Vous êtes sur la page 1sur 20

REVIEW ARTICLE

published: 20 April 2012


doi: 10.3389/fneur.2012.00063

Alzheimers disease: a clinical practice-oriented review


Lusa Alves 1,2,3 *, Ana Soa A. Correia 1 , Rita Miguel 1 , Paulo Alegria 4 and Paulo Bugalho 1,2,3
1
Servio de Neurologia, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
2
Faculdade de Cincias Mdicas, Universidade nova de Lisboa, Lisboa, Portugal
3
Centro de Estudos de Doenas Crnicas, Lisboa, Portugal
4
Hospital Beatriz ngelo, Loures, Portugal

Edited by: Investigation in the eld of Alzheimers disease (AD), the commonest cause of dementia,
Joo Massano, Centro Hospitalar de
has been very active in recent years and it may be difcult for the clinician to keep up with
So Joo and Faculty of Medicine
University of Porto, Portugal all the innovations and to be aware of the implications they have in clinical practice. The
Reviewed by: authors, thus, reviewed recent literature on the theme in order to provide the clinician with
Paula I. Moreira, University of an updated overview, intended to support decision-making on aspects of diagnosis and
Coimbra, Portugal management. This article begins to focus on the concept of AD and on its pathogenesis.
Stefano F. Cappa, Vita-Salute San
Afterward, epidemiology and non-genetic risk factors are approached. Genetics, including
Raffaele University, Italy
Gainotti Guido, Policlinico Gemelli, genetic risk factors and guidelines for genetic testing, are mentioned next. Recommenda-
Italy tions for diagnosis of AD, including recently proposed criteria, are then reviewed. Data on
*Correspondence: the variants of AD is presented. First approach to the patient is dealt with next, followed
Lusa Alves, Servio de Neurologia, by neuropsychological evaluation. Biomarkers, namely magnetic resonance imaging, sin-
Hospital de Egas Moniz, Centro
gle photon emission tomography, FDG PET, PiB PET, CSF tau, and A analysis, as well as
Hospitalar de Lisboa Ocidental, Rua
da Junqueira 126, 1349-019 Lisboa, available data on their diagnostic accuracy, are also discussed. Factors predicting rate of dis-
Portugal. ease progression are briey mentioned. Finally, non-pharmacological and pharmacological
e-mail: asiulselva@gmail.com treatments, including established and emerging drugs, are addressed.
Keywords: Alzheimer, AD pathogenesis, AD genetics, AD diagnosis, AD variants, AD neuropsychological evaluation,
AD biomarkers, AD treatment

CONCEPT OF ALZHEIMERS DISEASE clinical course of cognitively normal persons remains to be estab-
The present conceptualization of Alzheimers disease (AD) is based lished, and it is acknowledged that some of these individuals will
on autopsy ndings of widespread neuritic plaques and neurob- never exhibit clinical symptoms in their lifetime (Sperling et al.,
rillary tangles (NFT), described for the rst time in 1906 by Alois 2011).
Alzheimer in a case with early symptom onset (Alzheimer et al.,
1995). The concept was subsequently generalized to late-onset PATHOGENESIS
cases when Blessed et al. observed identical pathology in elderly AD is the most frequent cerebral proteopathy (Jucker and Walker,
patients (Blessed et al., 1968; Seshadri et al., 2011). 2011). Macroscopically, the AD brain is characterized by atro-
The term AD may have distinct meanings in different con- phy of the hippocampal formation and of the cerebral cortex,
texts. AD has, in some settings, referred to the neuropathological primarily involving the fronto-temporal association cortex, com-
criteria for AD, and, in other, to the clinical syndrome of progres- bined with ventricular enlargement, especially of the temporal
sive cognitive decline, typically at the stage of AD dementia (Sper- horn, all of these ndings being greater than expected for age
ling et al., 2011). The National Institute on Aging-Alzheimers (Walsh and Selkoe, 2004; Perl, 2010). Microscopically, its neu-
Association (NIA-AA) workgroup on diagnostic guidelines for AD ropathological hallmarks are the combined presence of extracel-
decided to dene AD as encompassing the underlying pathophysi- lular amyloid-containing plaques and intraneuronal NFTs, the
ological disease process (Sperling et al., 2011) as opposed to having latter being formed by abnormally hyperphosphorylated tau pro-
AD connote only the clinical stages of disease as proposed by the tein (Terry, 1963; Alzheimer et al., 1995). The -amyloid (A)
International Working Group for New Research Criteria for the peptide and tau protein are thought to play a critical role in AD
Diagnosis of AD (Dubois et al., 2010). The NIA-AA workgroup development, but several other mechanisms of neurodegenera-
thus considers that AD can be used to refer to dementia stages, as tion have been proposed, including pro-inammatory responses
well as to MCI and pre-MCI phases. (Wyss-Coray, 2007), mitochondrial dysfunction (Reddy, 2011),
Evidence from genetic at-risk and aging cohorts suggests that oxidative stress (Cai et al., 2011), genetic and environmental factors
there may be a time lag of at least a decade between the begin- (Nelson et al., 2011), and apoptosis (Cai et al., 2011). The deleteri-
ning of the pathological cascade of AD and the onset of clinical ous effects of these pathological changes provide the substrate for
impairment. The NIA-AA workgroup postulates that individu- the etiopathogenesis of AD, and converge, ultimately, to synaptic
als with biomarker evidence of early AD pathology (AD-P) are dysfunction and neuronal cell loss (Gtz et al., 2004; Walsh and
at increased risk of progression to AD dementia (AD-D). How- Selkoe, 2004; LaFerla and Oddo, 2005; Arendt, 2009; Takahashi
ever, the ability of the biomarkers of AD-P to predict the ulterior et al., 2010). This process occurs in particularly vulnerable brain

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 1


Alves et al. Alzheimers disease: a review

areas, such as those responsible for memory and cognition, namely in vitro studies suggest that, in the early stages of AD, A oligomers,
the limbic and association cortices and some subcortical nuclei through a potent pro-inammatory response induction, attenu-
with large cortical projections (Walsh and Selkoe, 2004; LaFerla, ate microglial phagocytic function and, consequently, impair the
2010; Perl, 2010). clearance of brillar A, promoting its deposition in the brain
The pathological interaction between A42 and tau proteins (Pan, 2011). In animal models, A oligomers can be found in the
and their relative contribution to neurodegeneration, synaptic and hipoccampal CA1 region and in the entorhinal cortex, prior to the
neuronal loss have been extensively investigated, but still remain development of amyloid plaques and NFT (Wirths et al., 2001).
to be completely elucidated. There is also robust evidence, from studies involving transgenic
The -amyloid protein, a physiological peptide with a char- mice and/or human AD patients, demonstrating that the early
acteristic -pleated sheet conguration, derives from sequential accumulation of intraneuronal A oligomers can induce down-
cleavage of the amyloid precursor protein (APP) by secretase, stream effects, such as mitochondrial dysfunction (LaFerla et al.,
followed by secretase (Glenner and Wong, 1984). A can vary in 2007; Amadoro et al., 2012), microgliosis and astrocytosis (Walsh
length at the c-terminus, according to the pattern of cleavage of and Selkoe, 2004), free radicals formation, oxidative stress and
APP. The A140 isoform (with a total of 40 amino acid residues) hyperphosphorylation of tau protein (Walsh and Selkoe, 2004;
is the most prevalent, followed by the A142 isoform (with 42 LaFerla and Oddo, 2005), synaptic dysfunction and neurotrans-
amino acid residues). The latter has hydrophobic properties and mitter decits (Walsh and Selkoe, 2004; Bao et al., 2012), leading
aggregates more readily than the A140 isoform, which turns it to synaptic dysruption and cognitive decline (Walsh and Selkoe,
more amyloidogenic and prone to polymerize (Perl, 2010). A 2004; LaFerla and Oddo, 2005; Arendt, 2009).
that escapes from proteolytic degradation aggregates and polymer- Besides amyloid plaques, the other major histopathological
izes in various structurally distinct forms, including oligomeric, hallmark of AD consists of intraneuronal neurobrillary lesions,
protobrillar, amylospheroid, and brillar forms. which appear as NFT in soma or apical dendrites, as neuropil
In the AD brain, neuritic plaques are composed of a central threads in distal dendrites and associated with A plaques in
core containing -amyloid protein, surrounded by clusters of dys- dystrophic neurites. These proteinaceous aggregates consist of
trophic axons and dendrites (or neurites) and by glial recruitment paired helical laments, formed by hyperphosphorylated tau pro-
(LaFerla and Oddo, 2005; Perl, 2010). A deposits also tend to tein. Tau is a microtubule-associated protein, responsible for the
accumulate in the walls of the leptomeningeal, cerebral cortical assembly and stability of microtubules in the neuronal cell and
and cerebellar blood vessels. Cerebral amyloid angiopathy is cor- for axoplasmatic transport. The microtubule connection is reg-
related with AD pathogenesis and may lead to vascular rupture and ulated by a complex interplay of isoform tau expression and
multiple lobar hemorrhages (Nicoll et al., 2004). The A deposi- tau phosphorylation (Perl, 2010). In the AD brain, tau pro-
tion on parenchyma or vascular walls in the brain appears to result tein becomes abnormally hyperphosphorylated at several Ser/Thr
from an increased anabolic activity or a decreased catabolic activity residues, detaches from axonal microtubules and aggregates into
of A. insoluble NFT. These changes result in disruption of axonal trans-
Several mutations involving the APP gene, or genes encod- port and intracellular organelles, including mitochondria (Reddy,
ing secretase complex components, can promote the amyloido- 2011). Several phosphokinases have been implicated in tau hyper-
genic pathway and increase the A42 /A40 ratio, promoting its phosphorylation, namely glycogen synthase kinase 3 (GSK3),
aggregation. The study of the early onset familiar forms of AD, cyclin dependent kinase 5 (CDK5) and extracellular signal-related
histopathologically indistinguishable from the sporadic form, pro- kinase 2 (ERK2; Ballard et al., 2011a). Tau protein is the main
vides compelling evidence for the A protein role in the initiation constituent of NFT, but other proteins have been identied, such
of the neurotoxic cascade, corroborated by experiments involving as ubiquitin (Perry et al., 1987), cholinesterases (Mesulam and
animal and tissue-culture models (Gtz et al., 2004). However, for Moran, 1987) and A4 amyloid protein (Hyman et al., 1989).
the AD sporadic form, which is the more prevalent, totaling more There is evidence, based on an animal and tissue-culture study,
than 95% of cases, the pathogenic trigger remains unidentied that neurobrillar degeneration may trigger or facilitate multi-
(Gtz et al., 2004). ple pathological changes, including intraneuronal A deposition,
The amyloid cascade hypothesis postulates that the excessive oxidative damage and glial activation, all of which can participate
formation and deposition of insoluble brillar A, with conse- in mitochondrial dysfunction and neuronal damage (Gtz et al.,
quent aggregation in plaques, is the initiating event in AD patho- 2004).
genesis. This rst insult triggers, secondarily, a neurotoxic cascade, Contrarily to what was observed for amyloid plaques, severity
including NFT formation, which ultimately leads to synaptic and of dementia has been strongly correlated with NFT density (in
neuronal loss in critical areas related with cognitive functions like studies involving human AD patients; Nagy et al., 1996), as well as
memory (Herrup, 2010). However, neuropathological investiga- with soluble oligomeric A (Arendt, 2009).
tions have found a weak correlation between cerebral amyloid Tau deposition and neurodegeneration occur in stereotyped
plaque burden and the severity of dementia (Terry et al., 1991; fashion, progressing over six stages: stages III represent the clin-
Nagy et al., 1996; Ingelsson, 2004). Moreover, evidence from earlier ically silent involvement of transentorhinal cortex; stages IIIIV
studies has suggested that the formation of soluble non-brillar are characterized by lesions in entorhinal/transentorhinal regions
A42 assemblies, termed oligomers and composed of small aggre- and correspond to the phase of mild cognitive decline; in stages
gates of 212 A peptides, rather than insoluble amyloid plaques, VVI, there is severe neocortical destruction and fully developed
may play a pivotal role in the AD neurodegenerative cascade. Some dementia (Braak and Braak, 1995; Perl, 2010). Recent investigation

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 2


Alves et al. Alzheimers disease: a review

involving human brains indicates that pre-tangle material, able to Late-onset AD has a substantial genetic component, with an
induce NFT pathology, develops early in noradrenergic projection estimated heritability of 5879% (Gatz et al., 2006; Wingo et al.,
neurons of the locus coeruleus, before involvement of the transen- 2012). It is probably governed by an array of low penetrance com-
torhinal region (Braak and Del Tredici, 2011). Tau pathology may mon risk alleles across a number of different loci (Avramopoulos,
then progress in a prodromical phase, during ve or more decades, 2009). In the early 1990s, the association between the APOE gene
until it reaches a clinical threshold (Hyman and Gomz-Isla, 1994; and late-onset AD was described (Corder et al., 1993; Saunders
Braak and Del Tredici, 2011; Nelson et al., 2011). These ndings et al., 1993; Strittmatter et al., 1993a,b). The APOE gene has been
indicate that AD is not a mere extension of normal aging and chal- repeatedly implicated in the pathogenesis of AD and a Genome-
lenge the traditional view suggesting that A deposition precedes Wide Association Study (GWAS) conrmed that this is the major
and triggers tau pathology (Gmez-Ramos and Asuncin Morn, susceptibility gene for late-onset forms (Coon et al., 2007). There
2007; Braak and Del Tredici, 2011). are three common alleles of APOE (2, 3, 4), corresponding to
Synaptic loss in the hippocampus and neocortex is widely con- six phenotypes. AD is associated with the 4 allele, the presence
sidered the major correlate of cognitive decline (Terry et al., 1991; of which increases the risk and reduces the average age at onset of
Coleman and Yao, 2003; Walsh and Selkoe, 2004). Defects in synap- AD in a dose-dependent manner (carriers of two APOE 4 alleles
tic transmission occur early in the disease, before the deposition of have a higher risk and an earlier onset of AD than heterozygous
amyloid plaques or NFTs, and progress slowly (Walsh and Selkoe, subjects). Estimates of the increased risk conferred by the APOE 4
2004; Arendt, 2009). Several studies using AD human samples have vary widely. In a recent study, the lifetime risk of AD at the age of
found reduced expression of a group of genes encoding proteins 85 ranged from 51 to 52% for APOE 4/4 male carriers to 6068%
involved in synaptic vesicle trafc, with consequent depletion of for APOE 4/4 female carriers and from 22 to 23% for APOE 4/3
neurotransmitter systems and synaptic/neuronal loss (Coleman male carriers to 3035% for APOE 4/3 female carriers. The odds
and Yao, 2003; Yao et al., 2003). Synaptic failure may lead to dis- ratio for AD of onset between the ages of 60 and 69 was of 35.1 in
ruption of neuronal circuits and subsequently result in cognitive APOE 4 homozygotes and of 4.2 in 4/3 heterozygotes (Genin
decline, even before structural cellular lesions (Yao et al., 2003; et al., 2011). APOE 4 allele is neither necessary nor sufcient for
Arendt, 2009). developing AD (Corder et al., 1993; Farrer et al., 1997).
Studies on AD human samples indicate a strong anatomical The early onset forms comprise about 67% of all cases of
correlation between synaptic loss markers and tangle forma- AD (Campion et al., 1999; Nussbaum and Ellis, 2003). Autoso-
tion (Honer et al., 1992; Callahan et al., 1999, 2002; Coleman mal dominant disease is usually found in early onset AD families
and Yao, 2003). Furthermore, investigation using animal models (Bertram and Tanzi, 2005). However, almost 40% of the patients
has demonstrated the combined occurrence of A42 and hyper- with early onset AD are sporadic cases (with a negative family
phosphorylated tau in hippocampal CA1 region, within neurites history; van Duijn et al., 1994; Campion et al., 1999). Three genes
and postsynaptically, during the early stages of AD pathogenesis have been implicated in early onset AD: APP, presenilin 1 (PSEN1),
(Takahashi et al., 2010). and presenilin 2 (PSEN2). The discovery of the rst autosomal
Despite major advances in our understanding of the AD neu- dominant mutations in APP (Goate et al., 1991) was followed
ropathology, it is still a matter of great debate and much remains by the identication of autosomal dominant mutations in the
to be explained. PSEN1 (Schellenberg et al., 1992; Sherrington et al., 1995) and
PSEN 2 genes (Levy-Lahad et al., 1995a,b; Rogaev et al., 1995).
GENETICS The mutations in any of these three genes result in a shift in the
GENETIC RISK FACTORS metabolism of APP such that more of a 42 aminoacid form of
AD can be divided into early (<6065 years) and late (>60 A is produced (Hardy, 1997). These mutations cause AD with
65 years) onset forms. According to family history,AD cases may be nearly complete penetrance. Nevertheless, they may present with
classied as autosomal dominant, familial or sporadic (Goldman heterogeneous phenotypes (Sherrington et al., 1996; Tedde et al.,
et al., 2011; Table 1). 2003), and recently an APP mutation that causes disease only in

Table 1 | Alzheimer Disease Genetics Division according to the American College of Medical Genetics and the National Society of Genetic
Counselors.

Feature Definition

Age of onset Early onset (67%)* <6065 years


Late-onset (9394%)* >6065 years
Family history Autosomal dominant (<5%) Disease that occurs in at least three individuals in two or more generations, with two of the individuals
being rst-degree relatives of the third
Familial (1525%) Disease that occurs in more than one individual and at least two of the affected individuals are
third-degree relatives or closer
Sporadic (75%) Isolated case in the family or cases separated by more than three degrees of relationship

*Campion et al. (1999), Nussbaum and Ellis (2003).

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 3


Alves et al. Alzheimers disease: a review

the homozygous state was described (Di Fede et al., 2009). In (3) Synaptic dysfunction and cell membrane processes (impli-
contrast to mutations in PSEN1, mutations in PSEN2 are a rela- cated genes: PICALM, BIN1, CD33, CD2AP, and EPHA1),
tively rare cause of familial AD (Sherrington et al., 1996; Campion including endocytosis (Hollingworth et al., 2011a). Although
et al., 1999). The age of symptom onset in patients with PSEN1 these processes diverge from the amyloid hypothesis, it was
mutations is generally 2565 years and it is similar among affected suggested that toxic A may have a modulatory effect on them
members of the same family. In contrast, individuals with PSEN2 (Morgan, 2011).
are typically older at presentation (4588 years) and the age of
onset is variable among relatives of the same family (Sherrington SORL1 is involved in the processing and trafcking of APP into
et al., 1996). Mutations in APP, PSEN1, and PSEN2 are found in recycling pathways, as demonstrated in a study involving in vitro
up to 82% of patients with autosomal dominant AD. It is likely experiments, mice models and human AD samples (Andersen
that additional genes inuence the pathophysiology of early onset et al., 2005). A recent meta-analysis of genetic data from case-
AD (Janssen et al., 2003; Goldman et al., 2011). APOE gene is a control studies suggests that mutations in SORL1 may play a role
risk factor for early onset AD (van Duijn et al., 1994) with odds in the pathogenesis of late-onset AD (Reitz et al., 2011), but their
ratio of 5.6 in 4 homozygous individuals and of 2.1 in heterozy- effect on the risk of disease seems to be modest (Rogaeva et al.,
gous 4/3 cases (Genin et al., 2011). A recent study estimates that 2007).
the early onset AD heritability is of 92100%, which is compati- Recently, a variable-length poly-T (deoxythymidine homopoly-
ble with an almost entirely genetically based disease. However, the mer) polymorphism in the TOMM40 gene, which is located next
concordance among siblings is of 21.6% and between parents and to the APOE gene in a region of strong linkage disequilibrium,
offspring of 10% or less. The authors (Wingo et al., 2012) consider was described and found to be associated with the age of onset of
that the most likely explanation for these results is that approxi- late-onset AD (Lutz et al., 2010; Roses, 2010; Roses et al., 2010).
mately 90% of early onset AD cases are due to autosomal recessive This association is still uncertain and it is unknown whether the
causes. poly-T repeat affects risk of AD through an APOE-dependent or
Since 2009, four GWAS and a three-stage analysis of the GWAS a totally independent mechanism (Cruchaga et al., 2011).
resulted in the identication of nine novel loci associated with
late-onset AD: CLU, PICALM, CR1, BIN1, ABCA7, MS4A cluster
(MS4A6A/MS4A4E), CD2AP, CD33, and EPHA1 (Harold et al., GENETIC TESTING
2009; Lambert et al., 2009; Seshadri et al., 2010; Hollingworth The American College of Medical Genetics and the National Soci-
et al., 2011a,b; Naj et al., 2011). Other studies replicated some ety of Genetic Counselors have recently dened guidelines con-
of these associations (Carrasquillo et al., 2010, 2011a,b; Corne- cerning genetic counseling and testing for AD (Goldman et al.,
veaux et al., 2010; Jun et al., 2010; Antnez et al., 2011a,b; Lambert 2011). Tests for genes associated with early onset AD (currently
et al., 2011; Kamboh et al., 2012). Examining the amount of APP, PSEN1, and PSEN2) are useful for: (1) symptomatic patients
genetic risk effect attributable to these genes (other than APOE), with early onset AD; (2) individuals with a family history of
the most strongly associated single-nucleotide polymorphisms at dementia with one or more cases of early onset AD; (3) individuals
each locus have population attributable fractions between 2.72 with a relative affected by a known mutation of APP, PSEN1, or
and 5.97%, with a cumulative population attributable fraction PSEN2.
for non-APOE loci estimated to be as much as 35% (Naj et al., In summary, AD genetic risk factors may be divided into: (1)
2011). rare autosomal dominant mutations (APP, PSEN1, PSEN2)
It has been suggested (Lambert et al., 2009; Lambert and genetic tests available in selected circumstances; (2) common
Amouyel, 2011) that, in familial early onset AD, the A peptides mutations with moderate effect (APOE) genetic testing not
accumulate through overproduction and that, in late-onset forms, recommended at present due to limited clinical utility and poor
the A excessive deposition is related to an insidious impairment predictive value; (3) common mutations with small effect (e.g.,
of clearance of A peptides (Mawuenyega et al., 2010). CLU, PICALM, CR1, BIN1, ABCA7, MS4A6A, MS4A4E, CD2AP,
Morgan (2011) described three new pathways implicated in CD33, and EPHA1) genes with poor predictive value individually.
late-onset AD: Genetic counseling is essential during the process of genetic
testing for both symptomatic and asymptomatic patients. Pedi-
(1) Immune system function (implicated genes: CLU, CR1, atric testing is not recommended (Goldman et al., 2011).
ABCA7, MS4A cluster, CD33 and EPHA1). Specic immune
responses may be capable of inducing A degradation, avoid- NON-GENETIC RISK FACTORS
ing accumulation of these peptides (Lambert and Amouyel, AD is a multifactorial disorder, whose causes remain largely
2011). It was also suggested that AD risk variants may unknown. Despite extensive research on genetic factors, the vast
cause changes to the complement system, which can re-ignite majority of AD cases are not directly linked to them. Instead,
programmed synaptic loss (Hollingworth et al., 2011b). a complex association between environmental or lifestyle and
(2) Cholesterol metabolism (implicated genes: APOE, CLU, and polygenetic factors seems to play a crucial role in sporadic AD
ABCA7 ). As cholesterol promotes synapse formation, interfer- vulnerability (Launer, 2002; Murray et al., 2011).
ence with cholesterol processing through AD risk gene activity Aging is, by far, the most well established risk factor for the
can be a mechanism of synaptic disintegration (Hollingworth development of sporadic AD. Several studies are unanimous in
et al., 2011b). showing an exponential growth in incidence rates between the

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 4


Alves et al. Alzheimers disease: a review

ages of 65 and 85 years, doubling every 5 years, and no gender dif- when compared to abstinence or heavy drinking, in a J-shaped
ferences in the AD incidence (Launer et al., 1999; Fratiglioni et al., relationship (Xu et al., 2009).
2000; Kawas and Corrada, 2006). In contrast, after the age of 85, Additional risk factors have been suggested, including migraine
the cumulative risk for developing AD seems to decrease only in (Tyas et al., 2001), high intake of saturated fat (Luchsinger and
men. Thus, the female gender is often associated with a higher Mayeux, 2004), high serum homocysteine (Kalmijn et al., 1999)
relative risk of AD (Andersen et al., 1999; Ruitenberg et al., 2001). and brinogen concentrations (Bots et al., 1998), peripheral
Modiable risk factors have received increasing attention. Epi- inammation (Engelhart et al., 2004), atrial brillation (Duron
demiological and clinical studies suggest that vascular and meta- and Hanon, 2010) and head injury (Guo et al., 2000).
bolic disorders are important risk factors for AD. Growing evi- In the elderly population, studies show a strong inverse asso-
dence has emerged suggesting that raised blood pressure (Qiu ciation between the levels of mental, social and physical activity
et al., 2006) and high levels of serum cholesterol (Kivipelto et al., and the dementia risk (Wang et al., 2002; Rovio et al., 2005). The
2005), particularly at adult age, may precede and increase the potential pathway explaining the effects of lifestyle factors on AD
risk of dementia, including AD, in late life. The biological path- risk can be explained by the cognitive reserve hypothesis, which
way linking long-standing hypertension or hypercholesterolemia states that educational or occupational stimulation may lead to a
to AD pathology can be mediated by atherosclerotic lesions and more effective and exible use of brain networks, resulting in an
other vascular changes. These lead to chronic or episodic cerebral increase of cognitive functional reserve against brain pathology or
hypoperfusion and may converge to initiate or accelerate selec- age-related changes. Thus, stimulating environments and physical
tive neurodegenerative processes in the aging brain, particularly exercise can be protective factors and may modulate the thresh-
in genetically susceptible hosts (Iadecola and Gorelick, 2003; de la old of clinical expression of AD pathology (Fratiglioni et al., 2004;
Torre, 2004; Qiu et al., 2006). The hypercholesterolemias effects Stern, 2006).
in AD incidence may also be due to an increased synthesis of A42 ,
through modulation of the cleavage pattern of APP, or to inter- EPIDEMIOLOGY
ference with the transport and metabolism of this peptide (Kuller AD is the most common cause of dementia (Fratiglioni et al., 2000;
and Lopez, 2011). Cardiovascular disease and carotid artery steno- Lobo et al., 2000). By 2005, the Delphi study estimated that there
sis are strong risk factors, supporting the view that chronic cerebral were 24.3 million people worldwide with dementia, with 4.6 mil-
hypoperfusion may promote selective neurodegenerative damages lion new cases arising every year. Among regional populations of
in susceptible brain areas (Ruitenberg et al., 2005) and suggesting individuals aged 60 years, those from North America and West-
that peripheral atherosclerosis biomarkers can be early indicators ern Europe exhibited the highest prevalence of dementia (6.4 and
of a subclinical phase of AD (Qiu et al., 2006; Muller et al., 2007). 5.4%, respectively), followed by those from Latin America (4.9%),
Cerebrovascular disease is an important pathological mechanism China and developing Western Pacic (4.0%) and Eastern Europe
adding severity to AD (Knopman, 2006). Similarly to hyper- (3.83.9%). The annual regional dementia incidence rates (per
tension, overweight and obesity initiated at adulthood seem to 1.000 individuals) were estimated to be 10.5 for North America,
increase the susceptibility for AD (Kivipelto et al., 2005), possibly 9.2 for Latin America, 8.8 for Western Europe, 8.0 for China and
through vascular dysfunction or through the effects of hormonal developing Western Pacic and 7.78.1 for Eastern Europe. It has
compounds that are secreted by the adipose tissue (Gustafson, been calculated that the number of people with dementia may rise
2006). Diabetes and impaired glucose tolerance have also been to 81.1 million by 2040 (Ferri et al., 2005).
associated with increased odds of dementia (Biessels et al., 2006; The estimated lifetime risk of AD is 1011% in males and 14
Qiu et al., 2007). The direct effect of glucose-mediated toxicity and 17% in females at the age of 85 (Genin et al., 2011). Women bear
hyperinsulinemia on amyloid metabolism and AD neurodegener- most of the burden of AD probably due to longer life expectancy
ative processes has been emphasized (Biessels et al., 2006), but the and longer post-diagnosis survival duration. However, reports of
exact pathophysiological mechanisms remain unclear. the association between gender and AD have been controversial
The vascular hypothesis suggests that midlife vascular risk fac- and its unclear whether women have a survival advantage or not
tors and disorders are involved in neurodegeneration, progression (Heyman et al., 1996; Lapane et al., 2001; Brookmeyer et al., 2002;
and clinical presentation of AD. The view that sporadic AD is pri- Larson et al., 2004; Helzner et al., 2008; Xie et al., 2008). In some
marily a vascular disease with neurodegenerative consequences (de studies, age-specic incidence of AD and prevalence controlled
la Torre, 2010) remains controversial, given that both cerebrovas- for age do not differ signicantly by gender (Hebert et al., 2001).
cular disease and AD are very prevalent in the elderly, and thus may Interestingly, a study reported that AD pathology is more likely to
coexist in an important proportion of patients. Furthermore, the be clinically expressed as dementia in women than in men (Barnes
identication of pure forms of AD, i.e., without simultaneous et al., 2005).
vascular disease, does not support a direct relationship between The prevalence and incidence rates for AD increase exponen-
vascular disease and AD (Romn and Royall, 2004). tially with age. AD rates rise from 2.8 per 1000 person-years in the
Concerning tobacco smoking, conicting outcomes have been age group 6569 years to 56.1 per 1000 person-years in the older
reported. Contradicting previous reports, recent studies have than 90-year age group (Kukull et al., 2002).
found that current smoking is associated with increased risk of AD substantially reduces life expectancy and increases the prob-
AD in older people (Cataldo et al., 2010). With regard to alco- ability of being admitted to a nursing home. The median survival
hol intake, the outcomes are also controversial, but suggest that times after diagnosis range from 8.3 years for individuals diag-
light to moderate consumption may decrease the risk of dementia, nosed with AD at the age of 653.4 years for persons diagnosed

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 5


Alves et al. Alzheimers disease: a review

as having AD at the age of 90. Diagnoses of AD at ages 65 and spectrum of the clinical disease. To fulll criteria for probable AD,
90 years are associated with approximately a 67 and 39% reduction a patient must meet the cornerstone clinical criterion A and at least
in median life span, respectively (Brookmeyer et al., 2002). Death one of the supportive biomarker criteria. Criterion A species that
from all causes by the age of 80 is expected in 61% of AD patients there must be an episodic memory decit within test conditions of
and in 30% of the general population. Nursing home admission encoding specicity. The presence of a biological footprint of the
by the age of 80 years is expected for 75% of the surviving AD disease is established either by criterion B (structural imaging),
patients, and only for 4% of the general population (Arrighi et al., criterion C (cerebrospinal uid), criterion D (molecular imag-
2010). ing), or criterion E (dominant mutation within the immediate
family). Apart from the incorporation of biomarkers, two relevant
DIAGNOSIS innovations characterize the Dubois criteria: (1) the presence of
The clinical diagnosis of dementia caused by AD can approach an a progressive memory decit is considered sufcient to make a
accuracy rate of 95%, but only when it is established by highly diagnosis of AD, even if it is the patients only cognitive decit; (2)
experienced clinicians observing selected patients who are gener- the declarative memory impairment necessary for diagnosis is of
ally followed up comprehensively over time. Outside of specialized the medial temporal lobe type (Carlesimo et al., 2011).
centers, AD dementia is correctly diagnosed only in about 50% of In 2011, the NIA-AA workgroup published recommendations
affected individuals (Mayeux et al., 2011). Accurate diagnosis of concerning the denition of the preclinical stages of AD (Sper-
AD is difcult because of the frequent presence, in older adults, ling et al., 2011), the diagnosis of MCI due to AD (MCI-AD;
of co-morbidities that can contribute to cognitive impairment. Albert et al., 2011) and the diagnosis of dementia due to AD (AD
A factor that may further complicate diagnosis is ignorance of dementia; McKhann et al., 2011), which also integrated biomarker
the patients previous baseline, which precludes the clinician from information. According to the NIA-AA workgroup, the major AD
correctly evaluating whether there was cognitive and functional biomarkers can be divided into those related to the process of brain
decline (a requisite incorporating MCI and dementia criteria) or A protein deposition, comprising low cerebrospinal uid (CSF)
not. There may not always be a reliable informant and self-reported A42 and positive positron emission tomography (PET) amyloid
estimates of function can be inaccurate (Mayeux et al., 2011). imaging, and those related to downstream neuronal degeneration
The diagnosis of AD is frequently based on the criteria of the or injury: elevated CSF tau, both total tau (t-tau) and phos-
Diagnostic and Statistical Manual of Mental Disorders, fourth edi- phorylated tau (p-tau); decreased 18uorodeoxyglucose (FDG)
tion (DSM-IV-TR; American Psychiatric Association, 2000) and uptake on PET in the temporoparietal cortex; and dispropor-
on the National Institute of Neurologic and Communicative Dis- tionate atrophy on structural magnetic resonance imaging (MRI)
orders and Stroke Alzheimer Disease and Related Disorders Asso- in medial, basal and lateral temporal lobe, and medial parietal
ciation (NINCDS-ADRDA) criteria (McKhann et al., 1984). Both cortex; McKhann et al., 2011).
sets of criteria require decits in memory and at least one other According to the NIA-AA recommendations (Albert et al.,
cognitive domain. The DSM-IV-TR criteria additionally stipulate 2011), in the presence of a change in cognition, objective impair-
that there must be an impact of the cognitive impairment on ment in at least one cognitive domain, preservation of inde-
social function or activities of daily living (ADL). According to the pendence in ADL (and inherent absence of dementia), clinical
NINCDS-ADRDA criteria, the AD diagnosis is classied as denite syndrome suggestive of AD and examination of potential causes
(clinical diagnosis with histologic conrmation), probable (typi- consistent with AD, an individual is classied as having MCI-AD-
cal clinical syndrome without histologic conrmation), or possible core clinical criteria in the following situations: (1) in the absence
(atypical clinical features but no alternative diagnosis apparent; of information on biomarkers; (2) in the event that they are unin-
no histologic conrmation). The NINCDS-ADRDA criteria have formative (neither clearly negative nor positive); or (3) in the case
been reasonably reliable for the diagnosis of probable AD: across that their information is conicting (e.g., low A and normal tau
more than a dozen clinical-pathological studies, they have had a in CSF). The suggestive clinical syndrome involves typically a
sensitivity of 81% and a specicity of 70% (Knopman et al., 2001). prominent impairment in episodic memory, but other patterns,
However, using the DSM-IV-TR and the 1984 NINCDS- such as visuo-spatial impairment, are also possible manifestations
ADRDA recommendations, the AD cases are discovered late in of underlying AD pathology and, as such, are compatible with a
the disease process. Therefore, substantial efforts have been made diagnosis of MCI-AD. A subject is attributed a diagnosis of MCI-
to create criteria for the clinical stage preceding dementia, i.e., mild AD with intermediate likelihood if he has one positive biomarker
cognitive impairment (MCI; Petersen and Negash, 2008), a state either reecting A deposition or neuronal injury. A person is diag-
in which, by denition, ADL are essentially normal. Nevertheless, nosed with MCI-AD with a high likelihood if both biomarkers are
MCI is a heterogenous entity, encompassing not only AD cases, but positive. An individual is attributed a diagnosis of MCI unlikely
also patients with degenerative diseases other than AD and individ- due to AD if both biomarkers are negative.
uals with non-degenerative causes of cognitive impairment. This Regarding AD dementia, the NIA-AA workgroup (McKhann
issue was solved with the creation of the concept of prodromal et al., 2011) proposes the following terminology: (1) probable AD
AD, which may be considered a subtype of MCI, by Dubois et al. dementia, (2) possible AD dementia, (3) probable or possible AD
(2007). The core principle of the research criteria for the diagnosis dementia with evidence of the AD pathophysiological process.
of AD proposed by this group (Dubois et al., 2007) is based upon The rst two concepts are intended for use in all clinical settings.
the presence of consistent episodic memory disturbance which, The third is to be used for research purposes only. The authors
together with biomarker positivity, recognizes AD across the full underline that dementia from all causes implies the presence of

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 6


Alves et al. Alzheimers disease: a review

cognitive decline from previous levels of performance, detected by The PCA syndrome, rst described by Benson et al. (1988),
history taking and objective assessment, involving a minimum of is characterized by the early appearance (before the age of 60)
two domains, interfering with ADL and not explained by delirium of alexia with agraphia (the most frequent symptoms, which
or major psychiatric disorder. According to McKhann et al. (2011), occur without signicant derangement of other language related
probable AD dementia is diagnosed when the patient meets cri- functions), Balints syndrome (optic ataxia, ocular apraxia, simul-
teria for dementia and, in addition, there is clear-cut history of tanagnosia, and visual agnosia) and Gerstmann syndrome, with
worsening of cognition by report or observation, insidious onset, acalculia as the predominant symptom (Galton et al., 2000; Tang-
and the initial and most prominent cognitive decits are in one Wai et al., 2004; McMonagle et al., 2006; Alladi et al., 2007). Some
of the following categories: memory, language, visuo-spatial func- cases present with predominant parietal dysfunction (involving
tion, or executive function. The NIA-AA workgroup recommends the dorsal stream of visual perception the where stream), mani-
that the diagnosis of probable AD dementia should not be applied fested by agraphia and apraxia, and others show occipital dysfunc-
when there is evidence of substantial concomitant cerebrovascular tion (involving the ventral stream the what stream), expressed
disease, core features of Dementia with Lewy bodies (DLB; other by symptoms of visual agnosia, prosopagnosia, achromatopsia
than dementia itself), prominent features of fronto-temporal lobar and alexia. A third PCA group has been suggested, consisting of
degeneration (FTLD), evidence of another concurrent, active neu- patients with cortical blindness, caused by degeneration of the
rological disease, or of a non-neurological medical comorbidity or occipital primary visual cortex (Galton et al., 2000). MRI stud-
use of medication that could have a substantial effect on cogni- ies have shown atrophy in regions related to the cognitive decits
tion. In persons who meet the core clinical criteria for probable found on neuropsychological evaluation, and relative preservation
AD dementia, the authors (McKhann et al., 2011) postulate an of temporal mesial regions. However, not all cases show conspic-
increased level of certainty of diagnosis in the presence of: docu- uous atrophy on MRI. Functional imaging shows hypofunction
mented decline and/or causative AD genetic mutation. According of the regions responsible for the cognitive decits. Many cases
to the same workgroup, a diagnosis of possible AD dementia is evolve to full-blown dementia, including affection of episodic
made when there is an atypical course, that is, when there is sud- memory and other cognitive domains. Nevertheless, some patients
den onset of cognitive impairment, or insufcient historical detail maintain exclusive derangement of posterior cortical functions
or objective cognitive documentation of progressive decline, or for many years, and some die without presenting the complete
when there is an etiologically mixed presentation. late stage AD pattern (Galton et al., 2000; Alladi et al., 2007).
McKhann et al. (2011) state that, in persons who meet the core Although PCA is dened as a syndrome, caused by different dis-
clinical criteria for probable AD dementia, biomarker evidence eases that affect the posterior cortex (including LBD, prion diseases
may increase the certainty that the basis of the clinical demen- and CBD), neuropathological examination shows a marked pre-
tia syndrome is the AD pathophysiological process. However, the dominance of AD cases (Renner et al., 2004). Renner et al. (2004)
authors do not advocate the use of AD biomarker tests for routine were not able to nd clinical differences between AD and non-AD
diagnostic purposes at the present time. They indicate the reasons PCA cases, although some LBD patients would eventually develop
for this limitation: the very good diagnostic accuracy of the core the visual hallucinations and extra-pyramidal symptoms charac-
clinical criteria; the need for more research validating the design of teristic of the disease (Tang-Wai et al., 2004; McMonagle et al.,
the biomarker incorporating criteria; the limited standardization 2006). AD parietal atrophy cases were also difcult to differentiate
of biomarkers from one site to another; and the limited access to from CBD cases.
biomarkers in many community settings. Primary progressive aphasia was rst described by Mesulam
as a syndrome consisting of progressive deterioration of lan-
AD VARIANTS guage and preservation of other cognitive functions, associated
Typically, AD presents with initial episodic memory dysfunction, with left temporal and frontal lobe atrophy (Mesulam, 2001). As
followed by progressive involvement of other cognitive domains, these cases were primarily associated with the FTLD spectrum
including praxis, visuo-spatial orientation, language, calculation of diseases, the existence of early language decits was usually
and executive functions. Decit progression mirrors the suc- considered as an exclusion criteria for AD. Further investigation
cessive involvement of different brain regions, beginning with has revealed that AD could underlie a higher percentage of PPA
hippocampal damage and spreading to lateral temporal regions, cases than was rst considered. The syndrome of primary apha-
parieto-occipital cortex and frontal lobe structures. However, AD sia was classically divided into two distinct patterns: non-uent
pathology is also found in patients presenting with different clini- aphasia and semantic dementia. Recently, a third pattern was pro-
cal symptoms, caused by initial damage on less frequently involved posed, consisting of patients who seem to share decits belonging
cortical regions. These atypical cases pose diagnostic difculties, to the other categories: slow speech and word nding difcul-
and are frequently identied as non-AD cases, including FTLD, ties (as in non-uent aphasia), with preservation of grammar and
Lewy-body disease (LBD) and cortico-basal degeneration (CBD), phonological functions and presence of decits in naming (sim-
in which non-memory decits are more frequent. Atypical AD ilar to semantic dementia). Difculty in sentence repetition and
patients present in at least two different patterns. The syndrome of in understanding long sentences, with preserved ability to under-
posterior cortical atrophy (PCA) involves dysfunction of the visuo- stand single words are also characteristic. This particular kind of
spatial areas of the occipital and parietal cortices. Primary pro- PPA, named logopenic aphasia, has been linked by imaging and
gressive aphasia (PPA) affects almost exclusively language related functional studies to atrophy of the left temporo-parietal junction,
structures. including the left posterior superior and middle temporal gyri

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 7


Alves et al. Alzheimers disease: a review

and inferior parietal lobule (Henry and Gorno-Tempini, 2010). non-demented and demented individuals in a trustworthy manner
AD pathology could be present in about 2030% of patients with (Galvin et al., 2005), is also helpful.
PPA. Interestingly, it appears to be linked with particular types of Behavioral and psychological symptoms, as well as co-morbid
PPA. While atypical Alzheimer cases rarely present with semantic conditions, should be identied, since they occur in the majority
and non-uent aphasia, AD pathology appears to be common of AD patients and are associated with decline in cognitive and
in logopenic patients, as proven by clinical-pathological studies functional ability (Apostolova and Cummings, 2008), decreased
(Alladi et al., 2007; Mesulam et al., 2008) and by imaging studies quality of life and increased institutionalization (Hort et al., 2010).
using Pittsburgh compound B (PiB; Rabinovici et al., 2008). Hort et al. (2010) also consider important to elicit past med-
A frontal variant of AD has also been reported, characterized ical history, co-morbidities, family and educational history. It is
by prominent executive and behavioral symptoms, mimicking the stressed that neurological and general physical examination are
behavioral variant of FTLD, but presenting with AD pathology helpful in distinguishing AD from other primary degenerative and
and selective deposition of amyloid in frontal regions (Johnson secondary dementias and co-morbidities.
et al., 1999; Alladi et al., 2007). These ndings have not been repli- Blood tests, according to the EFNS guidelines (Hort et al., 2010),
cated in other studies, and the existence of this variant remains are useful in excluding co-morbidities and should include vitamin
controversial. B12, folate, thyroid stimulating hormone, calcium, glucose, com-
The prevalence of AD atypical variants is unknown. In the study plete blood cell count, renal and liver function. Syphilis, Borrelia
by Galton et al. (2000), atypical cases represented 14% of all AD and HIV serological tests should be considered in high risk cases
cases followed in the institution. This was, however, a tertiary cen- or when there are suggestive clinical features.
ter of referral, in which atypical cases could be over-represented. Structural brain imaging is useful in excluding potentially sur-
In the mentioned study, PCA was present in 6 out of 26 atypical gically treatable diseases and in detecting specic ndings for AD.
cases, while 20 had the aphasic variant (Galton et al., 2000). Lit- For the former, CT and MRI are similarly good and it is consen-
tle is known about the relative predominance of AD pathology in sual that such an imaging procedure should be carried out once in
the total number of patients presenting with focal cortical symp- every patient. However, MRI is more sensitive to subtle vascular
toms, as compared to FTLD, DLB, and CBD pathology. In a study changes and to alterations specic of certain conditions. For prac-
performed in 100 patients with focal cortical syndromes, AD was tice purposes, a standard MRI protocol involving at least coronal
present in all PCA cases and in more than half of the mixed aphasic T1 and axial T2 or uid-attenuated inversion recovery sequences
cases (a group which included patients with the logopenic variant). should be used (Hort et al., 2010).
Half of the cortical basal syndrome (CBS) cases, about 40% of the Electroencephalography (EEG) may help to differentiate
patients with the non-uent aphasia syndrome and a small per- between AD, subjective complaints and psychiatric diagnoses. EEG
centage with behavioral FTLD and semantic dementia syndromes can also be useful in the differential diagnosis of atypical clinical
had AD pathology (Alladi et al., 2007). This suggests that, while a presentations of AD. Even though reduced alpha power, increased
diagnosis of PCA represents a high probability of AD, symptoms theta power and lower mean frequency are characteristic of AD
characteristic of FTLD (particularly the progressive non-uent patients, EEG can be normal early in the course of the disease in
aphasia type) or CBS should not rule out this diagnosis. up to 14% of cases (Hort et al., 2010).

FIRST APPROACH TO THE PATIENT NEUROPSYCHOLOGICAL EVALUATION


The European Federation of Neurological Societies (EFNS) guide- Episodic memory should be assessed because it is the function
lines for the diagnosis and management of AD (Hort et al., 2010) most commonly impaired early in AD as a result of dysfunction
recommend that the evaluation of a subject suspected of having of mesial temporal structures, which are responsible for consoli-
AD should include history, from the patient and a close infor- dation. Retrieval, which depends on frontal lobe and subcortical
mant, focused on the affected cognitive domains, the course of the structures, is less affected (Hort et al., 2010).
illness, the impact on the ADL and any associated non-cognitive Impaired delayed recall is not, per se, evidence of an AD-related
symptoms. memory disorder. Authentic decits in encoding and storage
Global assessment of cognitive functions should be undertaken, processes (features typical of AD) must be differentiated from
using, for example, Mini-Mental State Examination (MMSE; Fol- non-AD decits that can also affect delayed recall, including atten-
stein et al., 1975). Such a screening test is useful for the identica- tional difculties, which may be present in depression, and inef-
tion of cases requiring more detailed evaluation. This is done using cient retrieval strategies, associated with normal aging, FTLD, or
neuropsychological tests assessing memory, executive functions, subcortical-frontal dementias (Dubois et al., 2007).
language, praxis and visuo-spatial abilities (Hort et al., 2010). Dubois et al. (2007) proposed the use of a neuropsychological
As the differentiation of dementia from MCI rests on the deter- tool the Grober-Buschke (GB) paradigm (Grober et al., 1988)
mination of whether or not there is signicant interference in the which, controlling for elaborative encoding at study and providing
ability to function at work or in usual daily activities, it is useful to a strong category cue at retrieval, should be able to compensate
assess ADL. However, there is no gold standard available for this for eventual encoding/retrieval decits and allow to identify the
purpose (Hort et al., 2010). In a review by Sikkes et al. (2009), memory impairment that is caused by reduced efciency of con-
out of several systematically reviewed scales, the informant-based solidation of the memory trace (Carlesimo et al., 2011). Within
questionnaires the Disability Assessment for Dementia and the this neuropsychological test paradigm, measures of sensitivity to
Bristol ADL were among the most useful. The AD8, a brief semantic cueing can successfully differentiate patients with AD
informant-based questionnaire that is able to differentiate between from healthy controls (Dubois et al., 2007).

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 8


Alves et al. Alzheimers disease: a review

Sarazin et al. (2007) found that the most sensitive and specic Vemuri et al. (2011) created atrophy maps using structural
test for diagnosis of prodromal AD was the Free and Cued Selective MRI and applied them for classication of new incoming patients.
Recall Reminding Test (FCSRT; Grober and Buschke, 1987). Their They identied 115 pathologically conrmed subjects with a single
study showed that impairment of free recall, total recall, and index dementing pathologic diagnosis (AD, LBD or FTLD-TDP-43) who
of sensitivity of cueing can identify prodromal AD in patients with had an MRI at the time of clinical diagnosis of dementia. Leave-
MCI with high sensitivity (79.7%) and specicity (89.9%). one-out classication showed reasonable performance compared
In a series of studies reviewed by Carlesimo et al. (2011), total to the autopsy gold standard: in AD, sensitivity was 90.7% and
immediate recall scores on the original or modied version of the specicity 84%.
GB paradigm achieved levels of sensitivity between 62 and 100% A recent study (Karow et al., 2010) showed no evidence that
and of specicity between 93.9 and 100% in the discrimination brain FDG PET was more sensitive than MRI to the degenera-
between AD and healthy elderly individuals. However, none of tion occurring in preclinical and mild AD. Thus, MRI, a practical
these studies veried the diagnosis of AD using a postmortem exam, might be used instead of more sophisticated ancillary tests
histologic examination. in clinical practice for early detection of AD.
On the other hand, the review by Carlesimo et al. (2011) New quantitative methods using MRI are promising biomark-
did not completely corroborate the superiority of the GB para- ers. These include: functional MRI and diffusion tensor imaging
digm over more traditional neuropsychological tools for analyzing (Hampel et al., 2008); diffusion weighted imaging, magnetiza-
memory disorders in patients suspected of having AD. In fact, tion transfer MRI and proton magnetic resonance spectroscopy
controversial results emerged from studies that compared the sen- (Kantarci and Jack, 2003); and MR volumetry methods. The latter
sitivity/specicity of the cued recall task in the GB paradigm with encompass cortical thickness measurement, deformation-based
the free recall task in the same or a different experimental para- and voxel-based morphometry (Hampel et al., 2008), as well as
digm in differentiating patients with full-blown AD or amnestic multiple-atlas propagation and segmentation technique (Leung
MCI from healthy individuals. The authors (Carlesimo et al., 2011) et al., 2010).
concluded that the GB procedure was useful for discriminating
whether an isolated memory decit in an elderly person is due to SINGLE PHOTON EMISSION TOMOGRAPHY
incipient AD or to other causes and for helping in the differential The EFNS guidelines (Hort et al., 2010) considered that SPECT
diagnosis between AD and other etiological forms of dementia. could increase diagnostic condence in the evaluation of demen-
In an interesting study involving 150 patients with an objective tia. However, and even though it is more widely available and
history of episodic memory dysfunction (Oksengard et al., 2010), it cheaper than PET, it was not included in the Dubois criteria
was found that delayed recall, objectively assessed by Rey Auditory (Dubois et al., 2007), because of its poor estimated diagnostic
Verbal Learning Test (RAVLT; Rey, 1958), was the most sensitive accuracy. It is not mentioned in the recommendations from the
marker in the detection of AD cases, when compared with MRI, NIA-AA workgroup (McKhann et al., 2011), either.
single photon emission tomography (SPECT), CSF t-tau, A42 , Pooling data in an exploratory manner from two clinical-
and p-tau. pathological studies, Dougall et al. (2004) found a weighted
sensitivity for a positive brain SPECT (that is, one showing a
BIOMARKERS pattern of bilateral temporo-parietal hypoperfusion) of 74% and
MAGNETIC RESONANCE IMAGING a weighted specicity of 91% against neuropathology. In theses
Atrophy of the medial temporal lobe atrophy (MTA) is a well- studies using pathological verication as a gold standard, clinical
recognized feature of AD, though there are limitations concerning criteria were more sensitive than brain SPECT (81 versus 74%)
its specicity, because marked hippocampal atrophy has also been and brain SPECT had a higher specicity than clinical criteria (91
shown in fronto-temporal dementia (Galton et al., 2001), demen- versus 70%).
tia with Lewy bodies (DLB; Barber et al., 2000), Parkinsons disease In the context of other clinical-pathological study, by Jagust
with dementia (Tam et al., 2005), vascular dementia (Barber et al., et al. (2001), the clinical diagnosis of probable AD was associated
2000) and hippocampal sclerosis (Barkhof et al., 2007). However, with an 84% likelihood of pathologic AD. A positive perfusion
only one of these studies was in autopsy-conrmed cases (Barkhof SPECT scan raised this likelihood to 92%, whereas a negative
et al., 2007) and thus misdiagnosis cannot be ruled out. Indeed, at SPECT scan lowered it to 70%. SPECT was more useful when
least two clinical-pathological studies showed high sensitivity and the clinical diagnosis was possible AD, with a likelihood of 67%
specicity rates in the discrimination between AD and non-AD without SPECT, increasing to 84% with a positive SPECT, and
dementias. decreasing to 52% with a negative SPECT.
Burton et al. (2009) assessed the diagnostic specicity of Dopaminergic SPECT imaging is useful to differentiate AD
MTA, rated visually on MRI using a standardized scale (Schel- from DLB with sensitivity and specicity around 85% (Hort et al.,
tens et al., 1992), blind to clinical or autopsy diagnosis, for AD 2010).
among individuals with AD, DLB and vascular cognitive impair-
ment (VCI). The study group consisted of 46 individuals who had POSITRON EMISSION TOMOGRAPHY
both antemortem MRI and an autopsy. Subjects were patholog- FDG PET
ically classied as AD, DLB, or VCI. MTA was a highly accurate In vivo brain FDG PET is a minimally invasive diagnostic
diagnostic marker for autopsy-conrmed AD (sensitivity of 91% imaging procedure used to evaluate cerebral glucose metabolism.
and specicity of 94%). The pattern of metabolic impairment of the posterior cingulate

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 9


Alves et al. Alzheimers disease: a review

and temporo-parietal cortices, including the precuneus, more changes in a similar fraction of cognitively intact elderly indi-
accentuated than frontal cortex decits, together with relative viduals. On the other hand, a small fraction of AD patients do
preservation of the primary sensorimotor and visual cortices, not show any increase in PiB uptake, which may be explained
basal ganglia, and cerebellum, constitutes the distinctive metabolic by inaccurate clinical diagnosis or the fact that PiB does not
phenotype of AD (Bohnen et al., 2012). bind to all brillar A conformations (Prvulovic and Hampel,
Hoffman et al. (2000) studied FDG PET imaging in individ- 2011).
uals with difcult-to-characterize memory loss or dementia who
eventually received pathologic conrmation of diagnosis. The sen- CSF ANALYSIS
sitivity, specicity, and diagnostic accuracy of bilateral temporo- The typical CSF pattern of AD consists of decreased levels of A42
parietal hypometabolism being associated with AD were 93, 63, and increased values of t-tau or p-tau. Studies show that CSF A42
and 82%, respectively. changes before total tau (Isaac et al., 2011).
In a study by Silverman et al. (2001), using neuropathologi- In a multicenter-study, CSF baseline concentrations of p-tau
cal diagnosis as gold standard, PET was 94% sensitive and 73% predicted conversion to AD in subjects with MCI with high accu-
specic in identifying AD. racy (80%) during an observation interval of 1.5 years (Ewers et al.,
According to a study by Minoshima et al. (2001), PET could 2007; Prvulovic and Hampel, 2011).
distinguish autopsy-conrmed AD and DLB patients with 90% A prospective cohort study (Visser et al., 2009) found that the
sensitivity and 80% specicity. CSF AD prole could identify patients with potential AD type
Another study, by Jagust et al. (2007), reported results of a dementia among patients with MCI at sensitivities in the range of
mixed sample of subjects with variable levels of cognitive impair- 8891% and specicities between 52 and 90%.
ment, who eventually underwent autopsy. Results showed that In a large cross-sectional study involving patients with AD,
PET had sensitivity of 84% and specicity of 74% for the patho- fronto-temporal dementia (FTD) and DLB, CSF concentrations
logic diagnosis of AD. The clinical diagnosis of AD was associated of p-tau-181 discriminated between DLB and AD with a sensi-
with a 70% probability of detecting AD pathology; with a pos- tivity of 94% and a specicity of 64%, while CSF concentrations
itive PET scan this increased to 84%, and with a negative PET of p-tau-231 performed particularly well in the separation of AD
scan this decreased to 31%. A diagnosis of not AD at ini- and FTD groups, with a sensitivity of 88% and a specicity of 92%
tial clinical evaluation was associated with a 35% probability (Hampel et al., 2004).
of AD pathology, increasing to 70% with a positive PET scan. Welge et al. (2009) found that combining CSF p-tau with
The probability of a postmortem diagnosis of AD for an ini- A42 /A38 resulted in a sensitivity of 94% for detection of AD
tial normal cognitive assessment and negative FDG PET ndings and 85% specicity for excluding non-AD dementias.
was 17%. In a clinical study by de Souza et al. (2011), the p-tau/A42
In a study by Foster et al. (2007), involving patients with ratio was the best biomarker for distinguishing AD from behav-
pathologically conrmed AD or FTLD, adding FDG PET to clin- ioral variant FTLD and SD, with a sensitivity of 91.7 and 98.3%,
ical information increased the accuracy of AD diagnosis from 86 respectively, and a specicity of 92.6 and 84.2%, respectively.
to 97%. Le Bastard et al. (2010) investigated the utility of CSF in cases
that had clinically ambiguous diagnoses, using autopsy-conrmed
PiB PET dementia diagnosis as gold standard. AD and non-AD patients
N -methyl-11C-2-(4-methylaminophenyl)-6-hydroxybenzothiazole, showed no signicant differences in CSF A42 and t-tau con-
also known as 11C-6-OH-BTA-1 or 11C-PiB, is an amyloid- centrations, whereas p-tau (specically p-tau-181) concentrations
binding PET tracer. were signicantly higher in AD compared to non-AD patients. The
The topological pattern of PiB binding in preclinical and clini- biomarker-based diagnostic model correctly classied 82% of the
cal AD patients comprises prefrontal cortex (PFC), medial parietal patients.
cortex, lateral temporal cortex, striatum and posterior cingulate Interestingly, in a study by Schoonenboom et al. (2012), CSF
cortex (PCC), with PFC being the region with earliest and most AD biomarker prole was seen in 47% of DLB cases, 38% of CBD
pronounced uptake (Prvulovic and Hampel, 2011). individuals, and in almost 30% of FTLD and vascular dementia
The diagnostic sensitivity of PiB to accurately classify AD patients. Individuals with psychiatric diseases and with subjective
patients and control subjects is reported to average approximately memory complaints had normal CSF biomarkers in 91 and 88%
90% (Prvulovic and Hampel, 2011). of cases, respectively.
Devanand et al. (2010) evaluated 11C-PiB regional binding
potential (BPND, cerebellar reference) in individuals with clinical CANDIDATE BLOOD-BASED BIOMARKERS
diagnosis of mild AD, MCI and controls. Using a precuneus BPND There is evidence of peripheral oxidative damage correlating with
cut-point of 0.4087 (values above this considered abnormal), in the occurrence of AD (Di Domenico et al., 2011). Thus, peripheral
the differentiation of AD from controls, sensitivity was 0.944 and oxidative biomarkers might be useful for early diagnosis and prog-
specicity 0.944. In distinguishing MCI from controls, sensitivity nosis. Complement factor H, alpha-2-macroglobulin, and clus-
was 0.273 and specicity 0.944. terin have all been consistently associated with Alzheimers type
A puzzling fact is that 1030% of asymptomatic healthy elderly pathology (Ballard et al., 2011a). Ijsselstijn et al. (2011) identied
subjects have increased PiB uptake, a nding that is consistent with a signicant increase in concentration of pregnancy zone protein
several autopsy studies which found AD-typical neuropathological (PZP) in pre-symptomatic AD, when compared with controls. At

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 10


Alves et al. Alzheimers disease: a review

present, the use of plasma based A cannot be recommended as another group of treatments a more or less systematic effect can
diagnostic biomarker for MCI and AD (Prvulovic and Hampel, also be expected, so that it is possible to advise on many aspects
2011). of the management of this form of dementia with some degree
of certainty (Hort et al., 2010; Ballard et al., 2011a; Massoud and
ROLE OF BIOMARKERS IN THE CLINICAL SETTING Lger, 2011). Decisions at the level of a single patient, especially in
It is currently difcult to understand the relative importance of moderate and advanced disease, often have to be, though, based in
different biomarkers when used together, and to interpret results expert opinions, ones previous experience or extrapolations from
when biomarker data conict with one another (Albert et al., other diseases.
2011). The currently available treatments for AD are symptomatic.
Much work is still needed to determine the sensitivity, speci- They are able to, at least transiently, ameliorate some aspects of
city, and predictive value of biomarkers for a diagnosis of AD cognition and function and reduce some neuropsychiatric symp-
in clinical samples. On the other hand, AD biomarkers allow toms, making patients and their entourage suffer less with the
the detection of AD pathology in living individuals who have no disease. If patients under treatment see their therapies removed
clinically discernible cognitive impairment. Such individuals are after a certain time of continuous administration they will be
considered to have preclinical AD, with the assumption that all essentially indistinguishable from patients never treated, meaning
eventually will develop symptomatic AD if they live long enough that this treatments are not intervening in disease progression.
(Mayeux et al., 2011). However, it is known that some older indi- The measures currently advocated to manage this form of
viduals with the pathophysiological process of AD may not become dementia involve adapting the patients environment and treating
symptomatic during their lifetime (Sperling et al., 2011). Use of the patient himself.
biomarkers in the clinical setting is thus currently unwarranted The interaction of a demented patient with the environment
because many individuals who satisfy the proposed research cri- around him is dysfunctional. Some measures can reduce the con-
teria may not develop the clinical features of AD in their lifetime sequences of this (Hort et al., 2010). The continuous education
(Sperling et al., 2011). of the caregivers, perhaps starting with setting real expectations as
Nevertheless, biomarkers can be used as optional tools when to what to expect in terms of long-term evolution of symptoms
considered appropriate by the clinician (McKhann et al., 2011). and treatment effect, is of primordial importance. The ability of
In our view, biomarkers should be used in MCI and demented patients to correctly use money, medications, transports and home
patients whose characteristics pose doubt as to etiology. The appliances should be assessed and continuous adaptation of the
choice of biomarkers depends on local availability and cost- facilities at home and other pertinent environments should be
effectiveness issues, must take into consideration the time depen- planned. Ability to drive should also be assessed according to global
dence of biomarker changes during disease progression and the (Iverson et al., 2010) and country-specic guidelines. Information
main alternative differential diagnosis in the case in question. about social security, legal and other related matters should be sys-
PREDICTORS OF RATE OF DISEASE PROGRESSION tematized. The caregivers should be advised of the possibility of
Factors that seem to increase the rate of disease progression (in their own exhaustion and strategies to avoid it should be foreseen.
terms of cognitive and/or functional status) include: higher edu- Objectives of intervention in the patient himself can be concep-
cation (Bruandet et al., 2008; Musicco et al., 2009, 2010; Roselli tually divided in two: (transiently) revert some cognitive decits
et al., 2009); younger onset (Musicco et al., 2009; Tschanz et al., and ameliorate functional capacity; and revert disturbing neu-
2011); increased baseline severity (Ito et al., 2011); psychosis (Stern ropsychiatric symptoms or behaviors. These treatments may be
et al., 1994; Lopez et al., 1997); extrapyramidal signs (Mortimer pharmacological or non-pharmacological.
et al., 1992; Stern et al., 1994); lower CSF A 42 levels, higher tau The mainstay of treatment consists of a small group of drugs
or p-tau-181 levels, lower p-tau-181/tau ratio and higher tau/A that showed consistent, albeit small and variable, benets in well-
42 ratios (Kester et al., 2009; Snider et al., 2009). designed clinical trials. They are the only currently approved treat-
ments for AD by the authorities of most countries and include the
TREATMENT cholinesterase inhibitors (donepezil, rivastigmine, galantamine)
The particular assembly of symptoms that each patient manifests and the NMDA receptor antagonist memantine (for a review of
as a result of AD changes along the course of the disease and is the evidence see Herrmann et al., 2011). As stated above for the
also different between patients. Added to the dynamic inuence treatment of AD in general, they show only a symptomatic effect,
of co-morbidities and co-medications, this fact may, at least par- although a neuroprotective potential has also been proposed.
tially, explain why the effect of many treatments may vary both Cholinesterase inhibitors are approved for mild to moderate
intra and inter individually. Clinical trials, unable to control all disease (usually MMSE between 16 and 26), where they proved
variables and limited by the complex task of detecting changes in to have effect in cognition, global outcome and function when
cognition or behavior, often reect these heterogeneities by show- compared to placebo (Birks and Harvey, 2006; Loy and Schneider,
ing a globally small, non-signicant or conicting effect for the 2006; Birks et al., 2009). Weaker evidence shows some effect in
treatment they are evaluating (Gauthier et al., 2010). Consider- severe AD (Herrmann et al., 2011) and the FDA has also approved
ing that many drugs (and other treatments) have signicant side donepezil for moderate to severe disease. These drugs show as
effects and costs, producing guidelines for the treatment of this well benet in some neuropsychiatric symptoms, particularly apa-
disease is not always clear-cut. Despite all this, good level of evi- thy, and somewhat less in psychosis. In clinical practice one can
dence exists for some effects of a small group of drugs, and for generally expect to observe slight amelioration in cognition and

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 11


Alves et al. Alzheimers disease: a review

stabilization in function, parameters which are presumed to return disturbance (Gauthier et al., 2010). For severe depression, most
to baseline or start degrading again after 6 to 12 months of treat- clinicians use antidepressive drugs, whose efcacy has been shown
ment. Differences in drug metabolism may justify switching from in many trials, although not in all. Sertraline (100 mg/day) is the
one cholinesterase inhibitor agent to another in cases of intoler- most documented drug, but other SSRIs and other classes can be
ance or lack of effect (Massoud et al., 2011). The most common tried, as long as side effects are considered (Gauthier et al., 2010;
side effects are gastrointestinal dysfunction, anorexia and sleep Ballard et al., 2011a).
disturbances. They can usually be avoided by titrating up the Neuroleptic drugs are commonly used to treat aggression, agi-
dose. The most concerning side effects of these medications are tation and psychosis. Efcacy has been demonstrated for risperi-
bradycardia and syncope, reasons for which it is advisable to eval- done (especially 2 mg/day) when prescribed for aggression, but
uate preexisting bradycardia or cardiac conduction blocks with for agitation and psychosis, and for use of other neuroleptics,
an electrocardiogram and monitor blood pressure. Rivastigmine results are weak or conicting. Benets, which are often mod-
administered in a transdermal patch has fewer side effects than its erate, must be weighed against potentially serious adverse events
oral form, maintaining the same benets (Winblad et al., 2007). like sedation, parkinsonism, chest infections, ankle edema, and
Memantine is approved for moderate to severe AD, where it an increased risk of stroke and death. As already stated, mini-
proved to have benecial effects in cognitive performance, func- mal effective doses should be used and long-term prescription
tion and global measures (McShane et al., 2006). Memantine avoided. Anticonvulsants, like carbamazepine (useful for agita-
may also be useful in the prevention and treatment of agitation, tion), and benzodiazepines may be helpful in selected cases. For
aggression, irritability and psychosis. As with cholinergic drugs, the rst one, potential drug interactions should not be forgotten,
one should expect stabilization of cognition and function for and, for the latter, very short term use should be foreseen. Simple
6 months after onset of treatment. Memantine is usually well tol- non-pharmacological treatments, such as increasing physical and
erated. Dose-limiting side effects are rare and consist of dizziness, social activity, aroma therapy (lavender and melissa), therapy with
headache, somnolence, and confusion. animals, music therapy and simulated presence therapy (audio or
There also seem to be additive benets of combining a video tapes with familiars), can be effective alternatives (Ballard
cholinesterase inhibitor and memantine (Tariot et al., 2004; Atri et al., 2008, 2011a; Gauthier et al., 2010).
et al., 2008; Lopez et al., 2009) albeit not all studies have supported The treatment of AD also involves management of co-
this (Porsteinsson et al., 2008). morbidities. They often cause sudden aggravation of cognitive
The short duration of most trials with these four drugs pre- deterioration or appearance of neuropsychiatric symptoms. In
cludes us from knowing the time for how long a difference from these situations, so called medical causes, such as metabolic and
placebo is maintained. The majority of experts advice caution in infectious diseases, as well as cerebrovascular pathologies, must be
stopping these drugs, although some reasons for discontinuation actively sought and treated.
may be advocated (Massoud et al., 2011). Other symptoms which must be dealt with (mostly in advanced
Concerning non-pharmacological approaches to help main- disease stages) include parkinsonism, gait instability, myoclonus,
tain or enhance the cognitive, functional and global status of these seizures, contractures, pressure ulcers, pain, and undernutrition.
patients, current evidence suggests that cognitive training and cog- Concerning emerging treatment drugs, they are expected to
nitive stimulation offer modest, albeit signicant, benets. The show less inter-individual variability and a more marked effect.
results are mainly limited to the cognitive domains on which the For that purpose, they must act directly in the mechanisms of
intervention is focused and, as these interventions are laborious, disease and be administered earlier in the course of AD, before sig-
cost-effectiveness studies are needed (Ballard et al., 2011b). nicant neurodegeneration has occurred. Hopefully, biomarkers
Another major topic in the treatment of AD is the approach will allow diagnosis before a dementia syndrome is installed and
to certain frequent and disturbing neuropsychiatric symptoms permit testing drugs at a stage when they could be (more) effective.
like apathy, depression, anxiety, psychosis, agitation, irritabil- Several lines of drug investigation are being pursued, some
ity, aggression and sleep disturbances. There are no authority targeting only symptomatic treatment, but the majority aiming
approved treatments for these situations, which is a consequence to show a disease-modifying effect (for reviews see Potter, 2010;
of lack of data, insufcient or conicting results and concerns Herrmann et al., 2011; Salomone et al., 2012).
with possible side effects. In general terms, non-pharmacological New cholinergic agents in investigation include direct and
interventions are advocated rst (sleep hygiene measures, for allosteric muscarinic acetylcholine receptor agonists and also
instance). When symptoms are very severe and menacing, phar- agonists of certain subtypes of nicotinic receptors.
macological treatment may be started immediately, accompanied A group of drugs aiming to reduce A production include:
by non-pharmacological measures. Another general rule is that rosiglitazone, which, among other modes of action, could inhibit
drug treatments employed in this setting should be administered secretase, was ineffective in a phase 3 trial, despite suggestion
in the minimal efcacious dose and envisaged as transitory. Regis- of efcacy in earlier data (Gold et al., 2010); semagacestat, a
tering all the events prior to these manifestations helps to identify secretase inhibitor, was tested in two phase 3 trials that had to
possible environmental triggers. be prematurely stopped due to serious systemic adverse events
For depression, when symptoms are mild and transient, non- (Schor, 2011); several NSAID, including tarenurbil, show a weak
pharmacological approaches should be tried, like structured activ- modulating or inhibiting effect on secretase, but the tested ones
ities, such as day programs and daily exercise (Ballard et al., 2008). were not efcacious in AD. Calcium channel blocker nilvadipine
Bright light therapy can aid sleep and reduce mood and behavioral could also reduce the production and augment the clearance of

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 12


Alves et al. Alzheimers disease: a review

A and is being clinically tested. Investigation of more potent and active clinical investigation. The most studied antibodies are bap-
specic inhibitors of secretase and efforts to produce secretase ineuzumab, which is under various phase 3 trials with different
stimulating drugs are being undertaken. In the line of preventing doses after a higher dose produced cerebral vasogenic edema in
A aggregation, tramiprosate, which, by binding to A monomers, almost 10% of the patients (Panza et al., 2011), and solanezumab,
prevents formation of oligomers, did not show global clinical ben- also in phase 3 trials. Other strategies are being studied, including
et in a phase 3 trial, although more studies are needed to clarify the use of intravenous immunoglobulins (IVIG).
its effects in some cognitive areas (Saumier et al., 2009). Other Lithium and valproate, besides other modes of action, could
drugs that could interfere in this stage of the A cascade are the reduce hyperphosphorylation of tau, but various clinical studies
zinc and copper chelators, curcumin (also proposed to have sec- have shown conicting results. Other compounds judged to act in
retase inhibiting properties), epigallocatechin-3-gallate (EGCG) tau phosphorylation and aggregation (including methylene blue)
and scyllo-inositol, a compound that promotes dissociation of A are under investigation.
oligomers. Other therapeutic strategies in development include the use
Immunotherapy in AD is essentially being tried as a way of of nerve growth factor, etanercept and phosphodiesterase-5
clearing A deposits through the use of antibodies against this inhibitors; interventions at the mitochondrial level (for instance
protein. Active immunization in the form of vaccines has pro- with latrepirdine and EGCG); inhibition of the receptor for
duced severe adverse effects (meningoencephalitis), but different advanced glycation end products (RAGE); and the use of deep
techniques, inducing immunization against only a part of the A brain stimulation (DBS). The roles of caffeine (or of whole cof-
molecule and not all of it, are being thought of. Passive immu- fee) and of physical activity in the treatment of AD also deserve
nization with monoclonal antibodies is at present under very clarication.

REFERENCES A., Martinez-Lage, M., Stijnen, T., Antnez, C., Boada, M., Lpez-Arrieta, E. (2011a). Alzheimers disease.
Albert, M. S., DeKosky, S. T., Dickson, Hofman, A., and for the EURODEM J., Moreno-Rey, C., Hernndez, I., Lancet 377, 10191031.
D., Dubois, B., Feldman, H. H., Fox, Incidence Research Group. (1999). Marn, J., Gayn, J., Alzheimers Ballard, C., Khan, Z., Clack, H., and
N. C., Gamst, A., Holtzman, D. M., Gender differences in the incidence Disease Neuroimaging Initiative, Corbett, A. (2011b). Nonpharma-
Jagust, W. J., Petersen, R. C., Snyder, of AD and vascular dementia: the Gonzlez-Prez, A., Real, L. M., cological treatment of Alzheimer
P. J., Carrillo, M. C., Thies, B., and EURODEM Studies. Neurology 53, Alegret, M., Trraga, L., Ramrez- disease. Can. J. Psychiatry 56,
Phelps, C. H. (2011). The diagnosis 19921997. Lorca, R., and Ruiz, A. (2011b). 589595.
of mild cognitive impairment due Andersen, O. M., Reiche, J., Schmidt, V., Genetic association of complement Bao, F., Wicklund, L., Lacor, P. N.,
to Alzheimers disease: recom- Gotthardt, M., Spoelgen, R., Behlke, receptor 1 polymorphism rs3818361 Klein, W. L., Nordberg, A., and
mendations from the National J., von Arnim, C. A., Breiderhoff, in Alzheimers disease. Alzheimers Marutle, A. (2012). Different -
Institute on Aging-Alzheimers T., Jansen, P., Wu, X., Bales, K. R., Dement. 7, e124e129. amyloid oligomer assemblies in
Association workgroups on diag- Cappai, R., Masters, C. L., Glie- Apostolova, L. G., and Cummings, J. L. Alzheimer brains correlate with
nostic guidelines for Alzheimers mann, J., Mufson, E. J., Hyman, (2008). Neuropsychiatric manifesta- age of disease onset and impaired
disease. Alzheimers Dement. 7, B. T., Paul, S. M., Nykjaer, A., tions in mild cognitive impairment: cholinergic activity. Neurobiol. Aging
270279. and Willnow, T. E. (2005). Neu- a systematic review of the literature. 33, 825.e1825.e13.
Alladi, S., Xuereb, J., Bak, T., Nestor, P., ronal sorting protein-related recep- Dement. Geriatr. Cogn. Disord. 25, Barber, R., Ballard, C., McKeith, I.
Knibb, J., Patterson, K., and Hodges, tor sorLA/LR11 regulates process- 115126. G., Gholkar, A., and OBrien, J.
J. R. (2007). Focal cortical presen- ing of the amyloid precursor pro- Arendt, T. (2009). Synaptic degener- T. (2000). MRI volumetric study
tations of Alzheimers disease. Brain tein. Proc. Natl. Acad. Sci. U.S.A. 102, ation in Alzheimers disease. Acta of dementia with Lewy bodies: a
130, 26362645. 1346113466. Neuropathol. 118, 167179. comparison with AD and vascular
Alzheimer, A., Stelzmann, R. A., Schnit- Antnez, C., Boada, M., Gonzlez- Arrighi, H. M., Neumann, P. J., Lieber- dementia. Neurology 54, 13041309.
zlein, H. N., and Murtagh, F. R. Prez, A., Gayn, J., Ramrez-Lorca, burg, I. M., and Townsend, R. J. Barkhof, F., Polvikoski, T. M., van
(1995). An English translation of R., Marn, J., Hernndez, I., Moreno- (2010). Lethality of Alzheimer dis- Straaten, E. C. W., Kalaria, R. N.,
Alzheimers 1907 paper, Uber eine Rey, C., Morn, F. J., Lpez- ease and its impact on nursing home Sulkava, R., Aronen, H. J., Niin-
eigenartige Erkankung der Hirn- Arrieta, J., Maulen, A., Rosende- placement. Alzheimer Dis. Assoc. Dis- ist, L., Rastas, S., Oinas, M., Schel-
rinde. Clin. Anat. 8, 429431. Roca, M., Noguera-Perea, F., Legaz- ord. 24, 9095. tens, P., and Erkinjuntti, T. (2007).
Amadoro, G., Corset, V., Atlante, A., Garca, A., Vivancos-Moreau, L., Atri, A., Shaughnessy, L. W., Locascio, J. The signicance of medial temporal
Florenzano, F., Capsoni, S., Bussani, Velasco, J., Carrasco, J. M., Alegret, J., and Growdon, J. H. (2008). Long- lobe atrophy: a post-mortem MRI
R., Mercanti, D., and Calissano, P. M., Antequera-Torres, M., Man- term course and effectiveness of study in the very old. Neurology 69,
(2012). Interaction between NH(2)- zanares, S., Romo,A., Blanca, I., Ruiz, combination therapy in Alzheimer 15211527.
tau fragment and A in Alzheimers S., Espinosa, A., Castao, S., Gar- disease. Alzheimer Dis. Assoc. Disord. Barnes, L. L., Wilson, R. S., Bienias, J.
disease mitochondria contributes to ca, B., Martnez-Herrada, B., Vinyes, 22, 209221. L., Schneider, J. A., Evans, D. A., and
the synaptic deterioration. Neuro- G., Lafuente, A., Becker, J. T., Galn, Avramopoulos, D. (2009). Genetics of Bennett, D. A. (2005). Sex differences
biol. Aging 33, 833.e1833.e25. J. J., Serrano-Ros, M., Alzheimers Alzheimers disease: recent advances. in the clinical manifestations of
American Psychiatric Association. Disease Neuroimaging Initiative, Genome Med. 1, 34. Alzheimer disease pathology. Arch.
(2000). Diagnostic and Statistical Vzquez, E., Trraga, L., Sez, M. Ballard, C., Day, S., Sharp, S., Wing, Gen. Psychiatry 62, 685691.
Manual of Mental Disorders, 4th E., Lpez, O. L., Real, L. M., and G., and Sorensen, S. (2008). Neu- Benson, D. F., Davis, R. J., and Snyder, D.
Edn. Washington, DC: American Ruiz, A. (2011a). The membrane- ropsychiatric symptoms in demen- B. (1988). Posterior cortical atrophy.
Psychiatric Association. spanning 4-domains, subfamily A tia: importance and treatment con- Arch. Neurol. 45, 789793.
Andersen, K., Launer, J., Dewey, E., (MS4A) gene cluster contains a siderations. Int. Rev. Psychiatry 20, Bertram, L., and Tanzi, R. E. (2005). The
Letenneur, L., Ott, A., Copeland, common variant associated with 396404. genetic epidemiology of neurode-
M., Dartigues, F., Kragh-Sorensen, Alzheimers disease. Genome Med. Ballard, C., Gauthier, S., Corbett, A., generative disease. J. Clin. Invest.
P., Baldereschi, M., Brayne, C., Lobo, 3, 33. Brayne, C., Aarsland, D., and Jones, 115, 14491457.

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 13


Alves et al. Alzheimers disease: a review

Biessels, G., Staekenborg, S., Brunner, E., Callahan, L. M., Vaules, W. A., and Cataldo, J., Prochaska, J., and Glantz, Paz, R., Dubois, B., and Sarazin,
Brayne, C., and Scheltens, P. (2006). Coleman, P. D. (1999). Quantitative S. (2010). Cigarette smoking is a M. (2011). Cerebrospinal uid bio-
Risk of dementia in diabetes melli- decrease in synaptophysin message risk factor for Alzheimers disease: markers in the differential diagno-
tus: a systematic review. Lancet Neu- expression and increase in cathepsin an analysis controlling for tobacco sis of Alzheimers disease from other
rol. 5, 6474. D message expression in Alzheimer industry afliation. J. Alzheimer Dis. cortical dementias. J. Neurol. Neuro-
Birks, J., Grimley Evans, J., Iakovidou,V., disease neurons containing neu- 19, 465480. surg. Psychiatr. 82, 240246.
Tsolaki, M., and Holt, F. E. (2009). robrillary tangles. J. Neuropathol. Coleman, P., and Yao, P. (2003). Synap- Devanand, D. P., Mikhno, A., Pelton,
Rivastigmine for Alzheimers dis- Exp. Neurol. 58, 275287. tic slaughter in Alzheimers disease. G. H., Cuasay, K., Pradhaban, G.,
ease. Cochrane Database Syst. Rev. 2, Callahan, L. M., Vaules, W. A., and Neurobiol. Aging 24, 10231027. Dileep Kumar, J. S., Upton, N., Lai,
CD001191. Coleman, P. D. (2002). Progressive Coon, K. D., Myers, A. J., Craig, D. W., R., Gunn, R. N., Libri, V., Liu, X., van
Birks, J., and Harvey, R. J. (2006). reduction of synaptophysin message Webster, J. A., Pearson, J. V., Lince, Heertum, R., Mann, J. J., and Parsey,
Donepezil for dementia due to in single neurons in Alzheimer dis- D. H., Zismann, V. L., Beach, T. G., R. V. (2010). Pittsburgh compound
Alzheimers disease. Cochrane Data- ease. J. Neuropathol. Exp. Neurol. 61, Leung, D., Bryden, L., Halperin, R. F., B (11C-PIB) and uorodeoxyglu-
base Syst. Rev. 1, CD001190. 384395. Marlowe, L., Kaleem, M., Walker, D. cose (18 F-FDG) PET in patients
Blessed, G., Tomlinson, B. E., and Roth, Campion, D., Dumanchin, C., Han- G., Ravid, R., Heward, C. B., Rogers, with Alzheimer disease, mild cogni-
M. (1968). The association between nequin, D., Dubois, B., Belliard, J., Papassotiropoulos, A., Reiman, tive impairment, and healthy con-
quantitative measures of dementia S., Puel, M., Thomas-Anterion, C., E. M., Hardy, J., and Stephan, D. trols. J. Geriatr. Psychiatry Neurol.
and of senile change in the cerebral Michon, A., Martin, C., Charbon- A. (2007). A high-density whole- 23, 185198.
grey matter of elderly subjects. Br. J. nier, F., Raux, G., Camuzat, A., genome association study reveals Di Domenico, F., Coccia, R., Buttereld,
Psychiatry 114, 797811. Penet, C., Mesnage, V., Martinez, that APOE is the major suscepti- D. A., and Perluigi, M. (2011). Cir-
Bohnen, N. I., Djang, D. S., Herholz, K., M., Clerget-Darpoux, F., Brice, A., bility gene for sporadic late-onset culating biomarkers of protein oxi-
Anzai, Y., and Minoshima, S. (2012). and Frebourg, T. (1999). Early-onset Alzheimers disease. J. Clin. Psychi- dation for Alzheimer disease: expec-
Effectiveness and safety of 18F-FDG autosomal dominant Alzheimer dis- atry 68, 613618. tations within limits. Biochim. Bio-
PET in the evaluation of dementia: ease: prevalence, genetic heterogene- Corder, E. H., Saunders, A. M., phys. Acta 1814, 17851795.
a review of the recent literature. J. ity, and mutation spectrum. Am. J. Strittmatter, W. J., Schmechel, D. E., Di Fede, G., Catania, M., Morbin, M.,
Nucl. Med. 53, 5971. Hum. Genet. 65, 664670. Gaskell, P. C., Small, G. W., Roses, Rossi, G., Suardi, S., Mazzoleni, G.,
Bots, L., van Kooten, F., Haverkate, F., Carlesimo, G. A., Perri, R., and Calt- A. D., Haines, J. L., and Pericak- Merlin, M., Giovagnoli, A. R., Prioni,
Meijer, P., Koudstaal, J., Grobbee, D., agirone, C. (2011). Category cued Vance, M. A. (1993). Gene dose of S., Erbetta, A., Falcone, C., Gobbi,
and Kluft, C. (1998). Coagulation recall following controlled encoding apolipoprotein E type 4 allele and M., Colombo, L., Bastone, A., Beeg,
and brinolysis markers and risk of as a neuropsychological tool in the the risk of Alzheimers disease in late M., Manzoni, C., Francescucci, B.,
dementia: the Dutch vascular factors diagnosis of Alzheimers disease: a onset families. Science 261, 921923. Spagnoli, A., Cant, L., Del Favero,
in dementia study. Haemostasis 28, review of the evidence. Neuropsy- Corneveaux, J. J., Myers, A. J., Allen, E., Levy, E., Salmona, M., and Tagli-
216222. chol. Rev. 21, 5465. A. N., Pruzin, J. J., Ramirez, M., avini, F. (2009). A recessive mutation
Braak, H., and Braak, E. (1995). Stag- Carrasquillo, M. M., Belbin, O., Hunter, Engel, A., Nalls, M. A., Chen, K., Lee, in the APP gene with dominant-
ing of Alzheimers disease-related T. A., Ma, L., Bisceglio, G. D., Zou, F., W., Chewning, K., Villa, S. E., Mee- negative effect on amyloidogenesis.
neurobrillary changes. Neurobiol. Crook, J. E., Pankratz, V. S., Dick- choovet, H. B., Gerber, J. D., Frost, D., Science 323, 14731477.
Aging 16, 271278. son, D. W., Graff-Radford, N. R., Benson, H. L., OReilly, S., Chibnik, Dougall, N. J., Bruggink, S., and
Braak, H., and Del Tredici, K. (2011). Petersen, R. C., Morgan, K., and L. B., Shulman, J. M., Singleton, A. Ebmeier, K. P. (2004). Systematic
Alzheimers pathogenesis: is there Younkin, S. G. (2010). Replication B., Craig, D. W., Van Keuren-Jensen, review of the diagnostic accuracy of
neuron to-neuron propagation? Acta of CLU, CR1, and PICALM associ- K. R., Dunckley, T., Bennett, D. A., 99mTc-HMPAO-SPECT in demen-
Neuropathol. 121, 589595. ations with Alzheimer disease. Arch. De Jager, P. L., Heward, C., Hardy, J., tia. Am. J. Geriatr. Psychiatry 12,
Brookmeyer, R., Corrada, M. M., Cur- Neurol. 67, 961964. Reiman, E. M., and Huentelman, M. 554570.
riero, F. C., and Kawas, C. (2002). Carrasquillo, M. M., Belbin, O., Hunter, J. (2010). Association of CR1, CLU Dubois, B., Feldman, H. H., Jacova,
Survival following a diagnosis of T. A., Ma, L., Bisceglio, G. D., Zou, F., and PICALM with Alzheimers dis- C., Cummings, J. L., Dekosky, S. T.,
Alzheimer disease. Arch. Neurol. 59, Crook, J. E., Pankratz, V. S., Sando, ease in a cohort of clinically charac- Barberger-Gateau, P., Delacourte, A.,
17641767. S. B., Aasly, J. O., Barcikowska, M., terized and neuropathologically ver- Frisoni, G., Fox, N. C., Galasko, D.,
Bruandet, A., Richard, F., Bombois, S., Wszolek, Z. K., Dickson, D. W., ied individuals. Hum. Mol. Genet. Gauthier, S., Hampel, H., Jicha, G.
Maurage, C. A., Masse, I., Amouyel, Graff-Radford, N. R., Petersen, R. 19, 32953301. A., Meguro, K., OBrien, J., Pasquier,
P., and Pasquier, F. (2008). Cognitive C., Morgan, K., and Younkin, S. Cruchaga, C., Nowotny, P., Kauwe, J. S., F., Robert, P., Rossor, M., Salloway,
decline and survival in Alzheimers G. (2011a). Replication of BIN1 Ridge, P. G., Mayo, K., Bertelsen, S., S., Sarazin, M., de Souza, L. C.,
disease according to education level. association with Alzheimers dis- Hinrichs, A., Fagan, A. M., Holtz- Stern, Y., Visser, P. J., and Scheltens,
Dement. Geriatr. Cogn. Disord. 25, ease and evaluation of genetic man, D. M., Morris, J. C., Goate, A. P. (2010). Revising the denition of
7480. interactions. J. Alzheimers Dis. 24, M., and Alzheimers Disease Neu- Alzheimers disease: a new lexicon.
Burton, E. J., Barber, R., Mukaetova- 751758. roimaging Initiative. (2011). Associ- Lancet Neurol. 9, 11181127.
Ladinska, E. B., Robson, J., Perry, Carrasquillo, M. M., Belbin, O., Hunter, ation and expression analyses with Dubois, B., Feldman, H. H., Jacova,
R. H., Jaros, E., Kalaria, R. N., and T. A., Ma, L., Bisceglio, G. D., Zou, F., single-nucleotide polymorphisms in C., Dekosky, S. T., Barberger-Gateau,
OBrien, J. T. (2009). Medial tem- Crook, J. E., Pankratz, V. S., Sando, TOMM40 in Alzheimer disease. P., Cummings, J., Delacourte, A.,
poral lobe atrophy on MRI dif- S. B., Aasly, J. O., Barcikowska, M., Arch. Neurol. 68, 10131019. Galasko, D., Gauthier, S., Jicha, G.,
ferentiates Alzheimers disease from Wszolek, Z. K., Dickson, D. W., de la Torre, J. C. (2004). Is Alzheimers Meguro, K., Obrien, J., Pasquier, F.,
dementia with Lewy bodies and Graff-Radford, N. R., Petersen, R. disease a neurodegenerative or a vas- Robert, P., Rossor, M., Salloway, S.,
vascular cognitive impairment: a C., Passmore, P., Morgan, K., and cular disorder? Data, dogma, and Stern, Y., Visser, P. J., and Schel-
prospective study with pathologi- for the Alzheimers Research UK dialectics. Lancet Neurol. 3, 184190. tens, P. (2007). Research criteria
cal verication of diagnosis. Brain (ARUK) consortium, and Younkin, de la Torre, J. C. (2010). Vascular risk for the diagnosis of Alzheimers
132(Pt 1), 195203. S. G. (2011b). Replication of EPHA1 factor detection and control may disease: revising the NINCDS-
Cai, Z., Zhao, B., and Ratka, A. (2011). and CD33 associations with late- prevent Alzheimers disease. Ageing ADRDA criteria. Lancet Neurol. 6,
Oxidative stress and -amyloid pro- onset Alzheimers disease: a multi- Res. Rev. 9, 218225. 734746.
tein in Alzheimers disease. Neuro- centre case-control study. Mol. Neu- de Souza, L. C., Lamari, F., Belliard, Duron, E., and Hanon, O. (2010). Atrial
molecular Med. 13, 223250. rodegener. 6, 54. S., Jardel, C., Houillier, C., De brillation and cognitive function.

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 14


Alves et al. Alzheimers disease: a review

Psychol. Neuropsychiatr. Vieil. 8, socially integrated lifestyle in late Goate, A., Chartier-Harlin, M. C., Mul- Kerkman, D., Ishiguro, K., Ohno, H.,
209214. life might protect against dementia. lan, M., Brown, J., Crawford, F., Vanmechelen, E., Vanderstichele, H.,
Engelhart, M., Geerlings, M., Meijer, Lancet Neurol. 3, 343353. Fidani, L., Giuffra, L., Haynes, A., McCulloch, C., Moller, H. J., Davies,
J., Kiliaan, A., Ruitenberg, A., van Galton, C. J., Gomez-Anson, B., Antoun, Irving, N., James, L., Mant, R., New- P., and Blennow, K. (2004). Mea-
Swieten, J., Stijnen, T., Hofman, N., Scheltens, P., Patterson, K., ton, P., Rooke, K., Roques, P., Tal- surement of phosphorylated tau epi-
A., Witteman, J., and Breteler, M. Graves, M., Sahakian, B. J., and bot, C., Pericak-Vance, M., Roses, A., topes in the differential diagnosis
(2004). Inammatory proteins in Hodges, J. R. (2001). Temporal Williamson, R., Rossor, M., Owen, of Alzheimer disease: a comparative
plasma and the risk of dementia. The lobe rating scale: application to M., and Hardy, J. (1991). Segrega- cerebrospinal uid study. Arch. Gen.
Rotterdam Study. Arch. Neurol. 61, Alzheimers disease and frontotem- tion of a missense mutation in the Psychiatry 61, 95102.
668672. poral dementia. J. Neurol. Neurosurg. amyloid precursor protein gene with Hampel, H., Brger, K., Teipel, S. J.,
Ewers, M., Buerger, K., Teipel, S. J., Psychiatr. 70, 165173. familial Alzheimers disease. Nature Bokde, A. L., Zetterberg, H., and
Scheltens, P., Schrder, J., Zinkowski, Galton, C. J., Patterson, K., Xuereb, 349, 704706. Blennow, K. (2008). Core candi-
R. P., Bouwman, F. H., Schnknecht, J. H., and Hodges, J. R. (2000). Gold, M., Alderton, C., Zvartau-Hind, date neurochemical and imaging
P., Schoonenboom, N. S., Andreasen, Atypical and typical presentations of M., Egginton, S., Saunders, A. biomarkers of Alzheimers disease.
N., Wallin, A., DeBernardis, J. Alzheimers disease: a clinical, neu- M., Irizarry, M., Craft, S., Lan- Alzheimers Dement. 4, 3848.
F., Kerkman, D. J., Heindl, B., ropsychological, neuroimaging and dreth, G., Linnamagi, U., and Hardy, J. (1997). Amyloid, the pre-
Blennow, K., and Hampel, H. (2007). pathological study of 13 cases. Brain Sawchak, S. (2010). Rosiglitazone senilins and Alzheimers disease.
Multicenter assessment of CSF- 123, 484498. monotherapy in mild-to-moderate Trends Neurosci. 20, 154159.
phosphorylated tau for the predic- Galvin, J. E., Roe, C. M., Powlishta, K. K., Alzheimers disease: results from a Harold, D., Abraham, R., Hollingworth,
tion of conversion of MCI. Neurol- Coats, M. A., Muich, S. J., Grant, E., randomized, double-blind, placebo- P., Sims, R., Gerrish, A., Hamshere,
ogy 69, 22052212. Miller, J. P., Storandt, M., and Morris, controlled phase III study. Dement. M. L., Pahwa, J. S., Moskvina, V.,
Farrer, L. A., Cupples, L. A., Haines, J. C. (2005). The AD8: a brief infor- Geriatr. Cogn. Disord. 30, 131146. Dowzell, K., Williams, A., Jones, N.,
J. L., Hyman, B., Kukull, W. A., mant interview to detect dementia. Goldman, J. S., Hahn, S. E., Cata- Thomas, C., Stretton, A., Morgan, A.
Mayeux, R., Myers, R. H., Pericak- Neurology 65, 559564. nia, J. W., LaRusse-Eckert, S., But- R., Lovestone, S., Powell, J., Proitsi,
Vance, M. A., Risch, N., and van Gatz, M., Reynolds, C. A., Fratiglioni, son, M. B., Rumbaugh, M., Strecker, P., Lupton, M. K., Brayne, C., Rubin-
Duijn, C. M. (1997). Effects of L., Johansson, B., Mortimer, J. A., M. N., Roberts, J. S., Burke, W., sztein, D. C., Gill, M., Lawlor, B.,
age, sex, and ethnicity on the asso- Berg, S., Fiske, A., and Pedersen, N. Mayeux, R., Bird, T., and Amer- Lynch, A., Morgan, K., Brown, K. S.,
ciation between apolipoprotein E L. (2006). Role of genes and envi- ican College of Medical Genetics Passmore, P. A., Craig, D., McGuin-
genotype and Alzheimer disease. A ronments for explaining Alzheimer and the National Society of Genetic ness, B., Todd, S., Holmes, C., Mann,
meta-analysis. APOE and Alzheimer disease. Arch. Gen. Psychiatry 63, Counselors. (2011). Genetic coun- D., Smith, A. D., Love, S., Kehoe,
Disease Meta Analysis Consortium. 168174. seling and testing for Alzheimer dis- P. G., Hardy, J., Mead, S., Fox, N.,
JAMA 278, 13491356. Gauthier, S., Cummings, J., Ballard, C., ease: joint practice guidelines of Rossor, M., Collinge, J., Maier, W.,
Ferri, C. P., Prince, M., Brayne, C., Bro- Brodaty, H., Grossberg, G., Robert, the American College of Medical Jessen, F., Schrmann, B., van den
daty, H., Fratiglioni, L., Ganguli, M., P., and Lyketsos, C. (2010). Man- Genetics and the National Society of Bussche, H., Heuser, I., Kornhu-
Hall, K., Hasegawa, K., Hendrie, H., agement of behavioral problems in Genetic Counselors. Genet. Med. 13, ber, J., Wiltfang, J., Dichgans, M.,
Huang, Y., Jorm, A., Mathers, C., Alzheimers disease. Int. Psychogeri- 597605. Frlich, L., Hampel, H., Hll, M.,
Menezes, P. R., Rimmer, E., Scazufca, atr. 22, 346372. Gmez-Ramos, P., and Asuncin Rujescu, D., Goate, A. M., Kauwe,
M., and Alzheimers Disease Interna- Genin, E., Hannequin, D., Wallon, Morn, M. (2007). Ultrastructural J. S., Cruchaga, C., Nowotny, P.,
tional. (2005). Global prevalence of D., Sleegers, K., Hiltunen, M., localization of intraneuronal Abeta- Morris, J. C., Mayo, K., Sleegers,
dementia: a Delphi consensus study. Combarros, O., Bullido, M. J., peptide in Alzheimer disease brains. K., Bettens, K., Engelborghs, S., De
Lancet 366, 21122117. Engelborghs, S., De Deyn, P., Berr, J. Alzheimers Dis. 11, 5359. Deyn, P. P., Van Broeckhoven, C.,
Folstein, M. F., Folstein, S. E., and C., Pasquier, F., Dubois, B., Tognoni, Grober, E., and Buschke, H. (1987). Livingston, G., Bass, N. J., Gurl-
McHugh, P. R. (1975). Mini-mental G., Fivet, N., Brouwers, N., Bet- Genuine memory decits in ing, H., McQuillin, A., Gwilliam, R.,
state: a practical method for grad- tens, K., Arosio, B., Coto, E., Del dementia. Dev. Neuropsychol. 3, Deloukas, P., Al-Chalabi, A., Shaw,
ing the cognitive state of patients for Zompo, M., Mateo, I., Epelbaum, 1336. C. E., Tsolaki, M., Singleton, A.
the clinicians. J. Psychiatr. Res. 12, J., Frank-Garcia, A., Helisalmi, S., Grober, E., Buschke, H., Crystal, H., B., Guerreiro, R., Mhleisen, T. W.,
189198. Porcellini, E., Pilotto, A., Forti, P., Bang, S., and Dresner, R. (1988). Nthen, M. M., Moebus, S., Jckel,
Foster, N. L., Heidebrink, J. L., Clark, Ferri, R., Scarpini, E., Siciliano, Screening for dementia by memory K. H., Klopp, N., Wichmann, H.
C. M., Jagust, W. J., Arnold, S. G., Solfrizzi, V., Sorbi, S., Spalletta, testing. Neurology 38, 900903. E., Carrasquillo, M. M., Pankratz, V.
E., Barbas, N. R., DeCarli, C. S., G., Valdivieso, F., Vepslinen, S., Guo, Z., Cupples, A., Kurz, A., Auerbach, S., Younkin, S. G., Holmans, P. A.,
Turner, R. S., Koeppe, R. A., Hig- Alvarez, V., Bosco, P., Mancuso, M., S., Volicer, L., Chui, H., Green, C., ODonovan, M., Owen, M. J., and
don, R., and Minoshima, S. (2007). Panza, F., Nacmias, B., Boss, P., Sadovnick, A., and Duara, R. (2000). Williams, J. (2009). Genome-Wide
FDG-PET improves accuracy in dis- Hanon, O., Piccardi, P., Annoni, Head injury and the risk of AD in Association Study identies variants
tinguishing frontotemporal demen- G., Seripa, D., Galimberti, D., the MIRAGE study. Neurology 54, at CLU and PICALM associated with
tia and Alzheimers disease. Brain Licastro, F., Soininen, H., Dartigues, 13161323. Alzheimers disease. Nat. Genet. 41,
130(Pt 10), 26162635. J. F., Kamboh, M. I., Van Broeck- Gustafson, D. (2006). Adiposity indices 10881093.
Fratiglioni, L., Launer, L. J., Andersen, hoven, C., Lambert, J. C., Amouyel, and dementia. Lancet Neurol. 5, Hebert, L. E., Scherr, P. A., McCann,
K., Breteler, M. M., Copeland, J. R., P., and Campion, D. (2011). 713720. J. J., Beckett, L. A., and Evans, D.
Dartigues, J. F., Lobo, A., Martinez- APOE and Alzheimer disease: a Gtz, J., Schild,A., Hoerndli, F., and Pen- A. (2001). Is the risk of develop-
Lage, J., Soininen, H., and Hofman, major gene with semi-dominant nanen, L. (2004). Amyloid-induced ing Alzheimers disease greater for
A. (2000). Incidence of dementia inheritance. Mol. Psychiatry 16, neurobrillary tangle formation in women than for men? Am. J. Epi-
and major subtypes in Europe: a 903907. Alzheimers disease: insight from demiol. 153, 132136.
collaborative study of population- Glenner, G. G., and Wong, C. W. (1984). transgenic mouse and tissue-culture Helzner, E. P., Scarmeas, N., Cosentino,
based cohorts. Neurologic Diseases Alzheimers disease: initial report of models. Int. J. Dev. Neurosci. 22, S., Tang, M. X., Schupf, N., and Stern,
in the Elderly Research Group. Neu- the purication and characteriza- 453465. Y. (2008). Survival in Alzheimer
rology 54(Suppl. 5), S10S15. tion of a novel cerebrovascular amy- Hampel, H., Buerger, K., Zinkowski, R., disease: a multiethnic, population-
Fratiglioni, L., Paillard-Borg, S., and loid protein. Biochem. Biophys. Res. Teipel, S. J., Goernitz, A., Andreasen, based study of incident cases. Neu-
Winblad, B. (2004). An active and Commun. 120, 885890. N., Sjoegren, M., DeBernardis, J., rology 71, 14891495.

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 15


Alves et al. Alzheimers disease: a review

Henry, M. L., and Gorno-Tempini, M. Jckel, K. H., Klopp, N., Wichmann, Ijsselstijn, L., Dekker, L. J., Stingl, C., frontal variant of Alzheimers Dis-
L. (2010). The logopenic variant of H. E., Pankratz, V. S., Sando, S. van der Weiden, M. M., Hofman, A., ease. Arch. Neurol. 56, 12331239.
primary progressive aphasia. Curr. B., Aasly, J. O., Barcikowska, M., Kros, J. M., Koudstaal, P. J., Sillevis Jucker, M., and Walker, L. C. (2011).
Opin. Neurol. 23, 633637. Wszolek, Z. K., Dickson, D. W., Smitt, P. A., Ikram, M. A., Breteler, Pathogenic protein seeding in
Herrmann, N., Chau, S. A., Kircan- Graff-Radford, N. R., Petersen, R. C., M. M., and Luider, T. M. (2011). Alzheimer disease and other neu-
ski, I., and Lanctt, K. L. (2011). Alzheimers Disease Neuroimaging Serum levels of pregnancy zone pro- rodegenerative disorders. Ann.
Current and emerging drug treat- Initiative, van Duijn, C. M., Breteler, tein are elevated in presymptomatic Neurol. 70, 532540.
ment options for Alzheimers dis- M. M., Ikram, M. A., DeStefano, Alzheimers disease. J. Proteome Res. Jun, G., Naj, A. C., Beecham, G. W.,
ease: a systematic review. Drugs 71, A. L., Fitzpatrick, A. L., Lopez, O., 10, 49024910. Wang, L. S., Buros, J., Gallins, P. J.,
20312065. Launer, L. J., Seshadri, S., CHARGE Ingelsson, M. (2004). Early Abeta accu- Buxbaum, J. D., Ertekin-Taner, N.,
Herrup, K. (2010). Reimagining Consortium, Berr, C., Campion, D., mulation and progressive synaptic Fallin, M. D., Friedland, R., Inzel-
Alzheimers disease an age- Epelbaum, J., Dartigues, J. F., Tzou- loss, gliosis, and tangle formation in berg, R., Kramer, P., Rogaeva, E., St
based hypothesis. J. Neurosci. 30, rio, C., Alprovitch, A., Lathrop, AD brain. Neurology 62, 925931. George-Hyslop, P., Alzheimers Dis-
1675516762. M., EADI1 consortium, Feulner, T. Isaac, M., Vamvakas, S., Abadie, E., ease Genetics Consortium, Cantwell,
Heyman, A., Peterson, B., Fillenbaum, M., Friedrich, P., Riehle, C., Kraw- Jonsson, B., Gispen, C., and Pani, L. B., Dombroski, B. A., Saykin, A.
G., and Pieper, C. (1996). The con- czak, M., Schreiber, S., Mayhaus, M., L. (2011). Qualication opinion J., Reiman, E. M., Bennett, D. A.,
sortium to establish a registry for Nicolhaus, S., Wagenpfeil, S., Stein- of novel methodologies in the Morris, J. C., Lunetta, K. L., Martin,
Alzheimers disease (CERAD). Part berg, S., Stefansson, H., Stefansson, predementia stage of Alzheimers E. R., Montine, T. J., Goate, A. M.,
XIV: demographic and clinical pre- K., Snaedal, J., Bjrnsson, S., Jons- disease: cerebro-spinal-uid related Blacker, D., Tsuang, D. W., Beekly,
dictors of survival in patients with son, P. V., Chouraki,V., Genier-Boley, biomarkers for drugs affecting amy- D., Cupples, L. A., Hakonarson, H.,
Alzheimers disease. Neurology 46, B., Hiltunen, M., Soininen, H., Com- loid burden regulatory considera- Kukull, W., Foroud, T. M., Haines,
656660. barros, O., Zelenika, D., Delepine, tions by European Medicines Agency J., Mayeux, R., Farrer, L. A., Pericak-
Hoffman, J. M., Welsh-Bohmer, K. A., M., Bullido, M. J., Pasquier, F., Mateo, focusing in improving benet/risk Vance, M. A., and Schellenberg, G. D.
Hanson, M., Crain, B., Hulette, C., I., Frank-Garcia, A., Porcellini, E., in regulatory trials. Eur. Neuropsy- (2010). Meta-analysis conrms CR1,
Earl, N., and Coleman, R. E. (2000). Hanon, O., Coto, E., Alvarez, V., chopharmacol. 21, 781788. CLU, and PICALM as alzheimer dis-
FDG PET imaging in patients with Bosco, P., Siciliano, G., Mancuso, Ito, K., Corrigan, B., Zhao, Q., French, ease risk loci and reveals interactions
pathologically veried dementia. J. M., Panza, F., Solfrizzi, V., Nacmias, J., Miller, R., Soares, H., Katz, E., with APOE genotypes. Arch. Neurol.
Nucl. Med. 41, 19201928. B., Sorbi, S., Boss, P., Piccardi, P., Nicholas, T., Billing, B., Anziano, 67, 14731484.
Hollingworth, P., Harold, D., Jones, Arosio, B., Annoni, G., Seripa, D., R., Fullerton, T., and Alzheimers Kalmijn, S., Launer, J., Lindemans, J.,
L., Owen, M. J., and Williams, J. Pilotto, A., Scarpini, E., Galimberti, Disease Neuroimaging Initiative. Bots, L., Hofman,A., and Breteler, M.
(2011a). Alzheimers disease genet- D., Brice, A., Hannequin, D., Licas- (2011). Disease progression model (1999). Total homocysteine and cog-
ics: current knowledge and future tro, F., Jones, L., Holmans, P. A., Jon- for cognitive deterioration from nitive decline in a community based
challenges. Int. J. Geriatr. Psychiatry sson. T., Riemenschneider, M., Mor- Alzheimers Disease Neuroimag- sample of elderly subjects: the Rot-
26, 793802. gan, K., Younkin, S. G., Owen, M. ing Initiative database. Alzheimers terdam Study. Am. J. Epidemiol. 150,
Hollingworth, P., Harold, D., Sims, J., ODonovan, M., Amouyel, P., and Dement. 7, 151160. 283289.
R., Gerrish, A., Lambert, J. C., Williams, J. (2011b). Common vari- Iverson, D. J., Gronseth, G. S., Reger, Kamboh, M. I., Minster, R. L.,
Carrasquillo, M. M., Abraham, R., ants at ABCA7, MS4A6A/MS4A4E, M. A., Classen, S., Dubinsky, R. Demirci, F. Y., Ganguli, M., Dekosky,
Hamshere, M. L., Pahwa, J. S., EPHA1, CD33 and CD2AP are asso- M., Rizzo, M., and Quality Stan- S. T., Lopez, O. L., and Bar-
Moskvina, V., Dowzell, K., Jones, N., ciated with Alzheimers disease. Nat. dards Subcomittee of the Ameri- mada, M. M. (2012). Association
Stretton, A., Thomas, C., Richards, Genet. 43, 429435. can Academy of Neurology. (2010). of CLU and PICALM variants
A., Ivanov, D., Widdowson, C., Honer, W. G., Dickson, D. W., Gleeson, Practice parameter update: evalua- with Alzheimers disease. Neurobiol.
Chapman, J., Lovestone, S., Powell, J., J., and Davies, P. (1992). Regional tion and management of driving risk Aging 33, 518521.
Proitsi, P., Lupton, M. K., Brayne, C., synaptic pathology in Alzheimers in dementia: report of the Qual- Kantarci, K., and Jack, C. R. Jr.
Rubinsztein, D. C., Gill, M., Lawlor, disease. Neurobiol. Aging 13, ity Standards Subcommittee of the (2003). Neuroimaging in Alzheimer
B., Lynch, A., Brown, K. S., Pass- 375382. American Academy of Neurology. disease: an evidence-based review.
more, P. A., Craig, D., McGuinness, Hort, J., OBrien, J. T., Gainotti, G., Pirt- Neurology 74, 13161324. Neuroimaging Clin. N. Am. 13,
B., Todd, S., Holmes, C., Mann, D., tila, T., Popescu, B. O., Rektorova, Jagust, W., Reed, B., Mungas, D., Ellis, 197209.
Smith, A. D., Beaumont, H., Warden, I., Sorbi, S., Scheltens, P., and on W., and Decarli, C. (2007). What Karow, D. S., McEvoy, L. K., Fennema-
D., Wilcock, G., Love, S., Kehoe, P. G., Behalf of the EFNS Scientist Panel on does uorodeoxyglucose PET imag- Notestine, C., Hagler, D. J. Jr, Jen-
Hooper, N. M., Vardy, E. R., Hardy, Dementia. (2010). EFNS guidelines ing add to a clinical diagnosis of nings, R. G., Brewer, J. B., Hoh, C.
J., Mead, S., Fox, N. C., Rossor, M., for the diagnosis and management dementia? Neurology 69, 871877. K., Dale, A. M., and Alzheimers
Collinge, J., Maier, W., Jessen, F., of Alzheimers disease. Eur. J. Neurol. Jagust, W., Thisted, R., Devous, M. D. Disease Neuroimaging Initiative.
Rther, E., Schrmann, B., Heun, 17, 12361248. Sr., Van Heertum, R., Mayberg, H., (2010). Relative capability of MR
R., Klsch, H., van den Bussche, H., Hyman, B. T., and Gomz-Isla, T. Jobst, K., Smith, A. D., and Borys, imaging and FDG PET to depict
Heuser, I., Kornhuber, J., Wiltfang, J., (1994). Alzheimers disease is a lami- N. (2001). SPECT perfusion imag- changes associated with prodromal
Dichgans, M., Frlich, L., Hampel, nar, regional, and neural system spe- ing in the diagnosis of Alzheimers and early Alzheimer disease. Radiol-
H., Gallacher, J., Hll, M., Rujescu, cic disease, not a global brain dis- disease: a clinical-pathologic study. ogy 256, 932942.
D., Giegling, I., Goate, A. M., Kauwe, ease. Neurobiol. Aging 15, 353354. Neurology 56, 950956. Kawas, C., and Corrada, M. (2006).
J. S., Cruchaga, C., Nowotny, P., Hyman, B. T., Van Hoesen, G. W., Janssen, J. C., Beck, J. A., Campbell, T. Alzheimers and dementia in the
Morris, J. C., Mayo, K., Sleegers, Beyreuther, K., and Masters, C. A., Dickinson, A., Fox, N. C., Harvey, oldest-old: a century of challenges.
K., Bettens, K., Engelborghs, S., De L. (1989). A4 amyloid protein R. J., Houlden, H., Rossor, M. N., and Curr. Alzheimer Res. 3, 411419.
Deyn, P. P., Van Broeckhoven, C., immunoreactivity is present in Collinge, J. (2003). Early onset famil- Kester, M. I., van der Vlies, A. E.,
Livingston, G., Bass, N. J., Gurl- Alzheimers disease neurobrillary ial Alzheimers disease: mutation fre- Blankenstein, M. A., Pijnenburg, Y.
ing, H., McQuillin, A., Gwilliam, tangles. Neurosci. Lett. 101, 352355. quency in 31 families. Neurology 60, A., van Elk, E. J., Scheltens, P., and
R., Deloukas, P., Al-Chalabi, A., Iadecola, C., and Gorelick, P. (2003). 235239. van der Flier, W. M. (2009). CSF
Shaw, C. E., Tsolaki, M., Singleton, Converging pathogenic mechanisms Johnson, J. K., Head, E., Kim, R., Starr, biomarkers predict rate of cogni-
A. B., Guerreiro, R., Mhleisen, T. in vascular and neurodegenerative A., and Cotman, C. W. (1999). Clin- tive decline in Alzheimer disease.
W., Nthen, M. M., Moebus, S., dementia. Stroke 34, 335337. ical and pathological evidence for a Neurology 73, 13531358.

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 16


Alves et al. Alzheimers disease: a review

Kivipelto, M., Ngandu, T., Fratiglioni, L., CR1 associated with Alzheimers dis- Levy-Lahad, E., Wasco, W., Poorkaj, P., Morris, J. C., Yarasheski, K. E., and
Viitanen, M., Kreholt, I., Winblad, ease. Nat. Genet. 41, 10941099. Romano, D. M., Oshima, J., Pettin- Bateman, R. J. (2010). Decreased
B., Helkala, E., Tuomilehto, J., Soini- Lambert, J. C., Zelenika, D., Hiltunen, gell, W. H., Yu, C. E., Jondro, P. D., clearance of CNS beta-amyloid in
nen, H., and Nissinen, A. (2005). M., Chouraki, V., Combarros, O., Schmidt, S. D., and Wang, K., Crow- Alzheimers disease. Science 330,
Obesity and vascular risk factors at Bullido, M. J., Tognoni, G., Fivet, ley, A. C., Fu, Y.-H., Guenette, S. Y., 1774.
midlife and the risk of dementia and N., Boland, A., Arosio, B., Coto, E., Galas, D., Nemens, E., Wijsman, E. Mayeux, R., Reitz, C., Brickman, A.
Alzheimer disease. Arch. Neurol. 62, Del Zompo, M., Mateo, I., Frank- M., Bird, T. D., Schellenberg, G. D., M., Haan, M. N., Manly, J. J.,
15561560. Garcia, A., Helisalmi, S., Porcellini, and Tanzi, R. E. (1995a). Candidate Glymour, M. M., Weiss, C. C.,
Knopman, D. (2006). Dementia and E., Pilotto, A., Forti, P., Ferri, R., gene for the chromosome 1 famil- Yaffe, K., Middleton, L., Hendrie,
cerebrovascular disease. Mayo Clin. Delepine, M., Scarpini, E., Sicil- ial Alzheimers disease locus. Science H. C., Warren, L. H., Hayden, K.
Proc. 81, 223230. iano, G., Solfrizzi, V., Sorbi, S., Spal- 269, 973977. M., Welsh-Bohmer, K. A., Breitner,
Knopman, D. S., DeKosky, S. T., Cum- letta, G., Ravaglia, G., Valdivieso, Levy-Lahad, E., Wijsman, E. M., J. C., and Morris, J. C. (2011).
mings, J. L., Chuit, H., Corey-Bloom, F., Alvarez, V., Bosco, P., Mancuso, Nemens, E., Anderson, L., Goddard, Operationalizing diagnostic criteria
J., Relkin, N., Small, G. W., Miller, M., Panza, F., Nacmias, B., Boss, K. A., Weber, J. L., Bird, T. D., for Alzheimers disease and other
B., and Stevens, J. C. (2001). Prac- P., Piccardi, P., Annoni, G., Seripa, and Schellenberg, G. D. (1995b). age-related cognitive impairment-
tice parameter: diagnosis of demen- D., Galimberti, D., Licastro, F., Lath- A familial Alzheimers disease locus Part 1. Alzheimers Dement. 7,
tia (an evidence-based review). Neu- rop, M., Soininen, H., and Amouyel, on chromosome 1. Science 269, 1534.
rology 56, 11431153. P. (2011). Evidence of the associ- 970973. McKhann, G., Drachman, D., Folstein,
Kukull, W. A., Higdon, R., Bowen, J. D., ation of BIN1 and PICALM with Lobo, A., Launer, L. J., Fratiglioni, L., M., Katzman, R., Price, D., and Stad-
McCormick, W. C., Teri, L., Schel- the AD risk in contrasting Euro- Andersen, K., Di Carlo, A., Breteler, lan, E. M. (1984). Clinical diagno-
lenberg, G. D., van Belle, G., Jolley, pean populations. Neurobiol. Aging M. M., Copeland, J. R., Dartigues, sis of Alzheimers disease: report of
L., and Larson, E. B. (2002). Demen- 32, 756.e11756.e15. J. F., Jagger, C., Martinez-Lage, the NINCDS-ADRDA Work Group
tia and Alzheimer disease incidence: Lapane, K. L., Gambassi, G., Landi, F., J., Soininen, H., and Hofman, A. under the auspices of Department
a prospective cohort study. Arch. Sgadari, A., Mor, V., and Bernabei, (2000). Prevalence of dementia and of Health and Human Services Task
Neurol. 59, 17371746. R. (2001). Gender differences in pre- major subtypes in Europe: a collab- Force on Alzheimers disease. Neu-
Kuller, L., and Lopez, O. (2011). Demen- dictors of mortality in nursing home orative study of population-based rology 34, 939944.
tia and Alzheimers disease: a new residents with AD. Neurology 56, cohorts. Neurologic Diseases in the McKhann, G. M., Knopman, D. S.,
direction. The 2010 Jay L. Fos- 650654. Elderly Research Group. Neurology Chertkow, H., Hyman, B. T., Jack,
ter Memorial Lecture. Alzheimers Larson, E. B., Shadlen, M. F., Wang, 54(Suppl. 5), S4S9. C. R. Jr., Kawas, C. H., Klunk,
Dement. 7, 540550. L., McCormick, W. C., Bowen, J. D., Lopez, O. L., Becker, J. T., Wahed, A. W. E., Koroshetz, W. J., Manly, J.
LaFerla, F. M. (2010). Pathways linking Teri, L., and Kukull, W. A. (2004). S., Saxton, J., Sweet, R. A., Wolk, J., Mayeux, R., Mohs, R. C., Mor-
Abeta and tau pathologies. Biochem. Survival after initial diagnosis of D. A., Klunk, W., and Dekosky, ris, J. C., Rossor, M. N., Schel-
Soc. Trans. 38, 993995. Alzheimer disease. Ann. Intern. Med. S. T. (2009). Long-term effects of tens, P., Carrillo, M. C., Thies, B.,
LaFerla, F. M., Green, K. N., and Oddo, S. 140, 501509. the concomitant use of meman- Weintraub, S., and Phelps, C. H.
(2007). Intracellular amyloid-beta in Launer, L. (2002). Demonstrating the tine with cholinesterase inhibition in (2011). The diagnosis of dementia
Alzheimers disease. Nat. Rev. Neu- case that AD is a vascular disease: Alzheimer disease. J. Neurol. Neuro- due to Alzheimers disease: recom-
rosci. 8, 499509. epidemiologic evidence. Ageing Res. surg. Psychiatr. 80, 600607. mendations from the National Insti-
LaFerla, F. M., and Oddo, S. (2005). Rev. 1, 6177. Lopez, O. L., Brenner, R. P., Becker, J. T., tute on Aging-Alzheimers Asso-
Alzheimers disease: abeta, tau and Launer, L. J., Andersen, K., Dewey, M. Ulrich, R. F., Boller, F., and DeKosky, ciation workgroups on diagnostic
synaptic dysfunction. Trends Mol. E., Letenneur, L., Ott, A., Amaducci, S. T. (1997). EEG spectral abnormal- guidelines for Alzheimers disease.
Med. 11, 170176. L. A., Brayne, C., Copeland, J. R. ities and psychosis as predictors of Alzheimers Dement. 7, 263269.
Lambert, J. C., and Amouyel, P. (2011). M., Dartigues, J.-F., Kragh-Sorensen, cognitive and functional decline in McMonagle, P., Deering, F., Berliner, Y.,
Genetics of Alzheimers disease: new P., Lobo, A., Martinez-Lage, J. M., probable Alzheimers disease. Neu- and Kertesz, A. (2006). The cognitive
evidences for an old hypothesis? Stijnen, T., Hofman, A., and for rology 48, 15211525. prole of posterior cortical atrophy.
Curr. Opin. Genet. Dev. 21, 295301. the EURODEM Incidence Research Loy, C., and Schneider, L. (2006). Neurology 66, 331338.
Lambert, J. C., Heath, S., Even, G., Group and Work Groups. (1999). Galantamine for Alzheimers dis- McShane, R., Areosa Sastre, A., and
Campion, D., Sleegers, K., Hiltunen, Rates and risk factors for demen- ease and mild cognitive impair- Minakaran, N. (2006). Memantine
M., Combarros, O., Zelenika, D., tia and Alzheimers disease: results ment. Cochrane Database Syst. Rev. for dementia. Cochrane Database
Bullido, M. J., Tavernier, B., Leten- from EURODEM pooled analyses. 1, CD001747. Syst. Rev. 2, CD003154.
neur, L., Bettens, K., Berr, C., Neurology 52, 7884. Luchsinger, A., and Mayeux, R. (2004). Mesulam, M., Wicklund, A., Johnson,
Pasquier, F., Fivet, N., Barberger- Le Bastard, N., Martin, J. J., Vanmeche- Dietary factors and Alzheimers dis- N., Rogalski, E., Lger, G. C., Rade-
Gateau, P., Engelborghs, S., De Deyn, len, E., Vanderstichele, H., De Deyn, ease. Lancet Neurol. 3, 579587. maker, A., Weintraub, S., and Bigio,
P., Mateo, I., Franck, A., Helisalmi, P. P., and Engelborghs, S. (2010). Lutz, M. W., Crenshaw, D. G., Saun- E. H. (2008). Alzheimer and fron-
S., Porcellini, E., Hanon, O., Euro- Added diagnostic value of CSF ders, A. M., and Roses, A. D. (2010). totemporal pathology in subsets of
pean Alzheimers Disease Initiative biomarkers in differential demen- Genetic variation at a single locus primary progressive aphasia. Ann.
Investigators, de Pancorbo, M. M., tia diagnosis. Neurobiol. Aging 31, and age of onset for Alzheimers dis- Neurol. 63, 709719.
Lendon, C., Dufouil, C., Jaillard, C., 18671876. ease. Alzheimers Dement. 6, 125131. Mesulam, M. M. (2001). Primary pro-
Leveillard, T., Alvarez, V., Bosco, P., Leung, K. K., Barnes, J., Ridgway, Massoud, F., Desmarais, J. E., and gressive aphasia. Ann. Neurol. 49,
Mancuso, M., Panza, F., Nacmias, G. R., Bartlett, J. W., Clarkson, Gauthier, S. (2011). Switch- 425432.
B., Boss, P., Piccardi, P., Annoni, M. J., Macdonald, K., Schuff, N., ing cholinesterase inhibitors in Mesulam, M. M., and Moran, M. A.
G., Seripa, D., Galimberti, D., Han- Fox, N. C., Ourselin, S., and older adults with dementia. Int. (1987). Cholinesterases within neu-
nequin, D., Licastro, F., Soininen, Alzheimers Disease Neuroimaging Psychogeriatr. 23, 372378. robrillary tangles related to age and
H., Ritchie, K., Blanch, H., Dar- Initiative. (2010). Automated cross- Massoud, F., and Lger, G. C. (2011). Alzheimers disease. Ann. Neurol. 22,
tigues, J. F., Tzourio, C., Gut, I., sectional and longitudinal hip- Pharmacological treatment of 223228.
Van Broeckhoven, C., Alprovitch, pocampal volume measurement in Alzheimer disease. Can. J. Psychiatry Minoshima, S., Foster, N. L., Sima, A. A.,
A., Lathrop, M., and Amouyel, P. mild cognitive impairment and 56, 579588. Frey, K. A., Albin, R. L., and Kuhl,
(2009). Genome-Wide Association Alzheimers disease. Neuroimage 51, Mawuenyega, K. G., Sigurdson, W., D. E. (2001). Alzheimers disease
Study identies variants at CLU and 13451359. Ovod, V., Munsell, L., Kasten, T., versus dementia with Lewy bodies:

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 17


Alves et al. Alzheimers disease: a review

cerebral metabolic distinction with J., Carlson, C. S., Carney, R. M., Alzheimers disease. Pro-CAA posi- Fratiglioni, L. (2006). Heart fail-
autopsy conrmation. Ann. Neurol. Carroll, S. L., Chui, H. C., Clark, tion statement. Neurobiol. Aging 24, ure and risk of dementia and
50, 358365. D. G., Corneveaux, J., Cotman, C. 589597. Alzheimer disease: a population-
Morgan, K. (2011). The three new path- W., Cummings, J. L., DeCarli, C., Nussbaum, R. L., and Ellis, C. E. (2003). based cohort study. Arch. Intern.
ways leading to Alzheimers dis- DeKosky, S. T., Diaz-Arrastia, R., Alzheimers disease and Parkin- Med. 166, 10031008.
ease. Neuropathol. Appl. Neurobiol. Dick, M., Dickson, D. W., Ellis, sons disease. N. Engl. J. Med. 348, Rabinovici, G. D., Jagust, W. J., Furst,
37, 353357. W. G., Faber, K. M., Fallon, K. B., 13561364. A. J., Ogar, J. M., Racine, C. A.,
Mortimer, J. A., Ebbitt, B., Jun, S. P., Farlow, M. R., Ferris, S., Frosch, Oksengard, A. R., Cavallin, L., Axelsson, Mormino, E. C., ONeil, J. P., Lal, R.
and Finch, M. D. (1992). Predic- M. P., Galasko, D. R., Ganguli, M., R., Andersson, C., Ngga, K., Win- A., Dronkers, N. F., Miller, B. L., and
tors of cognitive and functional pro- Gearing, M., Geschwind, D. H., blad, B., Eriksdotter-Jnhagen, M., Gorno-Tempini, M. L. (2008). A
gression in patients with probable Ghetti, B., Gilbert, J. R., Gilman, S., and Wahlund, L. O. (2010). Lack of amyloid and glucose metabolism in
Alzheimers disease. Neurology 42, Giordani, B., Glass, J. D., Growdon, accuracy for the proposed Dubois three variants of primary progressive
16891696. J. H., Hamilton, R. L., Harrell, L. criteriain Alzheimers disease: a val- aphasia. Ann. Neurol. 64, 388401.
Muller, M., Grobbee, D., Aleman, A., E., Head, E., Honig, L. S., Hulette, idation study from the Swedish brain Reddy, P. H. (2011). Abnormal
Bots, M., and van der Schouw, Y. C. M., Hyman, B. T., Jicha, G. A., power initiative. Dement. Geriatr. tau, mitochondrial dysfunction,
(2007). Cardiovascular disease and Jin, L. W., Johnson, N., Karlawish, Cogn. Disord. 30, 374380. impaired axonal transport of mito-
cognitive performance in middle- J., Karydas, A., Kaye, J. A., Kim, R., Pan, X. (2011). Microglial phagocyto- chondria, and synaptic deprivation
aged and elderly men. Atherosclerosis Koo, E. H., Kowall, N. W., Lah, J. sis induced by brillar -amyloid is in Alzheimers disease. Brain Res.
190, 143149. J., Levey, A. I., Lieberman, A. P., attenuated by oligomeric -amyloid: 1415, 136148.
Murray, I., Proza, J., Sohrabji, F., and Lopez, O. L., Mack, W. J., Marson, implications for Alzheimers disease. Reitz, C., Tokuhiro, S., Clark, L. N., Con-
Lawler, J. (2011). Vascular and meta- D. C., Martiniuk, F., Mash, D. C., Mol. Neurodegener. 6, 45. rad, C., Vonsattel, J. P., Hazrati, L. N.,
bolic dysfunction in Alzheimers dis- Masliah, E., McCormick, W. C., Panza, F., Frisardi, V., Imbimbo, B. Palots, A., Lantigua, R., Medrano,
ease: a review. Exp. Biol. Med. 236, McCurry, S. M., McDavid, A. N., P., Seripa, D., Paris, F., Santamato, M., Z Jimnez-Velzquez, I., Var-
772782. McKee, A. C., Mesulam, M., Miller, A., DOnofrio, G., Logroscino, G., darajan, B., Simkin, I., Haines, J.
Musicco, M., Palmer, K., Salamone, G., B. L., Miller, C. A., Miller, J. W., Pilotto, A., and Solfrizzi, V. (2011). L., Pericak-Vance, M. A., Farrer, L.
Lupo, F., Perri, R., Mosti, S., Spalletta, Parisi, J. E., Perl, D. P., Peskind, E., Anti--amyloid immunotherapy for A., Lee, J. H., Rogaeva, E., George-
G., di Iulio, F., Pettenati, C., Crav- Petersen, R. C., Poon, W. W., Quinn, Alzheimers disease: focus on bap- Hyslop, P. S., and Mayeux, R. (2011).
ello, L., and Caltagirone, C. (2009). J. F., Rajbhandary, R. A., Raskind, ineuzumab. Curr. Alzheimer Res. 8, SORCS1 alters amyloid precursor
Predictors of progression of cogni- M., Reisberg, B., Ringman, J. M., 808817. protein processing and variants may
tive decline in Alzheimers disease: Roberson, E. D., Rosenberg, R. N., Perl, D. (2010). Neuropathology of increase Alzheimers disease risk.
the role of vascular and sociode- Sano, M., Schneider, L. S., Seeley, W., Alzheimers disease. Mt. Sinai J. Med. Ann. Neurol. 69, 4764.
mographic factors. J. Neurol. 256, Shelanski, M. L., Slifer, M. A., Smith, 77, 3242. Renner, J. A., Burns, J. M., Hou, C. E.,
12881295. C. D., Sonnen, J. A., Spina, S., Stern, Perry, G., Friedman, R., Shaw, G., McKeel, D. W., Storandt, M., and
Musicco, M., Salamone, G., Calta- R. A., Tanzi, R. E., Trojanowski, J. and Chau, C. (1987). Ubiqui- Morris, J. C. (2004). Progressive pos-
girone, C., Cravello, L., Fadda, L., Q., Troncoso, J. C., Van Deerlin, V. tin is detected in neurobrillary terior cortical dysfunction. A clini-
Lupo, F., Mosti, S., Perri, R., and M., Vinters, H. V., Vonsattel, J. P., tangles and senile plaque neu- copathological series. Neurology 63,
Palmer, K. (2010). Neuropsycholog- Weintraub, S., Welsh-Bohmer, K. rites of Alzheimers disease brains. 11751180.
ical predictors of rapidly progressing A., Williamson, J., Woltjer, R. L., Proc. Natl. Acad. Sci. U.S.A. 84, Rey, A. (1958). LExamen Clinique en
patients with Alzheimers disease. Cantwell, L. B., Dombroski, B. A., 30333036. Psychologie. Paris: Presses Universi-
Dement. Geriatr. Cogn. Disord. 30, Beekly, D., Lunetta, K. L., Martin, Petersen, R. C., and Negash, S. (2008). taires de France.
219228. E. R., Kamboh, M. I., Saykin, A. Mild cognitive impairment: an Rogaev, E. I., Sherrington, R., Rogaeva,
Nagy, Z., Jobst, K. A., Esiri, M. M., J., Reiman, E. M., Bennett, D. overview. CNS Spectr. 13, 4553. E. A., Levesque, G., Ikeda, M., Liang,
Morris, J. H., King, E. M., Mac- A., Morris, J. C., Montine, T. J., Porsteinsson, A. P., Grossberg, G. T., Y., Chi, H., Lin, C., Holman, K.,
Donald, B., Litcheld, S., Barnetson, Goate, A. M., Blacker, D., Tsuang, Mintzer, J., Olin, J. T., and Meman- Tsuda, T., Mar, L., Sorbi, S., Nacmias,
L., and Smith, A. D. (1996). Hip- D. W., Hakonarson, H., Kukull, tine MEM-MD-12 Study Group. B., Piacentini, S., Amaducci, L., Chu-
pocampal pathology reects mem- W. A., Foroud, T. M., Haines, J. (2008). Memantine treatment in makov, I., Cohen, D., Lannfelt, L.,
ory decit and brain imaging mea- L., Mayeux, R., Pericak-Vance, patients with mild to moderate Fraser, P. E., Rommens, J. M., and St
surements in Alzheimers disease: M. A., Farrer, L. A., and Schel- Alzheimers disease already receiv- George-Hyslop, P. H. (1995). Famil-
clinicopathologic correlations using lenberg, G. D. (2011). Common ing a cholinesterase inhibitor: a ial Alzheimers disease in kindreds
three sets of pathologic diagnostic variants at MS4A4/MS4A6E, randomized, double-blind, placebo- with missense mutations in a gene
criteria. Dementia 7, 7681. CD2AP, CD33 and EPHA1 controlled trial. Curr. Alzheimer Res. on chromosome 1 related to the
Naj, A. C., Jun, G., Beecham, G. W., are associated with late-onset 5, 8389. Alzheimers disease type 3 gene.
Wang, L. S., Vardarajan, B. N., Alzheimers disease. Nat. Genet. 43, Potter, P. E. (2010). Investigational med- Nature 376, 775778.
Buros, J., Gallins, P. J., Buxbaum, 436441. ications for treatment of patients Rogaeva, E., Meng, Y., Lee, J. H., Gu,
J. D., Jarvik, G. P., Crane, P. K., Nelson, P. T., Head, E., Schmitt, F. A., with Alzheimer disease. J. Am. Y., Kawarai, T., Zou, F., Katayama, T.,
Larson, E. B., Bird, T. D., Boeve, B. Davis, P. R., Neltner, J. H., Jicha, G. Osteopath. Assoc. 110(Suppl. 8), Baldwin, C. T., Cheng, R., Hasegawa,
F., Graff-Radford, N. R., De Jager, A., Abner, E. L., Smith, C. D., Van S27S36. H., Chen, F., Shibata, N., Lunetta,
P. L., Evans, D., Schneider, J. A., Eldik, L. J., Kryscio, R. J., and Scheff, Prvulovic, D., and Hampel, H. (2011). K. L., Pardossi-Piquard, R., Bohm,
Carrasquillo, M. M., Ertekin-Taner, S. W. (2011). Alzheimers disease Amyloid (A) and phospho-tau C., Wakutani, Y., Cupples, L. A.,
N., Younkin, S. G., Cruchaga, C., is not brain aging: neuropatho- (p-tau) as diagnostic biomarkers in Cuenco, K. T., Green, R. C., Pinessi,
Kauwe, J. S., Nowotny, P., Kramer, P., logical, genetic, and epidemiologi- Alzheimers disease. Clin. Chem. Lab. L., Rainero, I., Sorbi, S., Bruni, A.,
Hardy, J., Huentelman, M. J., Myers, cal human studies. Acta Neuropathol. Med. 49, 367374. Duara, R., Friedland, R. P., Inzel-
A. J., Barmada, M. M., Demirci, 121, 571587. Qiu, C., Ronchi, D., and Fratiglioni, berg, R., Hampe, W., Bujo, H., Song,
F. Y., Baldwin, C. T., Green, R. C., Nicoll, J. A., Yamada, M., Frack- L. (2007). The epidemiology of the Y. Q., Andersen, O. M., Willnow, T.
Rogaeva, E., St George-Hyslop, P., owiak, J., Mazur-Kolecka, B., dementias: an update. Curr. Opin. E., Graff-Radford, N., Petersen, R.
Arnold, S. E., Barber, R., Beach, and Weller, R. O. (2004). Cere- Psychiatry 20, 380385. C., Dickson, D., Der, S. D., Fraser,
T., Bigio, E. H., Bowen, J. D., bral amyloid angiopathy plays a Qiu, C., Winblad, B., Marengoni, P. E., Schmitt-Ulms, G., Younkin,
Boxer, A., Burke, J. R., Cairns, N. direct role in the pathogenesis of A., Klarin, I., Fastbom, J., and S., Mayeux, R., Farrer, L. A., and

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 18


Alves et al. Alzheimers disease: a review

St George-Hyslop, P. (2007). The Alphase Study. J. Nutr. Health Aging C., Du, Y., Rotter, J. I., Aulchenko, brain metabolism and long-term
neuronal sortilin-related receptor 13, 808812. Y. S., Harris, T. B., Petersen, R. outcome. JAMA 286, 21202127.
SORL1 is genetically associated with Saunders, A. M., Strittmatter, W. J., C., Berr, C., Owen, M. J., Lopez- Snider, B. J., Fagan, A. M., Roe, C., Shah,
Alzheimer disease. Nat. Genet. 39, Schmechel, D., George-Hyslop, P. H., Arrieta, J.,Varadarajan, B. N., Becker, A. R., Grant, E. A., Xiong, C., Morris,
168177. Pericak-Vance, M. A., Joo, S. H., J. T., Rivadeneira, F., Nalls, M. J. C., and Holtzman, D. M. (2009).
Romn, G. C., and Royall, D. R. Rosi, B. L., Gusella, J. F., Crapper- A., Graff-Radford, N. R., Campion, Cerebrospinal uid biomarkers and
(2004). A diagnostic dilemma: is MacLachlan, D. R., Alberts, M. J., D., Auerbach, S., Rice, K., Hof- rate of cognitive decline in very mild
Alzheimers dementia Alzheimers Hulette, C., Crain, B., Goldgaber, D., man, A., Jonsson, P. V., Schmidt, dementia of the Alzheimer type.
disease, vascular dementia, or both? and Roses, A. D. (1993). Association H., Lathrop, M., Mosley, T. H., Arch. Neurol. 66, 638645.
Lancet Neurol. 3, 141. of apolipoprotein E allele epsilon 4 Au, R., Psaty, B. M., Uitterlinden, Sperling, R. A., Aisen, P. S., Beckett, L.
Roselli, F., Tartaglione, B., Federico, F., with late-onset familial and sporadic A. G., Farrer, L. A., Lumley, T., A., Bennett, D. A., Craft, S., Fagan, A.
Lepore, V., Defazio, G., and Livrea, P. Alzheimers disease. Neurology 43, Ruiz, A., Williams, J., Amouyel, P., M., Iwatsubo, T., Jack, C. R. Jr., Kaye,
(2009). Rate of MMSE score change 14671472. Younkin, S. G., Wolf, P. A., Launer, J., Montine, T. J., Park, D. C., Reiman,
in Alzheimers disease: inuence of Schellenberg, G. D., Bird, T. D., Wijs- L. J., Lopez, O. L., van Duijn, E. M., Rowe, C. C., Siemers, E., Stern,
education and vascular risk fac- man, E. M., Orr, H. T., Anderson, C. M., Breteler, M. M., CHARGE Y., Yaffe, K., Carrillo, M. C., Thies, B.,
tors. Clin. Neurol. Neurosurg. 111, L., Nemens, E., White, J. A., Bonny- Consortium, GERAD1 Consortium, Morrison-Bogorad, M., Wagster, M.
327330. castle, L., Weber, J. L., Alonso, M. and EADI1 Consortium. (2010). V., and Phelps, C. H. (2011). Toward
Roses, A. D. (2010). An inherited E., Potter, H., Heston, L. L., Mar- Genome-wide analysis of genetic dening the preclinical stages of
variable poly-T repeat genotype tin, G. M. (1992). Genetic linkage loci associated with Alzheimer dis- Alzheimers disease: recommenda-
in TOMM40 in Alzheimer disease. evidence for a familial Alzheimers ease. JAMA 303, 18321840. tions from the National Insti-
Arch. Neurol. 67, 536541. disease locus on chromosome 14. Sherrington, R., Froelich, S., Sorbi, S., tute on Aging-Alzheimers Asso-
Roses, A. D., Lutz, M. W., Amrine- Science 258, 668671. Campion, D., Chi, H., Rogaeva, E. A., ciation workgroups on diagnostic
Madsen, H., Saunders, A. M., Cren- Scheltens, P., Leys, D., Barkhof, F., Levesque, G., Rogaev, E. I., Lin, C., guidelines for Alzheimers disease.
shaw, D. G., Sundseth, S. S., Huentel- Huglo, D., Weinstein, H. C., Verm- Liang, Y., Ikeda, M., Mar, L., Brice, Alzheimers Dement. 7, 280292.
man, M. J., Welsh-Bohmer, K. A., ersch, P., Kuiper, M., Steinling, M., A., Agid, Y., Percy, M. E., Clerget- Stern, Y. (2006). Cognitive reserve and
and Reiman, E. M. (2010). A Wolters, E. C., and Valk, J. (1992). Darpoux, F., Piacentini, S., Marcon, Alzheimer disease. Alzheimer Dis.
TOMM40 variable-length polymor- Atrophy of medial temporal lobes on G., Nacmias, B., Amaducci, L., Fre- Assoc. Disord. 20, S69S74.
phism predicts the age of late- MRI in probable Alzheimers dis- bourg, T., Lannfelt, L., Rommens, Stern,Y.,Albert, M., Brandt, J., Jacobs, D.
onset Alzheimers disease. Pharma- ease and normal ageing: diagnostic J. M., and St George-Hyslop, P. H. M., Tang, M. X., Marder, K., Bell, K.,
cogenomics J. 10, 375384. value and neuropsychological corre- (1996). Alzheimers disease associ- Sano, M., Devanand, D. P., Bylsma,
Rovio, S., Kreholt, I., Helkala, E., Viita- lates. J. Neurol. Neurosurg. Psychiatr. ated with mutations in presenilin 2 F., and Laeche, G. (1994). Utility
nen, M., Winblad, B., Tuomilehto, 55, 967972. is rare and variably penetrant. Hum. of extrapyramidal signs and psy-
J., Soininen, H., Nissinen, A., and Schoonenboom, N. S., Reesink, F. E., Mol. Genet. 5, 985988. chosis as predictors of cognitive and
Kivipelto, M. (2005). Leisure-time Verwey, N. A., Kester, M. I., Teunis- Sherrington, R., Rogaev, E. I., Liang, Y., functional decline, nursing home
physical activity at midlife and the sen, C. E., van de Ven, P. M., Pij- Rogaeva, E. A., Levesque, G., Ikeda, admission, and death in Alzheimers
risk of dementia and Alzheimers nenburg, Y. A., Blankenstein, M. A., M., Chi, H., Lin, C., Li, G., Holman, disease: prospective analyses from
disease. Lancet Neurol. 4, 705711. Rozemuller, A. J., Scheltens, P., and K., Tsuda, T., Mar, L., Foncin, J. F., the Predictors Study. Neurology 44,
Ruitenberg, A., Heijer, T., Bakker, S., van der Flier, W. M. (2012). Cere- Bruni, A. C., Montesi, M. P., Sorbi, 23002307.
van Swieten, J., Koudstaal, P., Hof- brospinal uid markers for differ- S., Rainero, I., Pinessi, L., Nee, L., Strittmatter, W. J., Saunders, A. M.,
man, A., and Breteler, M. (2005). ential dementia diagnosis in a large Chumakov, I., Pollen, D., Brookes, Schmechel, D., Pericak-Vance, M.,
Cerebral hypoperfusion and clinical memory clinic cohort. Neurology 78, A., Sanseau, P., Polinsky, R. J., Wasco, Enghild, J., Salvesen, G. S., and
onset of dementia: the Rotterdam 4754. W., Da Silva, H. A., Haines, J. L., Roses, A. D. (1993a). Apolipopro-
Study. Ann. Neurol. 57, 789794. Schor, N. F. (2011). What the halted Perkicak-Vance, M. A., Tanzi, R. E., tein E: high-avidity binding to beta-
Ruitenberg, A., Ott, A., Swieten, J., Hof- phase III -secretase inhibitor trial Roses, A. D., Fraser, P. E., Rommens, amyloid and increased frequency of
man, A., and Breteler, M. (2001). may (or may not) be telling us. Ann. J. M., and St George-Hyslop, P. H. type 4 allele in late-onset familial
Incidence of dementia: does gender Neurol. 69, 237239. (1995). Cloning of a gene bearing Alzheimer disease. Proc. Natl. Acad.
make a difference? Neurobiol. Aging Seshadri, S., Beiser, A., Au, R., Wolf, P. A., missense mutations in early-onset Sci. U.S.A. 90, 19771981.
22, 575580. Evans, D. A., Wilson, R. S., Petersen, familial Alzheimers disease. Nature Strittmatter, W. J., Weisgraber, K. H.,
Salomone, S., Caraci, F., Leggio, G. M., R. C., Knopman, D. S., Rocca, W. 375, 754760. Huang, D. Y., Dong, L. M., Salvesen,
Fedotova, J., and Drago, F. (2012). A., Kawas, C. H., Corrada, M. M., Sikkes, S. A., de Lange-de Klerk, E. S., G. S., Pericak-Vance, M., Schmechel,
New pharmacological strategies for Plassman, B. L., Langa, K. M., and Pijnenburg, Y. A., Scheltens, P., and D., Saunders, A. M., Goldgaber, D.,
treatment of Alzheimers disease: Chui, H. C. (2011). Operationalizing Uitdehaag, B. M. (2009). A system- and Roses, A. D. (1993b). Bind-
focus on disease-modifying drugs. diagnostic criteria for Alzheimers atic review of Instrumental Activities ing of human apolipoprotein E
Br. J. Clin. Pharmacol. 73, 504517. disease and other age-related cogni- of Daily Living scales in dementia: to synthetic amyloid beta peptide:
Sarazin, M., Berr, C., De Rotrou, J., Fab- tive impairment-Part 2. Alzheimers room for improvement. J. Neurol. isoform-specic effects and implica-
rigoule, C., Pasquier, F., Legrain, S., Dement. 7, 3552. Neurosurg. Psychiatr. 80, 712. tions for late-onset Alzheimer dis-
Michel, B., Puel, M., Volteau, M., Seshadri, S., Fitzpatrick, A. L., Ikram, Silverman, D. H., Small, G. W., Chang, ease. Proc. Natl. Acad. Sci. U.S.A. 90,
Touchon, J., Verny, M., and Dubois, M. A., DeStefano, A. L., Gudna- C. Y., Lu, C. S., Kung De Aburto, M. 80988102.
B. (2007). Amnestic syndrome of son, V., Boada, M., Bis, J. C., Smith, A., Chen, W., Czernin, J., Rapoport, Takahashi, R. H., Capetillo-Zarate, E.,
the medial temporal type identies A. V., Carassquillo, M. M., Lam- S. I., Pietrini, P., Alexander, G. E., Lin, M. T., Milner, T. A., and
prodromal AD: a longitudinal study. bert, J. C., Harold, D., Schrijvers, Schapiro, M. B., Jagust, W. J., Hoff- Gouras, G. K. (2010). Co-occurrence
Neurology 69, 18591867. E. M., Ramirez-Lorca, R., Debette, man, J. M., Welsh-Bohmer, K. A., of Alzheimers disease beta-amyloid
Saumier, D., Duong, A., Haine, D., S., Longstreth, W. T. Jr., Janssens, Alavi, A., Clark, C. M., Salmon, E., de and tau pathologies at synapses.
Garceau, D., and Sampalis, J. (2009). A. C., Pankratz, V. S., Dartigues, J. Leon, M. J., Mielke, R., Cummings, Neurobiol. Aging 31, 11451152.
Domain-specic cognitive effects of F., Hollingworth, P., Aspelund, T., J. L., Kowell, A. P., Gambhir, S. S., Tam, C. W. C., Burton, E. J., McKeith,
tramiprosate in patients with mild Hernandez, I., Beiser, A., Kuller, L. Hoh, C. K., and Phelps, M. E. (2001). I. G., Burn, D. J., and OBrien, J.
to moderate Alzheimers disease: H., Koudstaal, P. J., Dickson, D. W., Positron emission tomography in T. (2005). Temporal lobe atrophy
ADAS-cog subscale results from the Tzourio, C., Abraham, R., Antunez, evaluation of dementia: regional on MRI in Parkinsons disease

www.frontiersin.org April 2012 | Volume 3 | Article 63 | 19


Alves et al. Alzheimers disease: a review

with dementia: a comparison with G. (2011). Progression of cogni- memory failure in Alzheimers dis- Study Collaborators. (2008). Sur-
Alzheimers disease and dementia tive, functional, and neuropsychi- ease. Neuron 44, 181193. vival times in people with demen-
with Lewy bodies. Neurology 64, atric symptom domains in a popula- Wang, X., Karp, A., Winblad, B., tia: analysis from population based
861865. tion cohort with Alzheimer demen- and Fratiglioni, L. (2002). Late- cohort study with 14 year follow-up.
Tang-Wai, D. F., Graff-Radford, N. R., tia: the Cache County Dementia life engagement in social and BMJ 336, 258262.
Boeve, B. F., Dickson, D. W., Parisi, Progression study. Am. J. Geriatr. leisure activities is associated with a Xu, G., Liu, X., Yin, Q., Zhu, W., Zhang,
J. E., Crook, R., Caselli, R. J., Knop- Psychiatry 19, 532542. decreased risk of dementia: a lon- R., and Fan, X. (2009). Alcohol con-
man, D. S., and Petersen, R. C. Tyas, L., Manfreda, J., Strain, A., gitudinal study from the Kungshol- sumption and transition of mild
(2004). Clinical, genetic, and neu- and Montgomery, R. (2001). Risk men project. Am. J. Epidemiol. 155, cognitive impairment to dementia.
ropathologic characteristics of pos- factors for Alzheimers disease: 10811087. Psychiatry Clin. Neurosci. 63, 4349.
terior cortical atrophy. Neurology 63, a population-based, longitudinal Welge,V., Fiege, O., Lewczuk, P., Mollen- Yao, P. J., Zhu, M., Pyun, E. I., Brooks, A.
11681174. study in Manitoba, Canada. Int. J. hauer, B., Esselmann, H., Klafki, H. I., Therianos, S., Meyers, V. E., and
Tariot, P. N., Farlow, M. R., Gross- Epidemiol. 30, 590597. W., Wolf, S., Trenkwalder, C., Otto, Coleman, P. D. (2003). Defects in
berg, G. T., Graham, S. M., McDon- van Duijn, C. M., de Knijff, P., Cruts, M., Kornhuber, J., Wiltfang, J., and expression of genes related to synap-
ald, S., Gergel, I., and Memantine M., Wehnert, A., Havekes, L. M., Bibl, M. (2009). Combined CSF tau, tic vesicle trafcking in frontal cor-
Study Group. (2004). Memantine Hofman, A., and Van Broeckhoven, p-tau181 and amyloid-beta 38/40/42 tex of Alzheimers disease. Neurobiol.
treatment in patients with moderate C. (1994). Apolipoprotein E4 allele for diagnosing Alzheimers disease. J. Dis. 12, 97109.
to severe Alzheimer disease already in a population-based study of Neural Transm. 116, 203212.
receiving donepezil: a randomized early-onset Alzheimers disease. Nat. Winblad, B., Grossberg, G., Frlich, Conict of Interest Statement: The
controlled trial. JAMA 291, 317324. Genet. 7, 7478. L., Farlow, M., Zechner, S., Nagel, authors declare that the research was
Tedde, A., Nacmias, B., Ciantelli, M., Vemuri, P., Simon, G., Kantarci, K., J., and Lane, R. (2007). IDEAL: conducted in the absence of any com-
Forleo, P., Cellini, E., Bagnoli, S., Whitwell, J. L., Senjem, M. L., Przy- a 6-month, double-blind, placebo- mercial or nancial relationships that
Piccini, C., Caffarra, P., Ghidoni, belski, S. A., Gunter, J. L., Josephs, controlled study of the rst skin could be construed as a potential con-
E., Paganini, M., Bracco, L., and K. A., Knopman, D. S., Boeve, B. F., patch for Alzheimer disease. Neurol- ict of interest.
Sorbi, S. (2003). Identication of Ferman, T. J., Dickson, D. W., Parisi, ogy 69(Suppl. 1), S14S22.
new presenilin gene mutations in J. E., Petersen, R. C., and Jack, C. Wingo, T. S., Lah, J. J., Levey, A. I., Received: 31 January 2012; paper pending
early-onset familial Alzheimer dis- R. Jr. (2011). Antemortem differen- and Cutler, D. J. (2012). Autosomal published: 13 February 2012; accepted:
ease. Arch. Neurol. 60, 15411544. tial diagnosis of dementia pathology recessive causes likely in early-onset 02 April 2012; published online: 20 April
Terry, R. D. (1963). The ne struc- using structural MRI: differential- Alzheimer disease. Arch. Neurol. 69, 2012.
ture of neurobrillary tangles in STAND. Neuroimage 55, 522531. 5964. Citation: Alves L, Correia ASA, Miguel
Alzheimers disease. J. Neuropathol. Visser, P. J., Verhey, F., Knol, D. L., Schel- Wirths, O., Multhaup, G., Czech, C., R, Alegria P and Bugalho P (2012)
Exp. Neurol. 22, 629642. tens, P., Wahlund, L. O., Freund- Blanchard, V., Moussaoui, S., Tremp, Alzheimers disease: a clinical practice-
Terry, R. D., Masliah, E., Salmon, D. Levi, Y., Tsolaki, M., Minthon, L., G., Pradier, L., Beyreuther, K., and oriented review. Front. Neur. 3:63. doi:
P., Butters, N., DeTeresa, R., Hill, Wallin, A. K., Hampel, H., Brger, Bayer, T. A. (2001). Intraneuronal 10.3389/fneur.2012.00063
R., Hansen, L. A., and Katzman, R. K., Pirttila, T., Soininen, H., Rikkert, Abeta accumulation precedes plaque This article was submitted to Frontiers
(1991). Physical basis of cognitive M. O., Verbeek, M. M., Spiru, L., formation in beta-amyloid precur- in Dementia, a specialty of Frontiers in
alterations in Alzheimers disease: and Blennow, K. (2009). Prevalence sor protein and presenilin-1 double- Neurology.
synapse loss is the major correlate of and prognostic value of CSF mark- transgenic mice. Neurosci. Lett. 306, Copyright 2012 Alves, Correia, Miguel,
cognitive impairment. Ann. Neurol. ers of Alzheimers disease pathology 116120. Alegria and Bugalho. This is an open-
30, 572580. in patients with subjective cogni- Wyss-Coray, T. (2007). Inammation access article distributed under the terms
Tschanz, J. T., Corcoran, C. D., Schwartz, tive impairment or mild cognitive in Alzheimer disease: driving force, of the Creative Commons Attribution
S., Treiber, K., Green, R. C., Norton, impairment in the DESCRIPA study: bystander or benecial response? Non Commercial License, which per-
M. C., Mielke, M. M., Piercy, K., a prospective cohort study. Lancet Nat. Med. 12, 10051015. mits non-commercial use, distribution,
Steinberg, M., Rabins, P. V., Leout- Neurol. 8, 619627. Xie, J., Brayne, C., Matthews, F. and reproduction in other forums, pro-
sakos, J. M., Welsh-Bohmer, K. A., Walsh, D. M., and Selkoe, D. J. (2004). E., and Medical Research Council vided the original authors and source are
Breitner, J. C., and Lyketsos, C. Deciphering the molecular basis of Cognitive Function and Ageing credited.

Frontiers in Neurology | Dementia April 2012 | Volume 3 | Article 63 | 20

Vous aimerez peut-être aussi