. Nature Genetics, 49, 1373–1384. https:// immunized with AN1792: Reduced functional decline in anti- body responders.
doi.org/10.1038/ng.3916 Current Alzheimer Research, 6, 144–151. https://doi.
Sinha, S., Anderson, J. P., Barbour, R., Basi, G. S., Caccavello, R., Davis, D., … org/10.2174/156720509787602852
John, V. (1999). Purification and cloning of amyloid precursor protein β‐ Vellas, B., Carrillo, M. C., Sampaio, C., Brashear, H. R., Siemers, E., Hampel, H., …
secretase from human brain. Nature, 402, 537–540. https://doi. Aisen, P. (2013). Designing drug trials for Alzheimer's disease: What we have
org/10.1038/990114 learned from the release of the phase III antibody trials: A report from the
Sisodia, S. S., & St George‐Hyslop, P. H. (2002). γ‐Secretase, Notch, Aβ and EU/US/CTAD Task Force. Alzheimer's & Dementia, 9, 438–444.
Alzheimer's disease: Where do the presenilins fit in? Nature Reviews. https://doi.org/10.1016/j.jalz.2013.03.007
Neuroscience, 3, 281–290. Vradenburg, G. (2015). A pivotal moment in Alzheimer's disease and dementia:
How global unity of purpose and action can beat the disease by 2025. Expert
Sperling, R., Mormino, E., & Johnson, K. (2014). The evolution of preclinical
Review of Neurotherapeutics, 15, 73–82.
Alzheimer's disease: Implications for prevention trials. Neuron, 84, 608–622.
Ward, A., Tardiff, S., Dye, C., & Arrighi, H. M. (2013). Rate of conversion from
Sperling, R., Salloway, S., Brooks, D. J., Tampieri, D., Barakos, J., Fox, N. C.,
prodromal Alzheimer's disease to Alzheimer's dementia: A sys- tematic review
… Grundman, M. (2012). Amyloid‐related imaging abnormalities in patients
of the literature. Dementia and Geriatric Cognitive Disorders Extra, 3, 320–
with Alzheimer's disease treated with bapineuzumab: A retro- spective analysis.
332.
Lancet Neurology, 11, 241–249. https://doi.org/ 10.1016/S1474‐
4422(12)70015‐7 Weiner, M. W., Aisen, P. S., Jack, C. R., Jagust, W. J., Trojanowski, J. Q., Shaw,
L., … the Alzheimer's Disease Neuroimaging Initiative. (2010). The Alzheimer's
Sperling, R. A., Rentz, D. M., Johnson, K. A., Karlawish, J., Donohue, M., disease neuroimaging initiative: Progress report and future plans. Alzheimer's &
Salmon, D. P., & Aisen, P. (2014). The A4 study: Stopping AD before Dementia, 6, 202–211.e7. https://doi.org/ 10.1016/j.jalz.2010.03.007
symptoms begin? Science Translational Medicine, 6, 228fs13–228fs13.
https://doi.org/10.1126/scitranslmed.3007941 Weingarten, M. D., Lockwood, A. H., Hwo, S. Y., & Kirschner, M. W. (1975). A
protein factor essential for microtubule assembly. Proceedings of the National
Spillantini, M. G., Murrell, J. R., Goedert, M., Farlow, M. R., Klug, A., & Academy of Sciences of the United States of America, 72, 1858–1862.
Ghetti, B. (1998). Mutation in the tau gene in familial multiple system
Weintraub, S., Carrillo, M. C., Farias, S. T., Goldberg, T. E., Hendrix, J. A., Jaeger,
tauopathy with presenile dementia. Proceedings of the National Acad- emy
J., … Randolph, C. (2018). Measuring cognition and function in the preclinical
of Sciences of the United States of America, 95, 7737–7741.
stage of Alzheimer's disease. Alzheimer's & Dementia: Translational
Spires‐Jones, T. L., Attems, J., & Thal, D. R. (2017). Interactions of patho- logical Research & Clinical Interventions, 4, 64–75. https://doi.
proteins in neurodegenerative diseases. Acta Neuropathologica, 134, 187–205. org/10.1016/j.trci.2018.01.003
https://doi.org/10.1007/s00401‐017‐1709‐7
Whitehouse, P. J., Price, D. L., Struble, R. G., Clark, A. W., Coyle, J. T., &
Strittmatter, W. J., Saunders, A. M., Schmechel, D., Pericak‐Vance, M., Delon, M. R. (1982). Alzheimer's disease and senile dementia: Loss of neurons
Enghild, J., Salvesen, G. S., & Roses, A. D. (1993). Apolipoprotein E: High‐ in the basal forebrain. Science, 215, 1237–1239.
avidity binding to beta‐amyloid and increased frequency of type
World Alzheimer Report. (2015). World Alzheimer report 2015: The global impact
4 allele in late‐onset familial Alzheimer disease. Proceedings of the National of dementia | Alzheimer's Disease International.
Academy of Sciences of the United States of America, 90, 1977–1981.
World Health Organization. (2018). The top 10 causes of death.
https://doi.org/10.1073/pnas.90.5.1977
Yan, R., Bienkowski, M. J., Shuck, M. E., Miao, H., Tory, M. C., Pauley, A. M.,
Szaruga, M., Munteanu, B., Lismont, S., Veugelen, S., Horré, K., Mercken, M., …
… Gurney, M. E. (1999). Membrane‐anchored aspartyl protease with
Chávez‐Gutiérrez, L. (2017). Alzheimer's‐causing mutations shift Aβ length by
Alzheimer's disease β‐secretase activity. Nature, 402, 533–537.
destabilizing γ‐secretase‐Aβn interactions. Cell, 170, 443–456.e14.
https://doi.org/10.1038/990107
https://doi.org/10.1016/j.cell.2017.07.004
Zhu, J.‐B., Tan, C.‐C., Tan, L., & Yu, J.‐T. (2017). State of play in Alzheimer's
Tcw, J., & Goate, A. M. (2017). Genetics of β‐amyloid precursor protein in disease genetics. Journal of Alzheimer's Disease, 58, 631–659.
Alzheimer's disease. Cold Spring Harbor Perspectives in Medicine, 7,
a024539. https://doi.org/10.1101/cshperspect.a024539
Thompson, S., Lanctôt, K. L., & Herrmann, N. (2004). The benefits and risks
How to cite this article: Reynolds DS. A short perspective on
associated with cholinesterase inhibitor therapy in Alzheimer's disease. Expert
the long road to effective treatments for Alzheimer's disease. Br J
Opinion on Drug Safety, 3, 425–440. https://doi.org/10.1517/
14740338.3.5.425 Pharmacol. 2019;176:3636–3648. https://doi.org/10.1111/ bph.14581
Tomlinson, B. E., Blessed, G., & Roth, M. (1970). Observations on the brains of
demented old people. Journal of the Neurological Sciences, 11, 205–242.
https://doi.org/10.1016/0022‐510X(70)90063‐8

You might also like