Neuroimaging biomarkers in Alzheimer’s disease and other dementias

Neuroimaging biomarkers in Alzheimer’s disease and other dementias

Accepted Manuscript Title: Neuroimaging biomarkers in Alzheimer’s disease and other dementias Author: Victor L. Villemagne Ga¨el Ch´etelat PII: DOI: R...

1MB Sizes 1 Downloads 84 Views

Accepted Manuscript Title: Neuroimaging biomarkers in Alzheimer’s disease and other dementias Author: Victor L. Villemagne Ga¨el Ch´etelat PII: DOI: Reference:

S1568-1637(16)30004-6 http://dx.doi.org/doi:10.1016/j.arr.2016.01.004 ARR 635

To appear in:

Ageing Research Reviews

Received date: Revised date: Accepted date:

11-12-2015 21-1-2016 22-1-2016

Please cite this article as: Villemagne, Victor L., Ch´etelat, Ga¨el, Neuroimaging biomarkers in Alzheimer’s disease and other dementias.Ageing Research Reviews http://dx.doi.org/10.1016/j.arr.2016.01.004 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Neuroimaging biomarkers in Alzheimer’s disease and other dementias Running title: Aβ imaging, metabolism and inflammation Victor L. Villemagne MDa,b,d* [email protected], Gaël Chételat PhDd,e,f,g a

Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Victoria 3084, Australia. b Department of Medicine, University of Melbourne, Austin Health, Victoria 3084, Australia. c The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia. d Institut National de la Santé et de la Recherche Médicale (Inserm), Unité 1077, Caen, France. e Université de Caen Basse-Normandie, Unité Mixte de Recherche (UMR), S1077, Caen, France. f Ecole Pratique des Hautes Etudes, UMR-S1077, 14000, Caen, France. g Unité 1077, Centre Hospitalier Universitaire de Caen, 14000, Caen, France. * Corresponding author at: Department of Nuclear Medicine, Centre for PET, Austin Health, 145 Studley Road, Heidelberg, Vic. 3084, Australia. Tel.: +61-3-9496 3321. Fax +61-3-9496 5663.

1

ABSTRACT In vivo imaging of amyloid ( has transformed the assessment of  pathology and its changes over time, extending our insight into A deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional or global A burden in the brain. This knowledge is essential for therapeutic trial recruitment and for the evaluation of anti- treatments. Although cross sectional evaluation of  burden does not strongly correlate with cognitive impairment, it does correlate with cognitive (especially memory) decline and with a higher risk for conversion to AD in the aging population and MCI subjects. This suggests that  deposition is a protracted pathological process starting well before the onset of symptoms. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of A are required to confirm this hypothesis and further elucidate the role of  deposition in the course of Alzheimer‟s disease.

Keywords: Alzheimer‟s disease; A; positron emission tomography; neurodegenerative disorders; brain imaging

2

1.

INTRODUCTION

Alzheimer‟s disease (AD) is a progressive and irreversible neurodegenerative disorder clinically characterized by memory loss and cognitive decline that severely affect the activities of daily living (Masters et al., 2006). AD is the leading cause of dementia in the elderly, leading invariably to death, usually within 7-10 years after diagnosis (Khachaturian, 1985). The progressive nature of the neurodegeneration suggests an age-dependent process that ultimately leads to synaptic failure and neuronal damage in cortical areas of the brain essential for memory and other cognitive domains (Isacson et al., 2002). AD not only has devastating effects on the sufferers and their caregivers, but it also has a tremendous socioeconomic impact on families and the health system; a burden which will only increase in the upcoming years as the population of most countries ages (Johnson et al., 2000). In the absence of reliable biomarkers, direct pathologic examination of brain tissue derived from either biopsy or autopsy remains the only definitive method for establishing a diagnosis of AD (O'Brien et al., 2000). The typical macroscopic picture is gross cortical atrophy, whilst microscopically, there is widespread cellular degeneration and diffuse synaptic and neuronal loss, accompanied by reactive gliosis and the presence of the pathological hallmarks of the disease: intracellular neurofibrillary tangles (NFT) and extracellular -amyloid () plaques (Jellinger, 1990; Masters, 2005; Masters and Beyreuther, 2005). Whilst NFT are intraneuronal bundles of paired helical filaments mainly composed of the aggregates of an abnormally phosphorylated form of tau protein, (Jellinger and Bancher, 1998; Michaelis et al., 2002) neuritic plaques consist of dense extracellular aggregates of , (Masters et al., 1985) surrounded by reactive gliosis and dystrophic neurites.  is a 4 kDa 39–43 amino acid metalloprotein derived from the proteolytic cleavage of the amyloid precursor protein (APP), by  and -secretases (Cappai and White, 1999). To date, all available genetic, pathological, biochemical and cellular evidence strongly supports the notion that an imbalance between the production and removal of leading to its progressive accumulation is central to the 3

pathogenesis of AD (Villemagne et al., 2006). The “Acentric theory” (Masters et al., 2006) postulates that A plaque deposition is the primary event in a cascade of effects that lead to neurofibrillary degeneration and dementia (Hardy, 1997), while other hypotheses postulate that A deposition occurs in parallel with other interacting pathological events, as a necessary, but not sufficient, cause of the pathological processes leading to AD dementia (Chetelat, 2013; Pimplikar et al., 2010; Small and Duff, 2008).

While biomarkers play a crucial role in the new diagnostic criteria for AD, (McKhann et al., 2011), the clinical diagnosis of AD is still largely based on progressive impairment of memory and decline in at least one other cognitive domain, as well as excluding other conditions that might also present with dementia such as frontotemporal lobar degeneration (FTLD), dementia with Lewy-bodies (DLB), stroke, brain tumour, normal pressure hydrocephalus or depression (Cummings et al., 1998; Larson et al., 1996). A variable period of up to five years of prodromal decline in cognition characterized by a relatively isolated impairment in recent episodic memory that may also be accompanied by impairments of working memory, known as amnestic Mild Cognitive Impairment (MCI), usually precedes the formal diagnosis of AD (Petersen, 2000; Petersen et al., 1999). At this point there is no cure for AD nor proven way to slow the rate of neurodegeneration. Symptomatic treatment with an acetylcholinesterase inhibitor or a glutamatergic moderator provides modest benefit in some patients usually by temporary stabilization rather than a noticeable improvement in memory function (Masters and Beyreuther, 2006).

2. A IMAGING IN ALZHEIMER’S DISEASE The development of PET tracers for  deposition has allowed in vivo assessment of  pathology. Since the first study published more than 10 years ago (Klunk et al., 2004), several hundreds of  PET studies have been performed, improving our knowledge on the 4

topography, timing, propagation, rates of accumulation and covariates of  accumulation in the living Human brain. On visual inspection, cortical 11C-PiB retention is usually higher in AD than in cognitively intact controls. Quantitative PET studies using diverse  radiotracers have shown a robust difference in retention between AD patients and age-matched controls (Barthel et al., 2011; Camus et al., 2012; Cselenyi et al., 2012; Fleisher et al., 2011; Furst et al., 2010; Jack et al., 2008a; Klunk et al., 2004; Kudo et al., 2007; Mintun et al., 2006; Morris et al., 2009; Pike et al., 2007; Price et al., 2005; Rabinovici et al., 2007; Rowe et al., 2010; Rowe et al., 2007; Villemagne et al., 2011a). The binding is more particularly elevated in frontal, cingulate, precuneus, striatum, parietal, and lateral temporal cortices, while occipital, sensorimotor and mesial temporal cortices are much less affected. Both quantitative and visual assessment of PET images present a pattern of radiotracer retention that seems to replicate the sequence of  deposition found at autopsy (Braak and Braak, 1997), with initial deposition in the orbitofrontal cortex, inferior temporal, cingulate gyrus and precuneus, followed by the remaining prefrontal cortex, lateral temporal and parietal cortices (Figure 1). Multimodal neuroimaging studies in early AD have shown that the regional pattern of  deposition is similar to the anatomy of the „default network‟, (Buckner et al., 2005; Sperling et al., 2009) a specific, anatomically defined brain system responsible for internal modes of cognition, such as self-reflection processes, conscious resting state, or episodic memory retrieval (Buckner et al., 2008; Wermke et al., 2008).

While the overall pattern of  deposition is roughly the same across different phenotypes, there are some particular cases associated with a particular regional distribution of 11C-PiB retention – e.g. carriers of mutations associated with familial AD have particularly elevated retention in subcortical gray matter nuclei, while congophilic angiopathy (CAA) and posterior

5

cortical atrophy present a posterior pattern of tracer retention (Klunk et al., 2007; Koivunen et al., 2008b; Villemagne et al., 2009a). Longitudinal studies with 11C-PiB have shown that significant, albeit small, increases in  deposition can be measured, and that these increases in  deposition are present in the whole spectrum of cognitive stages, from cognitively unimpaired individuals to AD dementia patients (Engler et al., 2006; Jack et al., 2009; Okello et al., 2009b; Resnick et al., 2010; Rinne et al., 2010; Sojkova et al., 2011; Villain et al., 2012; Villemagne et al., 2011b). accumulation is also observed in individuals considered to have low  loads with about 7% of them progressing above the threshold in about 2.5 years (Vlassenko et al., 2011). In fact, both accumulators and non-accumulators could be identified, especially within individuals with low- deposition.  deposition has been estimated to take about 11 years for a PIB-negative subject to reach the threshold of PiB positivity (1·5 SUVR), and about 1520 years to go from this threshold to the levels usually observed in mild AD (Jack et al., 2013b; Villain et al., 2012; Villemagne et al., 2013). accumulation is thus a slow process that is thought to remain relatively constant across clinical stages i.e. similar in cognitively normal, and MCI accumulators (Jack et al., 2013b; Villemagne et al., 2013), yet tends to accelerate with increasing  load and to slow down, approaching a plateau, at the latest stages of amyloidosis (i.e. in individuals with highest  burden) (Jack et al., 2013b; Villain et al., 2012; Villemagne et al., 2013). This slower rate of  deposition might be attributed to a saturation of the process and/or to the extensive neuronal death that precludes production.

3. A DEPOSITION IN NON DEMENTED INDIVIDUALS AND ITS RELATION WITH COGNITION

6

About 25-35% of elderly subjects performing within normal limits on cognitive tests present with high cortical

11

C-PiB retention, predominantly in the prefrontal and posterior

cingulate/precuneus regions (Aizenstein et al., 2008; Mintun et al., 2006; Mormino et al., 2009; Reiman et al., 2009; Rowe et al., 2010; Rowe et al., 2007; Villemagne et al., 2008b). These findings are in perfect agreement with post mortem reports that ~25% of non-demented older individuals over the age of 75 have A plaques, (Davies et al., 1988; Forman et al., 2007; Morris and Price, 2001). Furthermore, the prevalence of subjects with high

11

C-PiB

retention increases each decade at the same rate as the reported prevalence of plaques in nondemented subjects in autopsy studies (Rowe et al., 2010). Within patients with MCI, approximately 50-70% present with high cortical

11

C-PiB retention (Forsberg et al., 2008;

Jansen et al., 2015; Kemppainen et al., 2007; Klunk, 2011; Lowe et al., 2009; Mormino et al., 2009; Pike et al., 2007; Price et al., 2005). Most studies found that subjects classified as nonamnestic MCI, especially the single-domain subtype, show low  levels in the brain consistent with a non-AD underlying pathological process (Lowe et al., 2009; Pike et al., 2007), although other reports did not (Wolk et al., 2009). The detection of A pathology at the presymptomatic stage is of crucial importance in order to both identify the group that may benefit the most from therapies aimed at reducing or eliminating  from the brain, as well as assess the potential effects of  deposition on cognition and/or neurodegeneration at the early stages of the disease process (Chetelat et al., 2010a; Chetelat et al., 2012).

Numerous cross-sectional studies have assessed the relationships between Adeposition and memory performances in demented and non-demented populations. While there is general agreement that Aburden does not correlate with memory impairment or disease severity at the dementia stage, the assessment of controls is more controversial, with studies reporting small or no significant relationships (Aizenstein et al., 2008; Jack et al., 2008a; Mintun et al., 2006; Rowe et al., 2010), while others reporting one (Pike et al., 2007; Sperling et al., 2012), 7

often restricted to specific conditions (e.g. only in females (Pike et al., 2011); only in nonApoE4 carriers (Pike et al., 2011); mainly in APOE4 carriers (Kantarci et al., 2012), or in low-educated individuals (Rentz et al., 2010). Some reports even found higher performance in Apositive controls compared to the A-negative (Chetelat et al., 2010b). In MCI patients, there is a more general agreement for a link between high Aburden and lower episodic memory performance (Mormino et al., 2009; Ong et al., 2013; Pike et al., 2007). The inconsistency in the findings of the relationship between Aand cognition might reflect several, sometimes convergent, sometimes divergent, facts i) Aimaging measures the aggregated fibrillar form of Awhile it has been postulated that only the soluble form of Ais toxic; ii) it is the presence but not the quantity of A deposition that matters; iii) fibrillar A is not directly causing cognitive impairment, but indirectly through other pathological processes, such as tau, that in turn drives neurodegeneration (f.e., A-related cognitive impairment was shown to be mediated by hippocampal atrophy in MCI patients (Mormino et al., 2009; Villeneuve et al., 2014; Wirth et al., 2013). Moreover, iv) cognitive reserve has been shown to impact not only on Adeposition but also on its association to cognition or neuronal injury (Rentz et al., 2010; Roe et al., 2008). Finally, it is also possible that v) the delay between A accumulation (which might start more than 15 years before the onset of dementia) and cognitive deficits/neurodegeneration (which generally occur later) prevents the detection of a link between these measures in cross-sectional studies. At the AD stage, there seems to be no longer correlation between glucose metabolism, cognition and  burden (Furst et al., 2010). This might reflect the fact that Ahas almost reach a plateau, or that the dose-dependent relationship of A to cognition and neurodegeneration only occurs in the early stage of amyloidosis (see also (Chetelat et al., 2010a)). While the results of cross-sectional studies relating A and cognition are discrepant, findings are more consistent in longitudinal studies assessing the effects of A deposition and later

8

change on cognitive decline and/or disease progression. Thus, high 11C-PiB cortical retention in non-demented elderly subjects is consistently associated with a greater risk of cognitive decline (Resnick et al., 2010; Villemagne et al., 2011b; Villemagne et al., 2008b). For example, the evaluation of either the cognitive trajectories (Resnick et al., 2010; Villemagne et al., 2008b) or changes in cognition prospectively (Doraiswamy et al., 2012b; Morris et al., 2009; Villemagne et al., 2011b) show that cognitively intact individuals with substantial  deposition are more likely to present with cognitive decline. Studies in MCI patients show that about 40-80% of MCI subjects with high

11

C-PiB retention progressed to AD over 2-3

years, being >10 times more likely to progress to AD than those with low A burden, highlighting the clinical relevance and potential impact in patient management of Aβ imaging (Doraiswamy et al., 2012a; Doraiswamy et al., 2014; Forsberg et al., 2008; Okello et al., 2009b; Ong et al., 2015; Rowe et al., 2013; Villemagne et al., 2011b; Wolk et al., 2009). 4. RELATION OF A IMAGING WITH OTHER BIOMARKERS

Another rapidly growing area is the exploration of the potential association between A burden as measured by PET, and CSF biomarkers of A deposition or neuroimaging biomarkers of neurodegeneration (Albert et al., 2011; Blennow et al., 2007; Clark et al., 2008; de Leon et al., 2006; Jack et al., 2010; McKhann et al., 2011; Morris et al., 2005; Shaw et al., 2007; Storandt et al., 2012; Sunderland et al., 2005; Thal et al., 2006; Wahlund and Blennow, 2003)(Jack et al., 2010; Jack et al., 2008a).

4.1

CSF

Analysis of CSF allows simultaneous measurement of the concentration of the two main hallmarks of AD, A and tau. It has been reported to be highly accurate in the diagnosis of AD as well of predicting cognitive decline (Blennow et al., 2007; de Leon et al., 2007; Hansson et al., 2007; Mattsson et al., 2011; Mattsson et al., 2009). The typical CSF profile in 9

AD is low A1-42 and high total (t-tau) and phosphorylated tau (p-tau) (Hampel et al., 2010; Strozyk et al., 2003). Slower A clearance from the brain (Bateman et al., 2006; Mawuenyega et al., 2010) is likely to lead to A deposition in the brain and lower CSF A concentrations. While there is no correlation between

11

C-PiB retention and CSF t-tau or p-

tau (Forsberg et al., 2010; Tolboom et al., 2009), several studies have reported a strong inverse correlation between A deposition in the brain as measured by 11C-PiB and CSF A142

(Fagan et al., 2006; Fagan et al., 2009; Fagan et al., 2007; Forsberg et al., 2008; Grimmer et

al., 2009; Koivunen et al., 2008a). Overall, there is about an 88% agreement between PIBPET and CSF in classifying cases as positive or negative, agreement that is higher (94%) in demented patients. Longitudinal A imaging and CSF studies have shown than while A accumulation in the brain followed a sigmoidal trajectory (Jack et al., 2013b; Villemagne et al., 2013), decreases in CSF A1-42 did not (Toledo et al., 2013). Moreover, a positive CSF in the absence of Adetected at PET is more frequent than the reverse. This likely reflects the fact that each technique is measuring a different pool of A(Toledo et al., 2015). While A imaging measures an insoluble fibrillar form of A, CSF measures reflect a more soluble diffusible form of A suggesting that changes in soluble A occur first (Toledo et al., 2013).

4.2

MRI

Hippocampal and cortical grey matter atrophy along with ventricular enlargement are typical MRI findings in AD and MCI (Drago et al., 2011; Frisoni et al., 2010). Furthermore, it has been shown that the rates of hippocampal atrophy might be predictive of conversion from MCI to AD (Jack et al., 2008b). The relationships between  deposition measured with PET and atrophy measured with MRI appears to be more complex and intricate. In AD, no relationship is usually found between deposition and atrophy (La Joie et al., 2012), and no association has been observed between hippocampal A load and hippocampal atrophy at any stage of the disease 10

spectrum. When comparing A-negative to A-positive non-demented individuals, results are rather discrepant with studies reporting hippocampal atrophy in the elderly with high global PiB (Jack et al., 2008a; Storandt et al., 2009) while others did not (Bourgeat et al., 2010; Dickerson et al., 2009). Similarly, significant correlations between global neocortical PiB and hippocampal atrophy has been reported in normal controls and MCI patients in some studies (Bourgeat et al., 2010; Mormino et al., 2009; Rowe et al., 2010), but not in others (Bourgeat et al., 2010; Dickerson et al., 2009). Moreover, between-group differences or correlations were inconsistently found in other brain areas, e.g. in the temporal pole (Dickerson et al., 2009) or anterior and posterior cingulate cortex (Becker et al., 2011; Storandt et al., 2009) or prefrontal and lateral parietal cortex (Becker et al., 2011). Interestingly, when assessing the links between regional A deposition and local atrophy (voxelwise), a significant relationship was found in the posterior cingulate-precuneus and anterior cingulate-medial prefrontal areas (with a trend in the temporo-parietal area), i.e. in the regions of highest  deposition (Chetelat et al., 2010a). This relationship was only found in elderly with subjective cognitive impairment but not in the MCI or in the AD patients, suggesting that this relationship occurs at a very particular stage of the disease, before objective memory impairment can be demonstrated. Also, a reverse relationships (larger temporal lobe volume in individuals with high A deposition) was found in the cognitively intact elderly, which might reflect brain reserve (Chetelat et al., 2010b). As mentioned above, education was found to modify the relation of plaques to cognition so that highly educated subjects have less susceptibility to -related cognitive impairment than those with lower education (Bennett et al., 2003; Rentz et al., 2010). It is thus possible that the high-PiB healthy controls represent those subjects with particularly high brain reserve reflected by larger brain (temporal) volume allowing them to better and/or longer tolerate the presence of A deposition. Moreover, the relationships between deposition and neurodegeneration might be further complicated by a different susceptibility/vulnerability to  at either a cellular - frontal neurons seems to be more 11

resistant to  than hippocampal neurons - (Resende et al., 2007; Roder et al., 2003), and/or by a regional compensatory upregulation of choline acetyltranserase (ChAT) activity in the frontal cortex and hippocampi of MCI and early AD subjects (DeKosky et al., 2002).

4.3

FDG

In FDG studies the typical „AD pattern‟ is of temporoparietal and posterior cingulate hypometabolism with sparing of the basal ganglia, thalamus, cerebellum, and primary sensorimotor cortex (Coleman, 2005; Devanand et al., 1997; Jagust et al., 2007; Salmon et al., 1994). Due to its high sensitivity (>90%) for detecting temporoparietal and posterior cingulate hypometabolism FDG-PET has improved diagnostic and prognostic accuracy in patients with probable AD (Kennedy et al., 1995a; Salmon et al., 1994; Silverman et al., 2002; Silverman et al., 2001; Small et al., 1995). A similar pattern of hypometabolism has been reported in MCI, (Chetelat et al., 2003; Chetelat et al., 2005; Mosconi et al., 2006a) asymptomatic subjects with mutations associated with familial AD, (Kennedy et al., 1995b; Rossor et al., 1996) and in subjects with a strong family history of AD (Mosconi et al., 2006b). FDG hypometabolism is correlated with cognition (Furst et al., 2010; Landau et al., 2009) and is predictive of future cognitive decline (Drzezga et al., 2005; Drzezga et al., 2003; Mosconi et al., 2004).

Overall, results regarding the relationships between FDG and A are similar as those between atrophy and A. Thus, no relationship is usually found in AD (La Joie et al., 2012) and discrepant findings are reported in non-demented individuals. Some studies found a significant link between PIB and local metabolism in the parietal and temporal cortex (Cohen et al., 2009; Edison et al., 2007; Klunk et al., 2004) but other studies reported no correlation in any brain region (Forsberg et al., 2008; Furst et al., 2010). In a recent report in a large sample of controls, MCI and AD assessing regional associations between A and hypometabolism, the authors concluded that regional fibrillar A deposition has little to no 12

association with regional hypometabolism showing that correlations were rather negative and low (or inexistent) in AD and healthy controls, but mostly positive in MCI patients (Altmann et al., 2015). As mentioned before for the temporal lobe volume (Chetelat et al., 2010b), this finding suggests that higher basal metabolism could reflect a reserve mechanism allowing to tolerate more A deposition while not showing cognitive impairment.

4.4

Neuroinflammation

Microglia are the primary resident immune surveillance cells in the brain and are thought to play a significant role in the pathogenesis of several neurodegenerative disorders including AD (Venneti et al., 2006). Activated microglia can be measured using radioligands for the translocator protein (TSPO), formerly known as the peripheral benzodiazepine receptor. The TSPO is upregulated on activated macrophages and microglia, and has been established as a biomarker of neuroinflammation in the central nervous system. Most reports found that in AD there is no association between A deposition and microglial activation measured by [11C]PK11195 (Edison et al., 2008a; Okello et al., 2009a; Wiley et al., 2009), while there is an association between [11C]PK11195 binding and both glucose hypometabolism and severity of dementia (Edison et al., 2008a; Yokokura et al., 2011). Another study used the MAO-B inhibitor L-deprenyl as a PET marker for astrocytosis, together with PIB to measure A, but no regional correlations were found between both PET tracers (Carter et al., 2012).

On hypotheses and facts (and their interpretation) Different hypotheses have been postulated on the sequence of biomarker changes leading to AD dementia (Jack et al., 2013a; Teipel et al., 2015) The dominant view of the physiopathological mechanisms underlying AD is that Aβ triggers a cascade of events that eventually leads to neurofibrillary tangle formation, followed by synaptic and neuronal injury and finally cognitive decline (Hardy, 1992; Hardy and Selkoe, 2002). Translating this

13

theoretical model in a biomarker model, it has been proposed that in preclinical AD,  deposition appears first, followed by biomarkers of neuronal injury (including FDG-PET hypometabolism and/or hippocampal atrophy), and then cognitive decline (Jack et al., 2013a; Jack et al., 2010). This biomarker model has been translated in criteria for preclinical AD with three stages corresponding to Aβ (CSF or PET) only (stage 1, A+N-), Aβ plus neuronal injury (stage 2, A+N+) and Aβ, neuronal injury and subtle cognitive change (stage 3) (Sperling et al., 2011a). In support of this Aβ-centric hypothesis we observe that: i) all known forms of autosomal-dominant AD involve genes related with the production of Aβ; ii)  deposition is detected with PET in normal individuals with no evidence of cognitive impairment or neuronal injury (Chetelat et al., 2013; Jack et al., 2012); iii)  deposition has been shown to be a slow and protracted process that starts >20 years before the onset of dementia (Jack et al., 2013b; Villain et al., 2012; Villemagne et al., 2013); iv) preclinical  deposition is associated with a higher rate of atrophy and conversion to MCI and AD (Chetelat et al., 2013; Chetelat et al., 2012; Morris et al., 2009).

However, the repeated and dismal record of therapeutic trials failures (Cummings et al., 2014; Doody et al., 2014; Doody et al., 2013; Salloway et al., 2014) and more specifically the lack of positive effects of anti- therapy on neuronal injury and/or cognitive markers casts serious doubts not only on the alleged efficacy of the therapeutic agents but also on Aβ as the single driver of the disease process. The other main counterargument have risen from neuropathology studies who have demonstrated that “tau comes first”, that tau pathology is found in the brain of cognitively normal elderly devoid of Aβ, and even reported evidence for tau pathology in children and young adults, decades before Aβ deposition (Braak and Braak, 1991, 1997; Braak and Del Tredici, 2011; Duyckaerts, 2011). To harmonize the biomarker model with the neuropathological evidence, a modified version of the model has been proposed where tau pathology in the mesial temporal cortex appears before Aβ, but at a 14

subthreshold level, not yet detectable with biomarkers (Jack et al., 2013a). According to this updated model, Aβ appears later but is detectable first, so the proposed sequence of biomarkers remains unchanged. However, several lines of evidence from neuroimaging studies are seen as not consistent with this view. More specifically, the postulated sequence of “Aβ first” has been questioned. First, regional variation in the relative degree of atrophy, hypometabolism and Aβ deposition, and low relationships between Aβ and neurodegeneration markers rather support to the view that AD is a multi-causal disease with different possible pathways and sequences of events (La Joie et al., 2012). Second, some cognitively normal elderly have evidence of neuronal injury without Aβ (A-N+). They have been classified as having “suspected non-AD pathophysiology” (SNAP) as they didn‟t fit with the postulated sequence of biomarker. As i) some of these patients will developed MCI and dementia (Knopman et al., 2012) while they don‟t present with more markers of non-AD pathophysiological processes (Knopman, 2013) ii) some A-N+ will convert to A+N+ and/or AD and iii) multimodal neuroimaging studies showed evidence for AD-like neurodegeneration independently from, or before, Aβ deposition (Altmann et al., 2015; Bateman et al., 2012; Jack et al., 2013c; Jagust et al., 2012; Reiman et al., 2012; Sheline et al., 2010), eventually, both Aβ-first and neurodegenerationfirst models of late-onset AD have been postulated (Chetelat, 2013; Jack and Holtzman, 2013). Another issue to be considered carefully is perithrehold values for both Aand neurodegeneration, that results in a classification that might not truly reflect the underlying pathological process(es). Yet, the debate remains on whether or not this AD-like neurodegeneration is part of the AD spectrum or not. As nicely phrased by Jack (Jack, 2014), “Some in the imaging/biomarker community argue that evidence of -amyloid deposition is required to label an individual as being in the “AD pathophysiological pathway” on the basis of biomarkers and hence SNAP is correctly labeled a non-AD condition. Others argue that because the neurodegenerative biomarkers in SNAP are AD-like, SNAP represents a “pre

15

fibrillar amyloid” part of the AD spectrum.” Still another position is to consider that, being labelled AD or not, the same should applied to both A+N- and A-N+, as there is no more reason to call preclinical AD the A+N- than the A-N+. Longitudinal studies will help clarify this issue.

This has led to the postulation of alternative models (Chetelat, 2013; Pimplikar et al., 2010; Small and Duff, 2008), re-opening intensive debates around the question of the role and relationships between tau and Aβ (Chetelat, 2013; Fox, 2012; Scheltens, 2013; Vishnu, 2013; Weiner, 2013). It has been proposed for instance that both cortical Aβ and mesial temporal tau-related pathological processes may occur independently of each other (Chetelat, 2013; Duyckaerts, 2011) and that, under the influence of environmental, genetic or yet unknown factors, they interact to promote the pathological processes leading to AD dementia (Chetelat, 2013). These models postulate that all neuroimaging biomarkers (atrophy, hypometablism, A deposition) should be considered as carrying the same weight, independently from the sequence they appear, where the addition of each biomarker is reflected in an incremental risk of progression to AD.

Neuroimaging (and especially A neuroimaging) offers an unique opportunity to test in vivo and longitudinally many of our hypotheses that could hardly be tested before. Yet, we have to remember that we are measuring macroscopic events detectable with neuroimaging techniques that are all limited by a sensitivity threshold and some measures that are just the end product of a biological/pathological process, not the biological process per se. Moreover, most of the results are amenable to multiple, even contradictory interpretations, where the same reports are cited in support of different hypotheses. So we need to be cautious.

16

Yet, encompassing these two different broad perspectives on AD physiopathology and biomarkers sequence, the following sequence proposed by Jack (Jack, 2014) seems to cover all aspects of what is known –and not yet known- to date: 1.

Most often before any significant A deposition, tau starts accumulating in the mesial

temporal cortex. Everyone in the population develops tau deposition at some point early in life. By itself, however, tau deposition in the mesial temporal cortex produces none to mild clinical symptomatology. 2.

Independently from tau accumulation in the mesial temporal cortex, A starts

accumulating in neocortical areas. 3.

At some point in time, A and tau interact, through some yet undetermined

mechanism(s), triggering the spreading of tauopathy from mesial temporal to widespread neocortical association areas that parallels progression of cognitive decline towards dementia. 4.

Progression of neurodegeneration and severe clinical symptoms reflect the accelerated

and expanding neocortical tauopathy, independently from the degree of A deposition.

Ongoing longitudinal studies will help identify those subjects that despite substantial A deposition do no develop the AD phenotype, thus allowing to isolate genetic or environmental factors that may convey resistance to A (Sojkova and Resnick, 2011). Perhaps selective tau imaging will help elucidate or bridge the gap between A deposition and the persistent decline in cognitive function observed in AD (Johnson et al., 2015; Villemagne et al., 2012). Preliminary A and tau imaging studies suggest that high cortical A and high hippocampal tau burden can be present in cognitively unimpaired individuals, suggesting that significant cognitive impairment is only observed when tau spreads into neocortical association areas (Delacourte et al., 2002; Villemagne et al., 2014).

17

5. A IMAGING IN OTHER NEURODEGENERATIVE CONDITIONS

A imaging in a wide spectrum of neurodegenerative conditions has allowed the assessment of the presence or absence in these different conditions. Although slightly lower than in AD, similar patterns of

11

C-PiB retention are usually observed in DLB (Gomperts et al., 2008;

Maetzler et al., 2009; Rowe et al., 2007). Cortical 11C-PiB retention, specifically in occipital areas, is also higher in subjects diagnosed with CAA (Johnson et al., 2007), while there is usually no cortical

11

C-PiB retention in patients with FTLD (Drzezga et al., 2008; Engler et

al., 2008; Rabinovici et al., 2007; Rowe et al., 2007).

8.1

Cerebral Amyloid Angiopathy

CAA, characterized by Aβ deposits in and around the media of small arteries and arterioles of the cerebral cortex and leptomeninges has been found to be present in most patients with AD (Jellinger and Attems, 2005). To this date, neuropathologic examination of the brain remains the only definitive method for diagnostic confirmation of CAA. However the combination of A imaging with 11C-PiB-PET and T2* susceptibility weighted (SWI) MR imaging (Knudsen et al., 2001) allows for the concomitant assessment of molecular and structural changes invivo. The combination of cerebral microhaemorrhages (MH) and superficial siderosis (SS) have been suggested as radiological markers for CAA (Feldman et al., 2008). A imaging studies have shown that both processes may be intimately associated with A deposition (Dhollander et al., 2011; Dierksen et al., 2010), preferentially in posterior areas of the brain, showing a distinct pattern of

11

C-PiB retention than the one observed in sporadic AD

(Johnson et al., 2007). Furthermore, focal A deposition might be useful in predicting new MH in CAA patients (Gurol et al., 2012). While deep subcortical MH are generally associated with vascular risk factors (VRF), lobar MH (particularly posterior) (Pettersen et al., 2008), are usually attributed to CAA (Knudsen et al., 2001; Vernooij et al., 2008). Lobar MH are a 18

frequent finding in AD patients or even in cognitively-normal older individuals, and they are strongly associated with increasing age and Aβ deposition (Yates et al., 2011). This association between A and vascular lesions has crucial implications not only for the selection and risk stratification of individuals undergoing anticoagulant therapies, but also in those enrolled in anti-A therapeutic trials (Weller et al., 2009).

8.2

Lewy Body Diseases

While AD is the most common cause of dementia in the elderly, DLB accounts for 20% of the cases (McKeith et al., 2005). The pathological hallmark of DLB is the presence of synuclein containing Lewy bodies (LB) within the neocortical and limbic regions, (McKeith et al., 2005) as well as substantial loss of pigmented dopaminergic neurons in the substantia nigra, reflected in a marked dopaminergic terminal denevation in the striatum (McKeith and Mosimann, 2004). More refined neuropathological techniques have revealed extensive synaptic deposits of -synuclein, more concordant with the DLB clinical phenotype than the usually sparse cortical LB (Schulz-Schaeffer, 2010). The overlap of cognitive symptoms early in the disease course of both AD and DLB present a challenge for clinicians that might find the differential diagnosis difficult to make. Post mortem histopathological studies have identified that 50-80% of DLB cases often have cortical A deposits with characteristics and a distribution similar to AD patients (Edison et al., 2008b; McKeith et al., 2005; Rowe et al., 2007). This has also been described as „mixed‟ DLB/AD or „Lewy body variant of AD‟. Socalled „pure‟ DLB is much less common and it is not clear if the clinical characteristics and prognosis differ from the mixed-pathology cases. On visual inspection, cortical

11

C-PiB

retention in DLB is generally lower and more variable than in AD, probably reflecting a larger spectrum of  deposition (Gomperts et al., 2008; Maetzler et al., 2009; Rowe et al., 2007). Therefore A imaging is not a useful tool in differentiating between AD and DLB, but

19

might assist in the differential diagnosis of those cases („pure‟ DLB) without A deposition in the brain (Armstrong et al., 2000). The contribution of A to the development of Lewy body diseases remains unclear, but cortical A deposits are associated with extensive -synuclein lesions and higher levels of insoluble -synuclein lesions (Pletnikova et al., 2005), as well as exacerbation of neuronal injury (Masliah et al., 2001). While A imaging cannot contribute to the differential diagnosis between AD and DLB, it may have prognostic relevance in these Lewy body diseases. For example,  burden in DLB patients was inversely correlated with the interval from onset of cognitive impairment to the full development of the DLB phenotype (Rowe et al., 2007).

8.3

Frontotemporal Lobar Degeneration

FTLD is a syndrome that can also be clinically difficult to distinguish from early onset AD, especially at the initial stages of the disease. Based on clinical phenotype, FTLD has been categorised mainly into two classes: behavioural variant of FTLD (bvFTLD) with distinct changes in behaviour and personality with little effect on language functions, and progressive aphasias (comprised by semantic dementia (SD), progressive non-fluent aphasia (PNFA), and logopenic aphasia (LPA)) showing progressive language deficits, with less obvious personality and behavioural changes (Rabinovici and Miller, 2010).

A deposition is not a pathological trait of FTLD, and definitive differential diagnosis can only be established after post mortem examination of the human brain. Neuropathological examination of the brain in FTLD shows variable frontal and temporal atrophy (Cairns et al., 2007; Hodges and Patterson, 2007; Snowden et al., 2007), microvacuolation and neuronal loss, with white matter myelin loss, astrocytic gliosis as well as neuronal and glial inclusions (Cairns et al., 2007). Three intraneuronal inclusion types have been identified in FTLD: (i) hyperphosphorylated tau (FTLD-tau), (ii) ubiquitin-hyperphosphorylated and proteolysed 20

TAR-DNA binding protein 43 (FTLD-TDP-43) and (iii) fused sarcoma positive (FUS) (Mott et al., 2005; Neumann et al., 2009; Neumann et al., 2006; Taniguchi et al., 2004). Taupositive inclusions account for approximately 40% of all FTLD cases, the remainder being tau negative and TDP-43/ubiquitin positive (Josephs et al., 2004). In bvFTLD, both tau and TDP-43 are equally prevalent; and while 90% of the SD cases show TDP-43 pathology, PNFA presents predominantly (70%) tau pathology (Josephs et al., 2004; Mackenzie et al., 2008). In contrast, LPA is though to be a language presentation of AD, with typical A and NFT (Leyton et al., 2011).

A imaging has been helpful in the differential diagnosis between FTLD and AD (Drzezga et al., 2008; Engler et al., 2008; Rabinovici et al., 2007; Rowe et al., 2007). Furthermore, A imaging has been used to ascertain the absence of AD pathology in the different FTLD aphasias (Drzezga et al., 2008; Leyton et al., 2011; Rabinovici et al., 2008). Despite displaying similar specificities, the diagnostic performance of A imaging proved to be more sensitive for the diagnosis of FTLD than FDG (Rabinovici et al., 2011).

8.4

Prion Diseases

No cortical 11C-PiB retention was observed in sporadic Creutzfeldt-Jakob disease (CJD) cases (Villemagne et al., 2009b). Due to the rapid progression and relatively short duration of symptomatic illness, sporadic CJD patients do not demonstrate appreciable PrP-plaque deposition at autopsy (Liberski, 2004). Although some sporadic CJD molecular sub-types can manifest larger PrP plaques (Hill et al., 2003), the most common subtype usually demonstrates only very fine “synaptic” and small perivacuolar deposits (Hill et al., 2003; Liberski, 2004). A preliminary study in transmissible spongiform encephalopathies showed that A imaging with 11C-BF227 might be able to distinguish between Gerstmann-Straussler-

21

Scheinker disease (GSS) and sporadic CJD disease with clear tracer retention in brain regions of PrP deposition (Okamura et al., 2010).

6. A

IMAGING

IN

THE

DEVELOPMENT

OF

DISEASE-SPECIFIC

THERAPEUTICS

Aβ imaging with PET is also contributing to the development of more effective therapies by allowing better selection of patients for anti-A therapy trials and providing a means to measure their impact on A burden (Ostrowitzki et al., 2011; Rinne et al., 2010). While these studies represent one of the principal applications of A imaging, they have brought forward an issue that needs to be addressed, namely that the advent of novel A tracers with differing pharmacological and pharmacokinetics properties, summed to the diverse arsenal of quantitative approaches and contrasting criteria for the selection of internal scaling region pose a challenge when trying to compare results from therapeutic trials that use different Atracers or quantitative tools.

Another aspect to be considered is prediction of therapeutic response. As new therapies enter clinical trials, the role of A imaging in vivo is becoming increasingly crucial.  imaging allows the in vivo assessment of brain  pathology and its changes over time, providing highly accurate, reliable, and reproducible quantitative statements of regional or global A burden in the brain. As mentioned before, therapies, especially those targeting irreversible neurodegenerative processes, have a better chance to succeed if applied early, making early detection of the underlying pathological process so important. Therefore, Aβ imaging is an ideal tool for the selection of adequate candidates for anti-Aβ therapeutic trials, while also monitoring -and potentially predicting- treatment response, thus aiding in reducing sample size, minimizing cost while maximizing outcomes. Preliminary studies are already showing 22

the utility of this technique (Ostrowitzki et al., 2011; Rinne et al., 2010). These preliminary results, taken together with the results from longitudinal studies, support the growing consensus that anti-Aβ therapy to be effective may need to be given early in the course of the disease, perhaps even before symptoms appear, (Sperling et al., 2011b) and that downstream mechanisms may also need to be addressed to successfully prevent the development of AD. Furthermore, the slow rate of A deposition indicates that the time window for altering A accumulation prior to the full manifestation of the clinical phenotype may be very wide (Villemagne et al., 2011b).

The contribution of Aβ imaging to the selection of patients for therapeutic trials needs to be complemented with adequate MRI sequences such as SWI or gradient recalled echo (GRE) for the detection of vascular pathology to rule out the possibility of vascular complications such as  related imaging abnormalities (ARIA), either as "vasogenic edema" and/or sulcal effusion (ARIA-E), or as hemosiderin deposits including MH and SS (ARIA-H) (Cordonnier, 2010; Sperling et al., 2011c). The presence of MH is impacting on patient selection in therapeutic trials, and some anti-Aβ immunotherapy trials are already excluding individuals with MH. To make matters worse, about one third of non-demented individuals with high Aβ burden and no prior history of cerebrovascular disease showed MH (Yates et al., 2011). While focal Aβ deposition might be a better predictor of a vascular event (Gurol et al., 2012), only longitudinal follow up of large cohorts of patients will allow elucidation of the relevant risk factors for lobar MH.

Finally, and given that there is no cure for AD, diagnosing the presence or absence of AD pathology in vivo raises ethical issues. While the presence of  deposition has undoubtedly been associated with a significantly greater risk of cognitive decline, there is still not enough longitudinal data available to attach a firm prognostic clinical significance to a “positive”

23

scan. This is especially true in cognitively normal elderly. In this situation, is it ethical to disclose the  scan result while it is not associated with a diagnosis so that it is an information on the risk of AD but not on the diagnosis? Also, is it ethical to deliver this information while there is no treatment that has proven efficacy against AD pathological process yet? Of course, this is useful for anti- clinical trials that are on-going, but these are still on the development/research stages. From the patient perspective, it is important to take into account their right to know, as well as their right not to know. More work is needed to evaluate the psychological impact of  PET result disclosure and determine the right way to disclose this information, as well as to further anticipate related societal and legislative impacts.

7. CODA

The clinical diagnosis of AD is typically based on progressive cognitive impairments whilst excluding other diseases. Clinical diagnosis of sporadic disease is challenging, however, often presenting mild and non-specific symptoms attributable to diverse and overlapping pathology presenting similar phenotypes. Overall, the accuracy of clinical diagnosis of AD compared to neuropathological examination ranges between 70-90% (Beach et al., 2012). While clinical criteria, together with current structural neuroimaging techniques (CT or MRI), are sufficiently sensitive and specific for the diagnosis of AD at the mid to late stages of the disease, they are focused on relatively non-specific features such as memory and functional activity decline or brain atrophy. Confirmation of diagnosis of AD still relies on autopsy. But how reliable is pathology? How much of a “gold standard” is autopsy? (Jellinger, 2010; Scheltens and Rockwood, 2011). A more sensible paradigm, adopting a more fluid model that integrates all cognitive, biochemical and imaging biomarkers that can provide a better predictive framework, has been proposed in the new diagnostic criteria for AD, (Dubois et al.,

24

2010; McKhann et al., 2011; Sperling and Johnson, 2010) MCI, (Albert et al., 2011) and preclinical AD (Sperling et al., 2011a). Along the same lines, Scheltens and Rockwood suggest that “a shift in focus from why people with AD have plaques and tangles to why not all people with plaques and tangles have AD would be welcome” (Scheltens and Rockwood, 2011). While most people with AD pathology develop AD dementia, there are some that while having the pathology do not. What makes them different? What conveys resistance to some or, conversely, what makes others more vulnerable to the same pathological burden? Identifying these factors will allow a more discerning selection of individuals that will benefit from early intervention with anti-A medication, while also providing insights into potential preventive strategies or even new therapeutic approaches (Sojkova and Resnick, 2011; Sperling et al., 2011b). While the new diagnostic criteria has blurred once again the boundaries between MCI and AD (Morris, 2012), it also allows for earlier diagnosis and therapeutic intervention, arguing that when there is a clear history of progressive cognitive decline, objective evidence from psychometric tests of episodic memory impairment and characteristic abnormalities in the CSF and/or neuroimaging studies such as A imaging, dementia might not be required for the diagnosis of probable AD (Dubois et al., 2010; McKhann et al., 2011; Sperling and Johnson, 2010). Thus, as the criteria for the diagnosis of AD changes, A imaging is likely to play an increasingly important role in clinical practice provided it is accessible and affordable (Villemagne and Rowe, 2010).

The pathological process usually begins decades before symptoms are evident, making it difficult to identify individuals at the earliest stage of pathology. This in turn precludes early intervention with disease-modifying medications during the presymptomatic period, which by arresting neuronal loss would presumably achieve the maximum benefits of such therapies (Villemagne et al., 2008a). Therefore, a change in the diagnostic paradigm is warranted, where diagnosis moves away from identification of signs and symptoms of neuronal failure –

25

indicating that central compensatory mechanisms have been exhausted and extensive synaptic and neuronal damage is present – to the non-invasive detection of specific biomarkers for particular traits underlying the pathological process (Clark et al., 2008). Given the complexity and sometimes overlapping characteristics of these disorders, and despite recent advances in molecular neurosciences, it is unlikely that a single biomarker will be able to provide the diagnostic certainty required for the early detection of neurodegenerative diseases like AD. Therefore, a multimodality approach combining biochemical and neuroimaging methods is warranted (Shaw et al., 2007).

ACKNOWLEDGEMENTS This work was supported in part by grant 1071430 of the National Health and Medical Research Council (NHMRC) of Australia. VLV is supported by the NHMRC through a Senior Research Fellowship. The Institut National de la Santé et de la Recherche Médicale (Inserm), the Fondation Plan Alzheimer (Alzheimer Plan 2008-2012), Programme Hospitalier de Recherche Clinique (PHRC National 2011, complément PHRC 2012), Agence Nationale de la Recherche (ANR LONGVIE 2007), and Région Basse Normandie also supported this work.

26

REFERENCES Aizenstein, H.J., Nebes, R.D., Saxton, J.A., Price, J.C., Mathis, C.A., Tsopelas, N.D., Ziolko, S.K., James, J.A., Snitz, B.E., Houck, P.R., Bi, W., Cohen, A.D., Lopresti, B.J., Dekosky, S.T., Halligan, E.M., Klunk, W.E., 2008. Frequent amyloid deposition without significant cognitive impairment among the elderly. Arch Neurol 65, 1509-1517. Albert, M.S., Dekosky, S.T., Dickson, D., Dubois, B., Feldman, H.H., Fox, N.C., Gamst, A., Holtzman, D.M., Jagust, W.J., Petersen, R.C., Snyder, P.J., Carrillo, M.C., Thies, B., Phelps, C.H., 2011. The diagnosis of mild cognitive impairment due to Alzheimer's disease: Recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 7, 270-279. Altmann, A., Ng, B., Landau, S.M., Jagust, W.J., Greicius, M.D., Alzheimer's Disease Neuroimaging, I., 2015. Regional brain hypometabolism is unrelated to regional amyloid plaque burden. Brain. Armstrong, R.A., Cairns, N.J., Lantos, P.L., 2000. Beta-amyloid deposition in the temporal lobe of patients with dementia with Lewy bodies: comparison with non-demented cases and Alzheimer's disease. Dement Geriatr Cogn Disord 11, 187-192. Barthel, H., Gertz, H.J., Dresel, S., Peters, O., Bartenstein, P., Buerger, K., Hiemeyer, F., Wittemer-Rump, S.M., Seibyl, J., Reininger, C., Sabri, O., 2011. Cerebral amyloid-beta PET with florbetaben ((18)F) in patients with Alzheimer's disease and healthy controls: a multicentre phase 2 diagnostic study. Lancet Neurol 10, 424-435. Bateman, R.J., Munsell, L.Y., Morris, J.C., Swarm, R., Yarasheski, K.E., Holtzman, D.M., 2006. Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nat Med 12, 856-861. Bateman, R.J., Xiong, C., Benzinger, T.L., Fagan, A.M., Goate, A., Fox, N.C., Marcus, D.S., Cairns, N.J., Xie, X., Blazey, T.M., Holtzman, D.M., Santacruz, A., Buckles, V., Oliver, A., Moulder, K., Aisen, P.S., Ghetti, B., Klunk, W.E., McDade, E., Martins, R.N., Masters, C.L., 27

Mayeux, R., Ringman, J.M., Rossor, M.N., Schofield, P.R., Sperling, R.A., Salloway, S., Morris, J.C., 2012. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N Engl J Med 367, 795-804. Beach, T.G., Monsell, S.E., Phillips, L.E., Kukull, W., 2012. Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005-2010. J Neuropathol Exp Neurol 71, 266-273. Blennow, K., Zetterberg, H., Minthon, L., Lannfelt, L., Strid, S., Annas, P., Basun, H., Andreasen, N., 2007. Longitudinal stability of CSF biomarkers in Alzheimer's disease. Neurosci Lett 419, 18-22. Bourgeat, P., Chetelat, G., Villemagne, V.L., Fripp, J., Raniga, P., Pike, K., Acosta, O., Szoeke, C., Ourselin, S., Ames, D., Ellis, K.A., Martins, R.N., Masters, C.L., Rowe, C.C., Salvado, O., 2010. Beta-amyloid burden in the temporal neocortex is related to hippocampal atrophy in elderly subjects without dementia. Neurology 74, 121-127. Braak, H., Braak, E., 1991. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol (Berl) 82, 239-259. Braak, H., Braak, E., 1997. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging 18, 351-357. Braak, H., Del Tredici, K., 2011. The pathological process underlying Alzheimer's disease in individuals under thirty. Acta Neuropathol 121, 171-181. Buckner, R.L., Andrews-Hanna, J.R., Schacter, D.L., 2008. The brain's default network: anatomy, function, and relevance to disease. Ann N Y Acad Sci 1124, 1-38. Buckner, R.L., Snyder, A.Z., Shannon, B.J., LaRossa, G., Sachs, R., Fotenos, A.F., Sheline, Y.I., Klunk, W.E., Mathis, C.A., Morris, J.C., Mintun, M.A., 2005. Molecular, structural, and functional characterization of Alzheimer's disease: evidence for a relationship between default activity, amyloid, and memory. J Neurosci 25, 7709-7717.

28

Cairns, N.J., Bigio, E.H., Mackenzie, I.R., Neumann, M., Lee, V.M., Hatanpaa, K.J., White, C.L., 3rd, Schneider, J.A., Grinberg, L.T., Halliday, G., Duyckaerts, C., Lowe, J.S., Holm, I.E., Tolnay, M., Okamoto, K., Yokoo, H., Murayama, S., Woulfe, J., Munoz, D.G., Dickson, D.W., Ince, P.G., Trojanowski, J.Q., Mann, D.M., 2007. Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol 114, 5-22. Camus, V., Payoux, P., Barre, L., Desgranges, B., Voisin, T., Tauber, C., La Joie, R., Tafani, M., Hommet, C., Chetelat, G., Mondon, K., de La Sayette, V., Cottier, J.P., Beaufils, E., Ribeiro, M.J., Gissot, V., Vierron, E., Vercouillie, J., Vellas, B., Eustache, F., Guilloteau, D., 2012. Using PET with (18)F-AV-45 (florbetapir) to quantify brain amyloid load in a clinical environment. Eur J Nucl Med Mol Imaging. Cappai, R., White, A.R., 1999. Amyloid beta. Int J Biochem Cell Biol 31, 885-889. Carter, S.F., Scholl, M., Almkvist, O., Wall, A., Engler, H., Langstrom, B., Nordberg, A., 2012. Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuteriumL-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18FFDG. J Nucl Med 53, 37-46. Chetelat, G., 2013. Alzheimer disease: Abeta-independent processes-rethinking preclinical AD. Nat Rev Neurol 9, 123-124. Chetelat, G., Desgranges, B., de la Sayette, V., Viader, F., Berkouk, K., Landeau, B., Lalevee, C., Le Doze, F., Dupuy, B., Hannequin, D., Baron, J.C., Eustache, F., 2003. Dissociating atrophy and hypometabolism impact on episodic memory in mild cognitive impairment. Brain 126, 1955-1967. Chetelat, G., Eustache, F., Viader, F., De La Sayette, V., Pelerin, A., Mezenge, F., Hannequin, D., Dupuy, B., Baron, J.C., Desgranges, B., 2005. FDG-PET measurement is more accurate than neuropsychological assessments to predict global cognitive deterioration in patients with mild cognitive impairment. Neurocase 11, 14-25.

29

Chetelat, G., La Joie, R., Villain, N., Perrotin, A., de La Sayette, V., Eustache, F., Vandenberghe, R., 2013. Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer's disease. NeuroImage. Clinical 2, 356-365. Chetelat, G., Villemagne, V.L., Bourgeat, P., Pike, K.E., Jones, G., Ames, D., Ellis, K.A., Szoeke, C., Martins, R.N., O'Keefe, G.J., Salvado, O., Masters, C.L., Rowe, C.C., 2010a. Relationship between atrophy and beta-amyloid deposition in Alzheimer disease. Ann Neurol 67, 317-324. Chetelat, G., Villemagne, V.L., Pike, K.E., Baron, J.C., Bourgeat, P., Jones, G., Faux, N.G., Ellis, K.A., Salvado, O., Szoeke, C., Martins, R.N., Ames, D., Masters, C.L., Rowe, C.C., 2010b. Larger temporal volume in elderly with high versus low beta-amyloid deposition. Brain 133, 3349-3358. Chetelat, G., Villemagne, V.L., Villain, N., Jones, G., Ellis, K.A., Ames, D., Martins, R.N., Masters, C.L., Rowe, C.C., 2012. Accelerated cortical atrophy in cognitively normal elderly with high beta-amyloid deposition. Neurology 78, 477-484. Clark, C.M., Davatzikos, C., Borthakur, A., Newberg, A., Leight, S., Lee, V.M., Trojanowski, J.Q., 2008. Biomarkers for early detection of Alzheimer pathology. Neurosignals 16, 11-18. Coleman, R.E., 2005. Positron emission tomography diagnosis of Alzheimer's disease. Neuroimaging Clin N Am 15, 837-846, x. Cordonnier, C., 2010. Brain microbleeds. Pract Neurol 10, 94-100. Cselenyi, Z., Jonhagen, M.E., Forsberg, A., Halldin, C., Julin, P., Schou, M., Johnstrom, P., Varnas, K., Svensson, S., Farde, L., 2012. Clinical Validation of 18F-AZD4694, an Amyloidbeta-Specific PET Radioligand. J Nucl Med 53, 415-424. Cummings, J.L., Morstorf, T., Zhong, K., 2014. Alzheimer's disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther 6, 37.

30

Cummings, J.L., Vinters, H.V., Cole, G.M., Khachaturian, Z.S., 1998. Alzheimer's disease: etiologies, pathophysiology, cognitive reserve, and treatment opportunities. Neurology 51, S2-17; discussion S65-17. Davies, L., Wolska, B., Hilbich, C., Multhaup, G., Martins, R., Simms, G., Beyreuther, K., Masters, C.L., 1988. A4 amyloid protein deposition and the diagnosis of Alzheimer's disease: prevalence in aged brains determined by immunocytochemistry compared with conventional neuropathologic techniques. Neurology 38, 1688-1693. de Leon, M.J., DeSanti, S., Zinkowski, R., Mehta, P.D., Pratico, D., Segal, S., Rusinek, H., Li, J., Tsui, W., Saint Louis, L.A., Clark, C.M., Tarshish, C., Li, Y., Lair, L., Javier, E., Rich, K., Lesbre, P., Mosconi, L., Reisberg, B., Sadowski, M., DeBernadis, J.F., Kerkman, D.J., Hampel, H., Wahlund, L.O., Davies, P., 2006. Longitudinal CSF and MRI biomarkers improve the diagnosis of mild cognitive impairment. Neurobiol Aging 27, 394-401. de Leon, M.J., Mosconi, L., Blennow, K., DeSanti, S., Zinkowski, R., Mehta, P.D., Pratico, D., Tsui, W., Saint Louis, L.A., Sobanska, L., Brys, M., Li, Y., Rich, K., Rinne, J., Rusinek, H., 2007. Imaging and CSF studies in the preclinical diagnosis of Alzheimer's disease. Ann N Y Acad Sci 1097, 114-145. DeKosky, S.T., Ikonomovic, M.D., Styren, S.D., Beckett, L., Wisniewski, S., Bennett, D.A., Cochran, E.J., Kordower, J.H., Mufson, E.J., 2002. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Ann Neurol 51, 145-155. Delacourte, A., Sergeant, N., Wattez, A., Maurage, C.A., Lebert, F., Pasquier, F., David, J.P., 2002. Tau aggregation in the hippocampal formation: an ageing or a pathological process? Exp Gerontol 37, 1291-1296. Devanand, D.P., Jacobs, D.M., Tang, M.X., Del Castillo-Castaneda, C., Sano, M., Marder, K., Bell, K., Bylsma, F.W., Brandt, J., Albert, M., Stern, Y., 1997. The course of

31

psychopathologic features in mild to moderate Alzheimer disease. Arch Gen Psychiatry 54, 257- 263. Dhollander, I., Nelissen, N., Van Laere, K., Peeters, D., Demaerel, P., Van Paesschen, W., Thijs, V., Vandenberghe, R., 2011. In vivo amyloid imaging in cortical superficial siderosis. J Neurol Neurosurg Psychiatry 82, 469-471. Dickerson, B.C., Bakkour, A., Salat, D.H., Feczko, E., Pacheco, J., Greve, D.N., Grodstein, F., Wright, C.I., Blacker, D., Rosas, H.D., Sperling, R.A., Atri, A., Growdon, J.H., Hyman, B.T., Morris, J.C., Fischl, B., Buckner, R.L., 2009. The cortical signature of Alzheimer's disease: regionally specific cortical thinning relates to symptom severity in very mild to mild AD dementia and is detectable in asymptomatic amyloid-positive individuals. Cereb Cortex 19, 497-510. Dierksen, G.A., Skehan, M.E., Khan, M.A., Jeng, J., Nandigam, R.N., Becker, J.A., Kumar, A., Neal, K.L., Betensky, R.A., Frosch, M.P., Rosand, J., Johnson, K.A., Viswanathan, A., Salat, D.H., Greenberg, S.M., 2010. Spatial relation between microbleeds and amyloid deposits in amyloid angiopathy. Ann Neurol 68, 545-548. Doody, R.S., Farlow, M., Aisen, P.S., Alzheimer's Disease Cooperative Study Data, A., Publication, C., 2014. Phase 3 trials of solanezumab and bapineuzumab for Alzheimer's disease. N Engl J Med 370, 1460. Doody, R.S., Raman, R., Farlow, M., Iwatsubo, T., Vellas, B., Joffe, S., Kieburtz, K., He, F., Sun, X., Thomas, R.G., Aisen, P.S., Alzheimer's Disease Cooperative Study Steering, C., Siemers, E., Sethuraman, G., Mohs, R., Semagacestat Study, G., 2013. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med 369, 341-350. Doraiswamy, P.M., Sperling, R.A., Coleman, R.E., Johnson, K.A., Reiman, E.M., Davis, M.D., Grundman, M., Sabbagh, M.N., Sadowsky, C.H., Fleisher, A.S., Carpenter, A., Clark, C.M., Joshi, A.D., Mintun, M.A., Skovronsky, D.M., Pontecorvo, M.J., 2012a. Amyloid-beta

32

assessed by florbetapir F 18 PET and 18-month cognitive decline: a multicenter study. Neurology 79, 1636-1644. Doraiswamy, P.M., Sperling, R.A., Coleman, R.E., Johnson, K.A., Reiman, E.M., Davis, M.D., Grundman, M., Sabbagh, M.N., Sadowsky, C.H., Fleisher, A.S., Carpenter, A., Clark, C.M., Joshi, A.D., Mintun, M.A., Skovronsky, D.M., Pontecorvo, M.J., 2012b. Amyloid-beta assessed by florbetapir F 18 PET and 18-month cognitive decline: A multicenter study. Neurology. Doraiswamy, P.M., Sperling, R.A., Johnson, K., Reiman, E.M., Wong, T.Z., Sabbagh, M.N., Sadowsky, C.H., Fleisher, A.S., Carpenter, A., Joshi, A.D., Lu, M., Grundman, M., Mintun, M.A., Skovronsky, D.M., Pontecorvo, M.J., Group, A.A.S., Group, A.A.S., 2014. Florbetapir F 18 amyloid PET and 36-month cognitive decline:a prospective multicenter study. Mol Psychiatry 19, 1044-1051. Drago, V., Babiloni, C., Bartres-Faz, D., Caroli, A., Bosch, B., Hensch, T., Didic, M., Klafki, H.W., Pievani, M., Jovicich, J., Venturi, L., Spitzer, P., Vecchio, F., Schoenknecht, P., Wiltfang, J., Redolfi, A., Forloni, G., Blin, O., Irving, E., Davis, C., Hardemark, H.G., Frisoni, G.B., 2011. Disease tracking markers for Alzheimer's disease at the prodromal (MCI) stage. J Alzheimers Dis 26 Suppl 3, 159-199. Drzezga, A., Grimmer, T., Henriksen, G., Stangier, I., Perneczky, R., Diehl-Schmid, J., Mathis, C.A., Klunk, W.E., Price, J., DeKosky, S., Wester, H.J., Schwaiger, M., Kurz, A., 2008. Imaging of amyloid plaques and cerebral glucose metabolism in semantic dementia and Alzheimer's disease. Neuroimage 39, 619-633. Drzezga, A., Grimmer, T., Riemenschneider, M., Lautenschlager, N., Siebner, H., Alexopoulus, P., Minoshima, S., Schwaiger, M., Kurz, A., 2005. Prediction of individual clinical outcome in MCI by means of genetic assessment and (18)F-FDG PET. J Nucl Med 46, 1625-1632.

33

Drzezga, A., Lautenschlager, N., Siebner, H., Riemenschneider, M., Willoch, F., Minoshima, S., Schwaiger, M., Kurz, A., 2003. Cerebral metabolic changes accompanying conversion of mild cognitive impairment into Alzheimer's disease: a PET follow-up study. Eur J Nucl Med Mol Imaging 30, 1104-1113. Dubois, B., Feldman, H.H., Jacova, C., Cummings, J.L., Dekosky, S.T., Barberger-Gateau, P., Delacourte, A., Frisoni, G., Fox, N.C., Galasko, D., Gauthier, S., Hampel, H., Jicha, G.A., Meguro, K., O'Brien, J., Pasquier, F., Robert, P., Rossor, M., Salloway, S., Sarazin, M., de Souza, L.C., Stern, Y., Visser, P.J., Scheltens, P., 2010. Revising the definition of Alzheimer's disease: a new lexicon. Lancet Neurol 9, 1118-1127. Duyckaerts, C., 2011. Tau pathology in children and young adults: can you still be unconditionally baptist? Acta Neuropathol 121, 145-147. Edison, P., Archer, H.A., Gerhard, A., Hinz, R., Pavese, N., Turkheimer, F.E., Hammers, A., Tai, Y.F., Fox, N., Kennedy, A., Rossor, M., Brooks, D.J., 2008a. Microglia, amyloid, and cognition in Alzheimer's disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol Dis 32, 412-419. Edison, P., Rowe, C.C., Rinne, J.O., Ng, S., Ahmed, I., Kemppainen, N., Villemagne, V.L., O'Keefe, G., Nagren, K., Chaudhury, K.R., Masters, C.L., Brooks, D.J., 2008b. Amyloid load in Parkinson's disease dementia and Lewy body dementia measured with [11C]PIB positron emission tomography. J Neurol Neurosurg Psychiatry 79, 1331-1338. Engler, H., Forsberg, A., Almkvist, O., Blomquist, G., Larsson, E., Savitcheva, I., Wall, A., Ringheim, A., Langstrom, B., Nordberg, A., 2006. Two-year follow-up of amyloid deposition in patients with Alzheimer's disease. Brain 129, 2856-2866. Engler, H., Santillo, A.F., Wang, S.X., Lindau, M., Savitcheva, I., Nordberg, A., Lannfelt, L., Langstrom, B., Kilander, L., 2008. In vivo amyloid imaging with PET in frontotemporal dementia. Eur J Nucl Med Mol Imaging 35, 100-106.

34

Fagan, A.M., Mintun, M.A., Mach, R.H., Lee, S.Y., Dence, C.S., Shah, A.R., Larossa, G.N., Spinner, M.L., Klunk, W.E., Mathis, C.A., Dekosky, S.T., Morris, J.C., Holtzman, D.M., 2006. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta(42) in humans. Ann Neurol 59, 512-519. Fagan, A.M., Mintun, M.A., Shah, A.R., Aldea, P., Roe, C.M., Mach, R.H., Marcus, D., Morris, J.C., Holtzman, D.M., 2009. Cerebrospinal fluid tau and ptau(181) increase with cortical amyloid deposition in cognitively normal individuals: implications for future clinical trials of Alzheimer's disease. EMBO Mol Med 1, 371-380. Fagan, A.M., Roe, C.M., Xiong, C., Mintun, M.A., Morris, J.C., Holtzman, D.M., 2007. Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol 64, 343-349. Feldman, H.H., Maia, L.F., Mackenzie, I.R., Forster, B.B., Martzke, J., Woolfenden, A., 2008. Superficial siderosis: a potential diagnostic marker of cerebral amyloid angiopathy in Alzheimer disease. Stroke 39, 2894-2897. Fleisher, A.S., Chen, K., Liu, X., Roontiva, A., Thiyyagura, P., Ayutyanont, N., Joshi, A.D., Clark, C.M., Mintun, M.A., Pontecorvo, M.J., Doraiswamy, P.M., Johnson, K.A., Skovronsky, D.M., Reiman, E.M., 2011. Using positron emission tomography and florbetapir F18 to image cortical amyloid in patients with mild cognitive impairment or dementia due to Alzheimer disease. Arch Neurol 68, 1404-1411. Forman, M.S., Mufson, E.J., Leurgans, S., Pratico, D., Joyce, S., Leight, S., Lee, V.M., Trojanowski, J.Q., 2007. Cortical biochemistry in MCI and Alzheimer disease: lack of correlation with clinical diagnosis. Neurology 68, 757-763. Forsberg, A., Almkvist, O., Engler, H., Wall, A., Langstrom, B., Nordberg, A., 2010. High PIB retention in Alzheimer's disease is an early event with complex relationship with CSF biomarkers and functional parameters. Curr Alzheimer Res 7, 56-66.

35

Forsberg, A., Engler, H., Almkvist, O., Blomquist, G., Hagman, G., Wall, A., Ringheim, A., Langstrom, B., Nordberg, A., 2008. PET imaging of amyloid deposition in patients with mild cognitive impairment. Neurobiol Aging 29, 1456-1465. Fox, N., 2012. When, where, and how does Alzheimer's disease start? Lancet Neurol 11, 1017-1018. Frisoni, G.B., Fox, N.C., Jack, C.R., Jr., Scheltens, P., Thompson, P.M., 2010. The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol 6, 67-77. Furst, A.J., Rabinovici, G.D., Rostomian, A.H., Steed, T., Alkalay, A., Racine, C., Miller, B.L., Jagust, W.J., 2010. Cognition, glucose metabolism and amyloid burden in Alzheimer's disease. Neurobiol Aging. Gomperts, S.N., Rentz, D.M., Moran, E., Becker, J.A., Locascio, J.J., Klunk, W.E., Mathis, C.A., Elmaleh, D.R., Shoup, T., Fischman, A.J., Hyman, B.T., Growdon, J.H., Johnson, K.A., 2008. Imaging amyloid deposition in Lewy body diseases. Neurology 71, 903-910. Grimmer, T., Riemenschneider, M., Forstl, H., Henriksen, G., Klunk, W.E., Mathis, C.A., Shiga, T., Wester, H.J., Kurz, A., Drzezga, A., 2009. Beta amyloid in Alzheimer's disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol Psychiatry 65, 927-934. Gurol, M.E., Dierksen, G., Betensky, R., Gidicsin, C., Halpin, A., Becker, A., Carmasin, J., Ayres, A., Schwab, K., Viswanathan, A., Salat, D., Rosand, J., Johnson, K.A., Greenberg, S.M., 2012. Predicting sites of new hemorrhage with amyloid imaging in cerebral amyloid angiopathy. Neurology 79, 320-326. Hampel, H., Blennow, K., Shaw, L.M., Hoessler, Y.C., Zetterberg, H., Trojanowski, J.Q., 2010. Total and phosphorylated tau protein as biological markers of Alzheimer's disease. Exp Gerontol 45, 30-40.

36

Hansson, O., Zetterberg, H., Buchhave, P., Andreasson, U., Londos, E., Minthon, L., Blennow, K., 2007. Prediction of Alzheimer's disease using the CSF Abeta42/Abeta40 ratio in patients with mild cognitive impairment. Dement Geriatr Cogn Disord 23, 316-320. Hardy, J., 1992. An 'anatomical cascade hypothesis' for Alzheimer's disease. Trends Neurosci 15, 200-201. Hardy, J., 1997. Amyloid, the presenilins and Alzheimer's disease. Trends Neurosci 20, 154159. Hardy, J., Selkoe, D.J., 2002. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297, 353-356. Hill, A.F., Joiner, S., Wadsworth, J.D., Sidle, K.C., Bell, J.E., Budka, H., Ironside, J.W., Collinge, J., 2003. Molecular classification of sporadic Creutzfeldt-Jakob disease. Brain 126, 1333-1346. Hodges, J.R., Patterson, K., 2007. Semantic dementia: a unique clinicopathological syndrome. Lancet Neurol 6, 1004-1014. Isacson, O., Seo, H., Lin, L., Albeck, D., Granholm, A.C., 2002. Alzheimer's disease and Down's syndrome: roles of APP, trophic factors and ACh. Trends Neurosci 25, 79-84. Jack, C.R., Jr., 2014. PART and SNAP. Acta Neuropathol 128, 773-776. Jack, C.R., Jr., Holtzman, D.M., 2013. Biomarker modeling of Alzheimer's disease. Neuron 80, 1347-1358. Jack, C.R., Jr., Knopman, D.S., Jagust, W.J., Petersen, R.C., Weiner, M.W., Aisen, P.S., Shaw, L.M., Vemuri, P., Wiste, H.J., Weigand, S.D., Lesnick, T.G., Pankratz, V.S., Donohue, M.C., Trojanowski, J.Q., 2013a. Tracking pathophysiological processes in Alzheimer's disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol 12, 207-216. Jack, C.R., Jr., Knopman, D.S., Jagust, W.J., Shaw, L.M., Aisen, P.S., Weiner, M.W., Petersen, R.C., Trojanowski, J.Q., 2010. Hypothetical model of dynamic biomarkers of the Alzheimer's pathological cascade. Lancet Neurol 9, 119-128.

37

Jack, C.R., Jr., Knopman, D.S., Weigand, S.D., Wiste, H.J., Vemuri, P., Lowe, V., Kantarci, K., Gunter, J.L., Senjem, M.L., Ivnik, R.J., Roberts, R.O., Rocca, W.A., Boeve, B.F., Petersen, R.C., 2012. An operational approach to National Institute on Aging-Alzheimer's Association criteria for preclinical Alzheimer disease. Ann Neurol 71, 765-775. Jack, C.R., Jr., Lowe, V.J., Senjem, M.L., Weigand, S.D., Kemp, B.J., Shiung, M.M., Knopman, D.S., Boeve, B.F., Klunk, W.E., Mathis, C.A., Petersen, R.C., 2008a. 11C PiB and structural MRI provide complementary information in imaging of Alzheimer's disease and amnestic mild cognitive impairment. Brain 131, 665-680. Jack, C.R., Jr., Lowe, V.J., Weigand, S.D., Wiste, H.J., Senjem, M.L., Knopman, D.S., Shiung, M.M., Gunter, J.L., Boeve, B.F., Kemp, B.J., Weiner, M., Petersen, R.C., 2009. Serial PIB and MRI in normal, mild cognitive impairment and Alzheimer's disease: implications for sequence of pathological events in Alzheimer's disease. Brain 132, 13551365. Jack, C.R., Jr., Weigand, S.D., Shiung, M.M., Przybelski, S.A., O'Brien, P.C., Gunter, J.L., Knopman, D.S., Boeve, B.F., Smith, G.E., Petersen, R.C., 2008b. Atrophy rates accelerate in amnestic mild cognitive impairment. Neurology 70, 1740-1752. Jack, C.R., Jr., Wiste, H.J., Lesnick, T.G., Weigand, S.D., Knopman, D.S., Vemuri, P., Pankratz, V.S., Senjem, M.L., Gunter, J.L., Mielke, M.M., Lowe, V.J., Boeve, B.F., Petersen, R.C., 2013b. Brain beta-amyloid load approaches a plateau. Neurology 80, 890-896. Jack, C.R., Jr., Wiste, H.J., Weigand, S.D., Knopman, D.S., Lowe, V., Vemuri, P., Mielke, M.M., Jones, D.T., Senjem, M.L., Gunter, J.L., Gregg, B.E., Pankratz, V.S., Petersen, R.C., 2013c. Amyloid-first and neurodegeneration-first profiles characterize incident amyloid PET positivity. Neurology 81, 1732-1740. Jagust, W., Reed, B., Mungas, D., Ellis, W., Decarli, C., 2007. What does fluorodeoxyglucose PET imaging add to a clinical diagnosis of dementia? Neurology 69, 871-877.

38

Jagust, W.J., Landau, S.M., Alzheimer's Disease Neuroimaging, I., 2012. Apolipoprotein E, not fibrillar beta-amyloid, reduces cerebral glucose metabolism in normal aging. J Neurosci 32, 18227-18233. Jansen, W.J., Ossenkoppele, R., Knol, D.L., Tijms, B.M., Scheltens, P., Verhey, F.R., Visser, P.J., Amyloid Biomarker Study, G., Aalten, P., Aarsland, D., Alcolea, D., Alexander, M., Almdahl, I.S., Arnold, S.E., Baldeiras, I., Barthel, H., van Berckel, B.N., Bibeau, K., Blennow, K., Brooks, D.J., van Buchem, M.A., Camus, V., Cavedo, E., Chen, K., Chetelat, G., Cohen, A.D., Drzezga, A., Engelborghs, S., Fagan, A.M., Fladby, T., Fleisher, A.S., van der Flier, W.M., Ford, L., Forster, S., Fortea, J., Foskett, N., Frederiksen, K.S., Freund-Levi, Y., Frisoni, G.B., Froelich, L., Gabryelewicz, T., Gill, K.D., Gkatzima, O., Gomez-Tortosa, E., Gordon, M.F., Grimmer, T., Hampel, H., Hausner, L., Hellwig, S., Herukka, S.K., Hildebrandt, H., Ishihara, L., Ivanoiu, A., Jagust, W.J., Johannsen, P., Kandimalla, R., Kapaki, E., Klimkowicz-Mrowiec, A., Klunk, W.E., Kohler, S., Koglin, N., Kornhuber, J., Kramberger, M.G., Van Laere, K., Landau, S.M., Lee, D.Y., de Leon, M., Lisetti, V., Lleo, A., Madsen, K., Maier, W., Marcusson, J., Mattsson, N., de Mendonca, A., Meulenbroek, O., Meyer, P.T., Mintun, M.A., Mok, V., Molinuevo, J.L., Mollergard, H.M., Morris, J.C., Mroczko, B., Van der Mussele, S., Na, D.L., Newberg, A., Nordberg, A., Nordlund, A., Novak, G.P., Paraskevas, G.P., Parnetti, L., Perera, G., Peters, O., Popp, J., Prabhakar, S., Rabinovici, G.D., Ramakers, I.H., Rami, L., Resende de Oliveira, C., Rinne, J.O., Rodrigue, K.M., Rodriguez-Rodriguez, E., Roe, C.M., Rot, U., Rowe, C.C., Ruther, E., Sabri, O., Sanchez-Juan, P., Santana, I., Sarazin, M., Schroder, J., Schutte, C., Seo, S.W., Soetewey, F., Soininen, H., Spiru, L., Struyfs, H., Teunissen, C.E., Tsolaki, M., Vandenberghe, R., Verbeek, M.M., Villemagne, V.L., Vos, S.J., van Waalwijk van Doorn, L.J., Waldemar, G., Wallin, A., Wallin, A.K., Wiltfang, J., Wolk, D.A., Zboch, M., Zetterberg, H., 2015. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA 313, 1924-1938.

39

Jellinger, K., 1990. Morphology of Alzheimer disease and related disorders, in: Maurer, K., Riederer, P., Beckmann, H. (Eds.), Alzheimer disease: epidemiology, neuropathology, neurochemistry, and clinics. Springer-Verlag, Berlin, pp. 61-77. Jellinger, K.A., 2010. Con: Can neuropathology really confirm the exact diagnosis? Alzheimers Res Ther 2, 11. Jellinger, K.A., Attems, J., 2005. Prevalence and pathogenic role of cerebrovascular lesions in Alzheimer disease. J Neurol Sci 229-230, 37-41. Jellinger, K.A., Bancher, C., 1998. Neuropathology of Alzheimer's disease: a critical update. J Neural Transm Suppl 54, 77-95. Johnson, K.A., Gregas, M., Becker, J.A., Kinnecom, C., Salat, D.H., Moran, E.K., Smith, E.E., Rosand, J., Rentz, D.M., Klunk, W.E., Mathis, C.A., Price, J.C., Dekosky, S.T., Fischman, A.J., Greenberg, S.M., 2007. Imaging of amyloid burden and distribution in cerebral amyloid angiopathy. Ann Neurol 62, 229-234. Johnson, K.A., Schultz, A., Betensky, R.A., Becker, J.A., Sepulcre, J., Rentz, D., Mormino, E., Chhatwal, J., Amariglio, R., Papp, K., Marshall, G., Albers, M., Mauro, S., Pepin, L., Alverio, J., Judge, K., Philiossaint, M., Shoup, T., Yokell, D., Dickerson, B., Gomez-Isla, T., Hyman, B., Vasdev, N., Sperling, R., 2015. Tau PET imaging in aging and early Alzheimer's disease. Ann Neurol. Johnson, N., Davis, T., Bosanquet, N., 2000. The epidemic of Alzheimer's disease. How can we manage the costs? Pharmacoeconomics 18, 215-223. Josephs, K.A., Holton, J.L., Rossor, M.N., Godbolt, A.K., Ozawa, T., Strand, K., Khan, N., Al-Sarraj, S., Revesz, T., 2004. Frontotemporal lobar degeneration and ubiquitin immunohistochemistry. Neuropathol Appl Neurobiol 30, 369-373. Kantarci, K., Lowe, V., Przybelski, S.A., Weigand, S.D., Senjem, M.L., Ivnik, R.J., Preboske, G.M., Roberts, R., Geda, Y.E., Boeve, B.F., Knopman, D.S., Petersen, R.C., Jack, C.R., Jr.,

40

2012. APOE modifies the association between Abeta load and cognition in cognitively normal older adults. Neurology 78, 232-240. Kemppainen, N.M., Aalto, S., Wilson, I.A., Nagren, K., Helin, S., Bruck, A., Oikonen, V., Kailajarvi, M., Scheinin, M., Viitanen, M., Parkkola, R., Rinne, J.O., 2007. PET amyloid ligand [11C]PIB uptake is increased in mild cognitive impairment. Neurology 68, 1603-1606. Kennedy, A.M., Frackowiak, R.S., Newman, S.K., Bloomfield, P., Seaward, J., Roques, P., Lewington, G., Cunningham, V.J., Rossor, M.N., 1995a. Deficits in cerebral glucose metabolism demonstrated by positron emission tomography in individuals at risk of familial Alzheimer's disease. Neurosci Lett 186, 17-20. Kennedy, A.M., Frackowiak, R.S., Newman, S.K., Bloomfield, P.M., Seaward, J., Roques, P., Lewington, G., Cunningham, V.J., Rossor, M.N., 1995b. Deficits in cerebral glucose metabolism demonstrated by positron emission tomography in individuals at risk of familial Alzheimer's disease. Neurosci Lett 186, 17-20. Khachaturian, Z.S., 1985. Diagnosis of Alzheimer's disease. Arch Neurol 42, 1097- 1105. Klunk, W.E., 2011. Amyloid imaging as a biomarker for cerebral beta-amyloidosis and risk prediction for Alzheimer dementia. Neurobiol Aging 32 Suppl 1, S20-36. Klunk, W.E., Engler, H., Nordberg, A., Wang, Y., Blomqvist, G., Holt, D.P., Bergstrom, M., Savitcheva, I., Huang, G.F., Estrada, S., Ausen, B., Debnath, M.L., Barletta, J., Price, J.C., Sandell, J., Lopresti, B.J., Wall, A., Koivisto, P., Antoni, G., Mathis, C.A., Langstrom, B., 2004. Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B. Ann Neurol 55, 306-319. Klunk, W.E., Price, J.C., Mathis, C.A., Tsopelas, N.D., Lopresti, B.J., Ziolko, S.K., Bi, W., Hoge, J.A., Cohen, A.D., Ikonomovic, M.D., Saxton, J.A., Snitz, B.E., Pollen, D.A., Moonis, M., Lippa, C.F., Swearer, J.M., Johnson, K.A., Rentz, D.M., Fischman, A.J., Aizenstein, H.J., DeKosky, S.T., 2007. Amyloid deposition begins in the striatum of presenilin-1 mutation carriers from two unrelated pedigrees. J Neurosci 27, 6174-6184.

41

Knopman, D.S., 2013. Alzheimer disease biomarkers and insights into mild cognitive impairment. Neurology 80, 978-980. Knopman, D.S., Jack, C.R., Jr., Wiste, H.J., Weigand, S.D., Vemuri, P., Lowe, V., Kantarci, K., Gunter, J.L., Senjem, M.L., Ivnik, R.J., Roberts, R.O., Boeve, B.F., Petersen, R.C., 2012. Short-term clinical outcomes for stages of NIA-AA preclinical Alzheimer disease. Neurology 78, 1576-1582. Knudsen, K.A., Rosand, J., Karluk, D., Greenberg, S.M., 2001. Clinical diagnosis of cerebral amyloid angiopathy: validation of the Boston criteria. Neurology 56, 537-539. Koivunen, J., Pirttila, T., Kemppainen, N., Aalto, S., Herukka, S.K., Jauhianen, A.M., Hanninen, T., Hallikainen, M., Nagren, K., Rinne, J.O., Soininen, H., 2008a. PET Amyloid Ligand [C]PIB Uptake and Cerebrospinal Fluid beta-Amyloid in Mild Cognitive Impairment. Dement Geriatr Cogn Disord 26, 378-383. Koivunen, J., Verkkoniemi, A., Aalto, S., Paetau, A., Ahonen, J.P., Viitanen, M., Nagren, K., Rokka, J., Haaparanta, M., Kalimo, H., Rinne, J.O., 2008b. PET amyloid ligand [11C]PIB uptake shows predominantly striatal increase in variant Alzheimer's disease. Brain 131, 18451853. Kudo, Y., Okamura, N., Furumoto, S., Tashiro, M., Furukawa, K., Maruyama, M., Itoh, M., Iwata, R., Yanai, K., Arai, H., 2007. 2-(2-[2-Dimethylaminothiazol-5-yl]Ethenyl)-6- (2[Fluoro]Ethoxy)Benzoxazole: A Novel PET Agent for In Vivo Detection of Dense Amyloid Plaques in Alzheimer's Disease Patients. J Nucl Med 48, 553-561. La Joie, R., Perrotin, A., Barre, L., Hommet, C., Mezenge, F., Ibazizene, M., Camus, V., Abbas, A., Landeau, B., Guilloteau, D., de La Sayette, V., Eustache, F., Desgranges, B., Chetelat, G., 2012. Region-specific hierarchy between atrophy, hypometabolism, and betaamyloid (Abeta) load in Alzheimer's disease dementia. J Neurosci 32, 16265-16273.

42

Landau, S.M., Harvey, D., Madison, C.M., Koeppe, R.A., Reiman, E.M., Foster, N.L., Weiner, M.W., Jagust, W.J., 2009. Associations between cognitive, functional, and FDG-PET measures of decline in AD and MCI. Neurobiol Aging. Larson, E.B., Edwards, J.K., O'Meara, E., Nochlin, D., Sumi, S.M., 1996. Neuropathologic diagnostic outcomes from a cohort of outpatients with suspected dementia. J Gerontol A Biol Sci Med Sci 51(suppl 6), M313-M318. Leyton, C.E., Villemagne, V.L., Savage, S., Pike, K.E., Ballard, K.J., Piguet, O., Burrell, J.R., Rowe, C.C., Hodges, J.R., 2011. Subtypes of progressive aphasia: application of the international consensus criteria and validation using {beta}-amyloid imaging. Brain. Liberski, P.P., 2004. Amyloid plaques in transmissible spongiform encephalopathies (prion diseases). Folia Neuropathol 42 Suppl B, 109-119. Lowe, V.J., Kemp, B.J., Jack, C.R., Jr., Senjem, M., Weigand, S., Shiung, M., Smith, G., Knopman, D., Boeve, B., Mullan, B., Petersen, R.C., 2009. Comparison of 18F-FDG and PiB PET in cognitive impairment. J Nucl Med 50, 878-886. Mackenzie, I.R., Foti, D., Woulfe, J., Hurwitz, T.A., 2008. Atypical frontotemporal lobar degeneration with ubiquitin-positive, TDP-43-negative neuronal inclusions. Brain 131, 12821293. Maetzler, W., Liepelt, I., Reimold, M., Reischl, G., Solbach, C., Becker, C., Schulte, C., Leyhe, T., Keller, S., Melms, A., Gasser, T., Berg, D., 2009. Cortical PIB binding in Lewy body disease is associated with Alzheimer-like characteristics. Neurobiol Dis 34, 107-112. Masliah, E., Rockenstein, E., Veinbergs, I., Sagara, Y., Mallory, M., Hashimoto, M., Mucke, L., 2001. Beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease. Proc Natl Acad Sci U S A 98, 12245-12250. Masters, C.L., 2005. Neuropathology of Alzheimer's Disease, in: Burns, A., O'Brien, J., Ames, D. (Eds.), Dementia (3rd Edition). Hodder Arnold, London, pp. 393-407.

43

Masters, C.L., Beyreuther, K., 2005. The neuropathology of Alzheimer's disease in the year 2005, in: Beal, M.F., Lang, A.E., Ludolph, A.C. (Eds.), Neurodegenerative Diseases: Neurobiology, Pathogenesis and Therapeutics. Cambridge University Press, Cambridge, pp. 433-440. Masters, C.L., Beyreuther, K., 2006. Alzheimer's centennial legacy: prospects for rational therapeutic intervention targeting the Abeta amyloid pathway. Brain 129, 2823-2839. Masters, C.L., Cappai, R., Barnham, K.J., Villemagne, V.L., 2006. Molecular mechanisms for Alzheimer's disease: implications for neuroimaging and therapeutics. J Neurochem 97, 17001725. Masters, C.L., Simms, G., Weinman, N.A., Multhaup, G., McDonald, B.L., Beyreuther, K., 1985. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 82, 4245-4249. Mattsson, N., Andreasson, U., Persson, S., Arai, H., Batish, S.D., Bernardini, S., BocchioChiavetto, L., Blankenstein, M.A., Carrillo, M.C., Chalbot, S., Coart, E., Chiasserini, D., Cutler, N., Dahlfors, G., Duller, S., Fagan, A.M., Forlenza, O., Frisoni, G.B., Galasko, D., Galimberti, D., Hampel, H., Handberg, A., Heneka, M.T., Herskovits, A.Z., Herukka, S.K., Holtzman, D.M., Humpel, C., Hyman, B.T., Iqbal, K., Jucker, M., Kaeser, S.A., Kaiser, E., Kapaki, E., Kidd, D., Klivenyi, P., Knudsen, C.S., Kummer, M.P., Lui, J., Llado, A., Lewczuk, P., Li, Q.X., Martins, R., Masters, C., McAuliffe, J., Mercken, M., Moghekar, A., Molinuevo, J.L., Montine, T.J., Nowatzke, W., O'Brien, R., Otto, M., Paraskevas, G.P., Parnetti, L., Petersen, R.C., Prvulovic, D., de Reus, H.P., Rissman, R.A., Scarpini, E., Stefani, A., Soininen, H., Schroder, J., Shaw, L.M., Skinningsrud, A., Skrogstad, B., Spreer, A., Talib, L., Teunissen, C., Trojanowski, J.Q., Tumani, H., Umek, R.M., Van Broeck, B., Vanderstichele, H., Vecsei, L., Verbeek, M.M., Windisch, M., Zhang, J., Zetterberg, H., Blennow, K., 2011. The Alzheimer's Association external quality control program for cerebrospinal fluid biomarkers. Alzheimers Dement 7, 386-395 e386.

44

Mattsson, N., Zetterberg, H., Hansson, O., Andreasen, N., Parnetti, L., Jonsson, M., Herukka, S.K., van der Flier, W.M., Blankenstein, M.A., Ewers, M., Rich, K., Kaiser, E., Verbeek, M., Tsolaki, M., Mulugeta, E., Rosen, E., Aarsland, D., Visser, P.J., Schroder, J., Marcusson, J., de Leon, M., Hampel, H., Scheltens, P., Pirttila, T., Wallin, A., Jonhagen, M.E., Minthon, L., Winblad, B., Blennow, K., 2009. CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. Jama 302, 385-393. Mawuenyega, K.G., Sigurdson, W., Ovod, V., Munsell, L., Kasten, T., Morris, J.C., Yarasheski, K.E., Bateman, R.J., 2010. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330, 1774. McKeith, I.G., Dickson, D.W., Lowe, J., Emre, M., O'Brien, J.T., Feldman, H., Cummings, J., Duda, J.E., Lippa, C., Perry, E.K., Aarsland, D., Arai, H., Ballard, C.G., Boeve, B., Burn, D.J., Costa, D., Del Ser, T., Dubois, B., Galasko, D., Gauthier, S., Goetz, C.G., GomezTortosa, E., Halliday, G., Hansen, L.A., Hardy, J., Iwatsubo, T., Kalaria, R.N., Kaufer, D., Kenny, R.A., Korczyn, A., Kosaka, K., Lee, V.M., Lees, A., Litvan, I., Londos, E., Lopez, O.L., Minoshima, S., Mizuno, Y., Molina, J.A., Mukaetova-Ladinska, E.B., Pasquier, F., Perry, R.H., Schulz, J.B., Trojanowski, J.Q., Yamada, M., 2005. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology 65, 18631872. McKeith, I.G., Mosimann, U.P., 2004. Dementia with Lewy bodies and Parkinson's disease. Parkinsonism Relat Disord 10 Suppl 1, S15-18. McKhann, G.M., Knopman, D.S., Chertkow, H., Hyman, B.T., Jack, C.R., Jr., Kawas, C.H., Klunk, W.E., Koroshetz, W.J., Manly, J.J., Mayeux, R., Mohs, R.C., Morris, J.C., Rossor, M.N., Scheltens, P., Carrillo, M.C., Thies, B., Weintraub, S., Phelps, C.H., 2011. The diagnosis of dementia due to Alzheimer's disease: Recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 7, 263-269.

45

Michaelis, M.L., Dobrowsky, R.T., Li, G., 2002. Tau neurofibrillary pathology and microtubule stability. J Mol Neurosci 19, 289-293. Mintun, M.A., Larossa, G.N., Sheline, Y.I., Dence, C.S., Lee, S.Y., Mach, R.H., Klunk, W.E., Mathis, C.A., DeKosky, S.T., Morris, J.C., 2006. [11C]PIB in a nondemented population: potential antecedent marker of Alzheimer disease. Neurology 67, 446-452. Mormino, E.C., Kluth, J.T., Madison, C.M., Rabinovici, G.D., Baker, S.L., Miller, B.L., Koeppe, R.A., Mathis, C.A., Weiner, M.W., Jagust, W.J., 2009. Episodic memory loss is related to hippocampal-mediated beta-amyloid deposition in elderly subjects. Brain 132, 1310-1323. Morris, J.C., 2012. Revised Criteria for Mild Cognitive Impairment May Compromise the Diagnosis of Alzheimer Disease Dementia. Arch Neurol, doi:10.1001/archneurol.2011.3152. Morris, J.C., Kimberly, A., Quaid, K., Holtzman, D.M., Kantarci, K., Kaye, J., Reiman, E.M., Klunk, W.E., Siemers, E.R., 2005. Role of biomarkers in studies of presymptomatic Alzheimer's disease. Alzheimers Dement 1, 145-151. Morris, J.C., Price, A.L., 2001. Pathologic correlates of nondemented aging, mild cognitive impairment, and early-stage Alzheimer's disease. J Mol Neurosci 17, 101-118. Morris, J.C., Roe, C.M., Grant, E.A., Head, D., Storandt, M., Goate, A.M., Fagan, A.M., Holtzman, D.M., Mintun, M.A., 2009. Pittsburgh Compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch Neurol 66, 1469-1475. Mosconi, L., De Santi, S., Li, Y., Li, J., Zhan, J., Tsui, W.H., Boppana, M., Pupi, A., de Leon, M.J., 2006a. Visual rating of medial temporal lobe metabolism in mild cognitive impairment and Alzheimer's disease using FDG-PET. Eur J Nucl Med Mol Imaging 33, 210-221. Mosconi, L., Perani, D., Sorbi, S., Herholz, K., Nacmias, B., Holthoff, V., Salmon, E., Baron, J.C., De Cristofaro, M.T., Padovani, A., Borroni, B., Franceschi, M., Bracco, L., Pupi, A.,

46

2004. MCI conversion to dementia and the APOE genotype: a prediction study with FDGPET. Neurology 63, 2332-2340. Mosconi, L., Sorbi, S., de Leon, M.J., Li, Y., Nacmias, B., Myoung, P.S., Tsui, W., Ginestroni, A., Bessi, V., Fayyazz, M., Caffarra, P., Pupi, A., 2006b. Hypometabolism exceeds atrophy in presymptomatic early-onset familial Alzheimer's disease. J Nucl Med 47, 1778-1786. Mott, R.T., Dickson, D.W., Trojanowski, J.Q., Zhukareva, V., Lee, V.M., Forman, M., Van Deerlin, V., Ervin, J.F., Wang, D.S., Schmechel, D.E., Hulette, C.M., 2005. Neuropathologic, biochemical, and molecular characterization of the frontotemporal dementias. J Neuropathol Exp Neurol 64, 420-428. Neumann, M., Rademakers, R., Roeber, S., Baker, M., Kretzschmar, H.A., Mackenzie, I.R., 2009. A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 132, 2922-2931. Neumann, M., Sampathu, D.M., Kwong, L.K., Truax, A.C., Micsenyi, M.C., Chou, T.T., Bruce, J., Schuck, T., Grossman, M., Clark, C.M., McCluskey, L.F., Miller, B.L., Masliah, E., Mackenzie, I.R., Feldman, H., Feiden, W., Kretzschmar, H.A., Trojanowski, J.Q., Lee, V.M., 2006. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314, 130-133. O'Brien, J., Ames, D., Burns, A., 2000. Dementia (2nd Edn). Arnold, London. Okamura, N., Shiga, Y., Furumoto, S., Tashiro, M., Tsuboi, Y., Furukawa, K., Yanai, K., Iwata, R., Arai, H., Kudo, Y., Itoyama, Y., Doh-ura, K., 2010. In vivo detection of prion amyloid plaques using [(11)C]BF-227 PET. Eur J Nucl Med Mol Imaging 37, 934-941. Okello, A., Edison, P., Archer, H.A., Turkheimer, F.E., Kennedy, J., Bullock, R., Walker, Z., Kennedy, A., Fox, N., Rossor, M., Brooks, D.J., 2009a. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology 72, 56-62.

47

Okello, A., Koivunen, J., Edison, P., Archer, H.A., Turkheimer, F.E., Nagren, K., Bullock, R., Walker, Z., Kennedy, A., Fox, N.C., Rossor, M.N., Rinne, J.O., Brooks, D.J., 2009b. Conversion of amyloid positive and negative MCI to AD over 3 years: an 11C-PIB PET study. Neurology 73, 754-760. Ong, K., Villemagne, V.L., Bahar-Fuchs, A., Lamb, F., Chetelat, G., Raniga, P., Mulligan, R.S., Salvado, O., Putz, B., Roth, K., Masters, C.L., Reininger, C.B., Rowe, C.C., 2013. (18)F-florbetaben Abeta imaging in mild cognitive impairment. Alzheimers Res Ther 5, 4. Ong, K.T., Villemagne, V.L., Bahar-Fuchs, A., Lamb, F., Langdon, N., Catafau, A.M., Stephens, A.W., Seibyl, J., Dinkelborg, L.M., Reininger, C.B., Putz, B., Rohde, B., Masters, C.L., Rowe, C.C., 2015. Abeta imaging with 18F-florbetaben in prodromal Alzheimer's disease: a prospective outcome study. J Neurol Neurosurg Psychiatry 86, 431-436. Ostrowitzki, S., Deptula, D., Thurfjell, L., Barkhof, F., Bohrmann, B., Brooks, D.J., Klunk, W.E., Ashford, E., Yoo, K., Xu, Z.X., Loetscher, H., Santarelli, L., 2011. Mechanism of Amyloid Removal in Patients With Alzheimer Disease Treated With Gantenerumab. Arch Neurol. Petersen, R.C., 2000. Mild cognitive impairment: transition between aging and Alzheimer's disease. Neurologia 15, 93-101. Petersen, R.C., Smith, G.E., Waring, S.C., Ivnik, R.J., Tangalos, E.G., Kokmen, E., 1999. Mild cognitive impairment: clinical characterization and outcome. Arch Neurol 56, 303-308. Pettersen, J.A., Sathiyamoorthy, G., Gao, F.Q., Szilagyi, G., Nadkarni, N.K., St GeorgeHyslop, P., Rogaeva, E., Black, S.E., 2008. Microbleed topography, leukoaraiosis, and cognition in probable Alzheimer disease from the Sunnybrook dementia study. Arch Neurol 65, 790-795. Pike, K.E., Ellis, K.A., Villemagne, V.L., Good, N., Chetelat, G., Ames, D., Szoeke, C., Laws, S.M., Verdile, G., Martins, R.N., Masters, C.L., Rowe, C.C., 2011. Cognition and beta-

48

amyloid in preclinical Alzheimer's disease: Data from the AIBL study. Neuropsychologia 49, 2384-2390. Pike, K.E., Savage, G., Villemagne, V.L., Ng, S., Moss, S.A., Maruff, P., Mathis, C.A., Klunk, W.E., Masters, C.L., Rowe, C.C., 2007. Beta-amyloid imaging and memory in nondemented individuals: evidence for preclinical Alzheimer's disease. Brain 130, 2837-2844. Pimplikar, S.W., Nixon, R.A., Robakis, N.K., Shen, J., Tsai, L.H., 2010. Amyloidindependent mechanisms in Alzheimer's disease pathogenesis. J Neurosci 30, 14946-14954. Pletnikova, O., West, N., Lee, M.K., Rudow, G.L., Skolasky, R.L., Dawson, T.M., Marsh, L., Troncoso, J.C., 2005. Abeta deposition is associated with enhanced cortical a-synuclein lesions in Lewy body diseases. Neurobiol Aging 26. Price, J.C., Klunk, W.E., Lopresti, B.J., Lu, X., Hoge, J.A., Ziolko, S.K., Holt, D.P., Meltzer, C.C., DeKosky, S.T., Mathis, C.A., 2005. Kinetic modeling of amyloid binding in humans using PET imaging and Pittsburgh Compound-B. J Cereb Blood Flow Metab 25, 1528-1547. Rabinovici, G.D., Furst, A.J., O'Neil, J.P., Racine, C.A., Mormino, E.C., Baker, S.L., Chetty, S., Patel, P., Pagliaro, T.A., Klunk, W.E., Mathis, C.A., Rosen, H.J., Miller, B.L., Jagust, W.J., 2007. 11C-PIB PET imaging in Alzheimer disease and frontotemporal lobar degeneration. Neurology 68, 1205-1212. Rabinovici, G.D., Jagust, W.J., Furst, A.J., Ogar, J.M., Racine, C.A., Mormino, E.C., O'Neil, J.P., Lal, R.A., Dronkers, N.F., Miller, B.L., Gorno-Tempini, M.L., 2008. Abeta amyloid and glucose metabolism in three variants of primary progressive aphasia. Ann Neurol 64, 388401. Rabinovici, G.D., Miller, B.L., 2010. Frontotemporal lobar degeneration: epidemiology, pathophysiology, diagnosis and management. CNS Drugs 24, 375-398. Rabinovici, G.D., Rosen, H.J., Alkalay, A., Kornak, J., Furst, A.J., Agarwal, N., Mormino, E.C., O'Neil, J.P., Janabi, M., Karydas, A., Growdon, M.E., Jang, J.Y., Huang, E.J., Dearmond, S.J., Trojanowski, J.Q., Grinberg, L.T., Gorno-Tempini, M.L., Seeley, W.W.,

49

Miller, B.L., Jagust, W.J., 2011. Amyloid vs FDG-PET in the differential diagnosis of AD and FTLD. Neurology 77, 2034-2042. Reiman, E.M., Chen, K., Liu, X., Bandy, D., Yu, M., Lee, W., Ayutyanont, N., Keppler, J., Reeder, S.A., Langbaum, J.B., Alexander, G.E., Klunk, W.E., Mathis, C.A., Price, J.C., Aizenstein, H.J., DeKosky, S.T., Caselli, R.J., 2009. Fibrillar amyloid-beta burden in cognitively normal people at 3 levels of genetic risk for Alzheimer's disease. Proc Natl Acad Sci U S A 106, 6820-6825. Reiman, E.M., Quiroz, Y.T., Fleisher, A.S., Chen, K., Velez-Pardo, C., Jimenez-Del-Rio, M., Fagan, A.M., Shah, A.R., Alvarez, S., Arbelaez, A., Giraldo, M., Acosta-Baena, N., Sperling, R.A., Dickerson, B., Stern, C.E., Tirado, V., Munoz, C., Reiman, R.A., Huentelman, M.J., Alexander, G.E., Langbaum, J.B., Kosik, K.S., Tariot, P.N., Lopera, F., 2012. Brain imaging and fluid biomarker analysis in young adults at genetic risk for autosomal dominant Alzheimer's disease in the presenilin 1 E280A kindred: a case-control study. Lancet Neurol 11, 1048-1056. Rentz, D.M., Locascio, J.J., Becker, J.A., Moran, E.K., Eng, E., Buckner, R.L., Sperling, R.A., Johnson, K.A., 2010. Cognition, reserve, and amyloid deposition in normal aging. Ann Neurol 67, 353-364. Resende, R., Pereira, C., Agostinho, P., Vieira, A.P., Malva, J.O., Oliveira, C.R., 2007. Susceptibility of hippocampal neurons to Abeta peptide toxicity is associated with perturbation of Ca2+ homeostasis. Brain Res 1143, 11-21. Resnick, S.M., Sojkova, J., Zhou, Y., An, Y., Ye, W., Holt, D.P., Dannals, R.F., Mathis, C.A., Klunk, W.E., Ferrucci, L., Kraut, M.A., Wong, D.F., 2010. Longitudinal cognitive decline is associated with fibrillar amyloid-beta measured by [11C]PiB. Neurology 74, 807-815. Rinne, J.O., Brooks, D.J., Rossor, M.N., Fox, N.C., Bullock, R., Klunk, W.E., Mathis, C.A., Blennow, K., Barakos, J., Okello, A.A., Rodriguez Martinez de Liano, S., Liu, E., Koller, M., Gregg, K.M., Schenk, D., Black, R., Grundman, M., 2010. 11C-PiB PET assessment of

50

change in fibrillar amyloid-beta load in patients with Alzheimer's disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol 9, 363-372. Roder, S., Danober, L., Pozza, M.F., Lingenhoehl, K., Wiederhold, K.H., Olpe, H.R., 2003. Electrophysiological studies on the hippocampus and prefrontal cortex assessing the effects of amyloidosis in amyloid precursor protein 23 transgenic mice. Neuroscience 120, 705-720. Roe, C.M., Mintun, M.A., D'Angelo, G., Xiong, C., Grant, E.A., Morris, J.C., 2008. Alzheimer disease and cognitive reserve: variation of education effect with carbon 11-labeled Pittsburgh Compound B uptake. Arch Neurol 65, 1467-1471. Rossor, M.N., Kennedy, A.M., Frackowiak, R.S., 1996. Clinical and neuroimaging features of familial Alzheimer's disease. Ann N Y Acad Sci 777, 49-56. Rowe, C.C., Bourgeat, P., Ellis, K.A., Brown, B., Lim, Y.Y., Mulligan, R., Jones, G., Maruff, P., Woodward, M., Price, R., Robins, P., Tochon-Danguy, H., O'Keefe, G., Pike, K.E., Yates, P., Szoeke, C., Salvado, O., Macaulay, S.L., O'Meara, T., Head, R., Cobiac, L., Savage, G., Martins, R., Masters, C.L., Ames, D., Villemagne, V.L., 2013. Predicting Alzheimer disease with beta-amyloid imaging: results from the Australian imaging, biomarkers, and lifestyle study of ageing. Ann Neurol 74, 905-913. Rowe, C.C., Ellis, K.A., Rimajova, M., Bourgeat, P., Pike, K.E., Jones, G., Fripp, J., TochonDanguy, H., Morandeau, L., O'Keefe, G., Price, R., Raniga, P., Robins, P., Acosta, O., Lenzo, N., Szoeke, C., Salvado, O., Head, R., Martins, R., Masters, C.L., Ames, D., Villemagne, V.L., 2010. Amyloid imaging results from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging 31, 1275-1283. Rowe, C.C., Ng, S., Ackermann, U., Gong, S.J., Pike, K., Savage, G., Cowie, T.F., Dickinson, K.L., Maruff, P., Darby, D., Smith, C., Woodward, M., Merory, J., Tochon-Danguy, H., O'Keefe, G., Klunk, W.E., Mathis, C.A., Price, J.C., Masters, C.L., Villemagne, V.L., 2007. Imaging beta-amyloid burden in aging and dementia. Neurology 68, 1718-1725.

51

Salloway, S., Sperling, R., Fox, N.C., Blennow, K., Klunk, W., Raskind, M., Sabbagh, M., Honig, L.S., Porsteinsson, A.P., Ferris, S., Reichert, M., Ketter, N., Nejadnik, B., Guenzler, V., Miloslavsky, M., Wang, D., Lu, Y., Lull, J., Tudor, I.C., Liu, E.C., Grundman, M., Yuen, E., Black, R., Brashear, H.R., Clinical, B., 2014. Two Phase 3 Trials of Bapineuzumab in Mild-to-Moderate Alzheimer's Disease. New Engl J Med 370, 322-333. Salmon, E., Sadzot, B., Maquet, P., Degueldre, C., Lemaire, C., Rigo, P., Comar, D., Franck, G., 1994. Differential diagnosis of Alzheimer's disease with PET. J Nucl Med 35, 391- 398. Scheltens, P., 2013. Dementia: Mild cognitive impairment--amyloid and beyond. Nat Rev Neurol 9, 493-495. Scheltens, P., Rockwood, K., 2011. How golden is the gold standard of neuropathology in dementia? Alzheimers Dement 7, 486-489. Schulz-Schaeffer, W.J., 2010. The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia. Acta Neuropathol 120, 131-143. Shaw, L.M., Korecka, M., Clark, C.M., Lee, V.M., Trojanowski, J.Q., 2007. Biomarkers of neurodegeneration for diagnosis and monitoring therapeutics. Nat Rev Drug Discov 6, 295303. Sheline, Y.I., Morris, J.C., Snyder, A.Z., Price, J.L., Yan, Z., D'Angelo, G., Liu, C., Dixit, S., Benzinger, T., Fagan, A., Goate, A., Mintun, M.A., 2010. APOE4 allele disrupts resting state fMRI connectivity in the absence of amyloid plaques or decreased CSF Abeta42. J Neurosci 30, 17035-17040. Silverman, D.H., Cummings, J.L., Small, G.W., Gambhir, S.S., Chen, W., Czernin, J., Phelps, M.E., 2002. Added clinical benefit of incorporating 2-deoxy-2-[18F]fluoro-D-glucose with positron emission tomography into the clinical evaluation of patients with cognitive impairment. Mol Imaging Biol 4, 283-293.

52

Silverman, D.H., Small, G.W., Chang, C.Y., Lu, C.S., Kung De Aburto, M.A., Chen, W., Czernin, J., Rapoport, S.I., Pietrini, P., Alexander, G.E., Schapiro, M.B., Jagust, W.J., Hoffman, J.M., Welsh-Bohmer, K.A., Alavi, A., Clark, C.M., Salmon, E., de Leon, M.J., Mielke, R., Cummings, J.L., Kowell, A.P., Gambhir, S.S., Hoh, C.K., Phelps, M.E., 2001. Positron emission tomography in evaluation of dementia: regional brain metabolism and longterm outcome. JAMA 286, 2120-2127. Small, G.W., Mazziotta, J.C., Collins, M.T., Baxter, L.R., Phelps, M.E., Mandelkern, M.A., Kaplan, A., La Rue, A., Adamson, C.F., Chang, L., al., e., 1995. Apolipoprotein E type 4 allele and cerebral glucose metabolism in relatives at risk for familial Alzheimer disease. JAMA 273, 942-947. Small, S.A., Duff, K., 2008. Linking Abeta and tau in late-onset Alzheimer's disease: a dual pathway hypothesis. Neuron 60, 534-542. Snowden, J., Neary, D., Mann, D., 2007. Frontotemporal lobar degeneration: clinical and pathological relationships. Acta Neuropathol 114, 31-38. Sojkova, J., Resnick, S.M., 2011. In vivo human amyloid imaging. Curr Alzheimer Res 8, 366-372. Sojkova, J., Zhou, Y., An, Y., Kraut, M.A., Ferrucci, L., Wong, D.F., Resnick, S.M., 2011. Longitudinal patterns of beta-amyloid deposition in nondemented older adults. Arch Neurol 68, 644-649. Sperling, R., Johnson, K., 2010. Pro: Can biomarkers be gold standards in Alzheimer's disease? Alzheimers Res Ther 2, 17. Sperling, R.A., Aisen, P.S., Beckett, L.A., Bennett, D.A., Craft, S., Fagan, A.M., Iwatsubo, T., Jack, C.R., Jr., Kaye, J., Montine, T.J., Park, D.C., Reiman, E.M., Rowe, C.C., Siemers, E., Stern, Y., Yaffe, K., Carrillo, M.C., Thies, B., Morrison-Bogorad, M., Wagster, M.V., Phelps, C.H., 2011a. Toward defining the preclinical stages of Alzheimer's disease:

53

Recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 7, 280-292. Sperling, R.A., Jack, C.R., Jr., Aisen, P.S., 2011b. Testing the right target and right drug at the right stage. Sci Transl Med 3, 111cm133. Sperling, R.A., Jack, C.R., Jr., Black, S.E., Frosch, M.P., Greenberg, S.M., Hyman, B.T., Scheltens, P., Carrillo, M.C., Thies, W., Bednar, M.M., Black, R.S., Brashear, H.R., Grundman, M., Siemers, E.R., Feldman, H.H., Schindler, R.J., 2011c. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer's Association Research Roundtable Workgroup. Alzheimers Dement 7, 367-385. Sperling, R.A., Johnson, K.A., Doraiswamy, P.M., Reiman, E.M., Fleisher, A.S., Sabbagh, M.N., Sadowsky, C.H., Carpenter, A., Davis, M.D., Lu, M., Flitter, M., Joshi, A.D., Clark, C.M., Grundman, M., Mintun, M.A., Skovronsky, D.M., Pontecorvo, M.J., 2012. Amyloid deposition detected with florbetapir F 18 ((18)F-AV-45) is related to lower episodic memory performance in clinically normal older individuals. Neurobiol Aging. Sperling, R.A., Laviolette, P.S., O'Keefe, K., O'Brien, J., Rentz, D.M., Pihlajamaki, M., Marshall, G., Hyman, B.T., Selkoe, D.J., Hedden, T., Buckner, R.L., Becker, J.A., Johnson, K.A., 2009. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron 63, 178-188. Storandt, M., Head, D., Fagan, A.M., Holtzman, D.M., Morris, J.C., 2012. Toward a multifactorial model of Alzheimer disease. Neurobiol Aging. Storandt, M., Mintun, M.A., Head, D., Morris, J.C., 2009. Cognitive decline and brain volume loss as signatures of cerebral amyloid-beta peptide deposition identified with Pittsburgh compound B: cognitive decline associated with Abeta deposition. Arch Neurol 66, 1476-1481. Strozyk, D., Blennow, K., White, L.R., Launer, L.J., 2003. CSF Abeta 42 levels correlate with amyloid-neuropathology in a population-based autopsy study. Neurology 60, 652-656.

54

Sunderland, T., Gur, R.E., Arnold, S.E., 2005. The use of biomarkers in the elderly: current and future challenges. Biol Psychiatry 58, 272-276. Taniguchi, S., McDonagh, A.M., Pickering-Brown, S.M., Umeda, Y., Iwatsubo, T., Hasegawa, M., Mann, D.M., 2004. The neuropathology of frontotemporal lobar degeneration with respect to the cytological and biochemical characteristics of tau protein. Neuropathol Appl Neurobiol 30, 1-18. Teipel, S., Drzezga, A., Grothe, M.J., Barthel, H., Chetelat, G., Schuff, N., Skudlarski, P., Cavedo, E., Frisoni, G.B., Hoffmann, W., Thyrian, J.R., Fox, C., Minoshima, S., Sabri, O., Fellgiebel, A., 2015. Multimodal imaging in Alzheimer's disease: validity and usefulness for early detection. Lancet Neurol 14, 1037-1053. Thal, L.J., Kantarci, K., Reiman, E.M., Klunk, W.E., Weiner, M.W., Zetterberg, H., Galasko, D., Pratico, D., Griffin, S., Schenk, D., Siemers, E., 2006. The Role of Biomarkers in Clinical Trials for Alzheimer Disease. Alzheimer Dis Assoc Disord 20, 6-15. Tolboom, N., van der Flier, W.M., Yaqub, M., Boellaard, R., Verwey, N.A., Blankenstein, M.A., Windhorst, A.D., Scheltens, P., Lammertsma, A.A., van Berckel, B.N., 2009. Relationship of cerebrospinal fluid markers to 11C-PiB and 18F-FDDNP binding. J Nucl Med 50, 1464-1470. Toledo, J.B., Bjerke, M., Da, X., Landau, S.M., Foster, N.L., Jagust, W., Jack, C., Jr., Weiner, M., Davatzikos, C., Shaw, L.M., Trojanowski, J.Q., Alzheimer's Disease Neuroimaging Initiative, I., 2015. Nonlinear Association Between Cerebrospinal Fluid and Florbetapir F-18 beta-Amyloid Measures Across the Spectrum of Alzheimer Disease. JAMA Neurol 72, 571581. Toledo, J.B., Xie, S.X., Trojanowski, J.Q., Shaw, L.M., 2013. Longitudinal change in CSF Tau and Abeta biomarkers for up to 48 months in ADNI. Acta Neuropathol 126, 659-670.

55

Venneti, S., Lopresti, B.J., Wiley, C.A., 2006. The peripheral benzodiazepine receptor (Translocator protein 18kDa) in microglia: from pathology to imaging. Prog Neurobiol 80, 308-322. Vernooij, M.W., van der Lugt, A., Ikram, M.A., Wielopolski, P.A., Niessen, W.J., Hofman, A., Krestin, G.P., Breteler, M.M., 2008. Prevalence and risk factors of cerebral microbleeds: the Rotterdam Scan Study. Neurology 70, 1208-1214. Villain, N., Chetelat, G., Grassiot, B., Bourgeat, P., Jones, G., Ellis, K.A., Ames, D., Martins, R.N., Eustache, F., Salvado, O., Masters, C.L., Rowe, C.C., Villemagne, V.L., 2012. Regional dynamics of amyloid-beta deposition in healthy elderly, mild cognitive impairment and Alzheimer's disease: a voxelwise PiB-PET longitudinal study. Brain 135, 2126-2139. Villemagne, V.L., Ataka, S., Mizuno, T., Brooks, W.S., Wada, Y., Kondo, M., Jones, G., Watanabe, Y., Mulligan, R., Nakagawa, M., Miki, T., Shimada, H., O'Keefe, G.J., Masters, C.L., Mori, H., Rowe, C.C., 2009a. High striatal amyloid beta-peptide deposition across different autosomal Alzheimer disease mutation types. Arch Neurol 66, 1537-1544. Villemagne, V.L., Burnham, S., Bourgeat, P., Brown, B., Ellis, K.A., Salvado, O., Szoeke, C., Macaulay, S.L., Martins, R., Maruff, P., Ames, D., Rowe, C.C., Masters, C.L., 2013. Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer's disease: a prospective cohort study. Lancet Neurol 12, 357-367. Villemagne, V.L., Cappai, R., Barnham, K.J., Cherny, R., Opazo, C., Novakovic, K.E., Rowe, C.C., Masters, C.L., 2006. The Aβ centric pathway of Alzheimer‟s disease, in: Barrow, C.J., Small, B.J. (Eds.), Abeta Peptide and Alzheimer‟s Disease. Springer-Verlag, London, pp. 532. Villemagne, V.L., Fodero-Tavoletti, M.T., Pike, K.E., Cappai, R., Masters, C.L., Rowe, C.C., 2008a. The ART of Loss: Aβ Imaging in the Evaluation of Alzheimer's Disease and other Dementias. Mol Neurobiol 38, 1-15.

56

Villemagne, V.L., Furumoto, S., Fodero-Tavoletti, M.T., Mulligan, R.S., Hodges, J., Harada, R., Yates, P., Piguet, O., Pejoska, S., Dore, V., Yanai, K., Masters, C.L., Kudo, Y., Rowe, C.C., Okamura, N., 2014. In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease. Eur J Nucl Med Mol Imaging 41, 816-826. Villemagne, V.L., Furumoto, S., Fodero-Tavoletti, M.T., Mulligan, R.S., Hodges, J., Piguet, O., Pejoska, S., Kudo, Y., Masters, C.L., Yanai, K., Rowe, C.C., Okamura, N., 2012. In vivo tau imaging in Alzheimer's disease and other dementias. Alzheimer & Dementia 8, P9. Villemagne, V.L., McLean, C.A., Reardon, K., Boyd, A., Lewis, V., Klug, G., Jones, G., Baxendale, D., Masters, C.L., Rowe, C.C., Collins, S.J., 2009b. 11C-PiB PET studies in typical sporadic Creutzfeldt-Jakob disease. J Neurol Neurosurg Psychiatry 80, 998-1001. Villemagne, V.L., Ong, K., Mulligan, R.S., Holl, G., Pejoska, S., Jones, G., O'Keefe, G., Ackerman, U., Tochon-Danguy, H., Chan, J.G., Reininger, C.B., Fels, L., Putz, B., Rohde, B., Masters, C.L., Rowe, C.C., 2011a. Amyloid Imaging with 18F-Florbetaben in Alzheimer Disease and Other Dementias. J Nucl Med 52, 1210-1217. Villemagne, V.L., Pike, K.E., Chetelat, G., Ellis, K.A., Mulligan, R.S., Bourgeat, P., Ackermann, U., Jones, G., Szoeke, C., Salvado, O., Martins, R., O'Keefe, G., Mathis, C.A., Klunk, W.E., Ames, D., Masters, C.L., Rowe, C.C., 2011b. Longitudinal assessment of Aβ and cognition in aging and Alzheimer disease. Ann Neurol 69, 181-192. Villemagne, V.L., Pike, K.E., Darby, D., Maruff, P., Savage, G., Ng, S., Ackermann, U., Cowie, T.F., Currie, J., Chan, S.G., Jones, G., Tochon-Danguy, H., O'Keefe, G., Masters, C.L., Rowe, C.C., 2008b. Aβ deposits in older non-demented individuals with cognitive decline are indicative of preclinical Alzheimer's disease. Neuropsychologia 46, 1688-1697. Villemagne, V.L., Rowe, C.C., 2010. Amyloid Ligands for Dementia. PET Clin 5, 33-53. Villeneuve, S., Reed, B.R., Wirth, M., Haase, C.M., Madison, C.M., Ayakta, N., Mack, W., Mungas, D., Chui, H.C., DeCarli, C., Weiner, M.W., Jagust, W.J., 2014. Cortical thickness mediates the effect of beta-amyloid on episodic memory. Neurology 82, 761-767. 57

Vishnu, V.Y., 2013. Can tauopathy shake the amyloid cascade hypothesis? Nat Rev Neurol 9, 356. Vlassenko, A.G., Mintun, M.A., Xiong, C., Sheline, Y.I., Goate, A.M., Benzinger, T.L., Morris, J.C., 2011. Amyloid-beta plaque growth in cognitively normal adults: longitudinal [11C]Pittsburgh compound B data. Ann Neurol 70, 857-861. Wahlund, L.O., Blennow, K., 2003. Cerebrospinal fluid biomarkers for disease stage and intensity in cognitively impaired patients. Neurosci Lett 339, 99-102. Weiner, M.W., 2013. Dementia in 2012: Further insights into Alzheimer disease pathogenesis. Nat Rev Neurol 9, 65-66. Weller, R.O., Preston, S.D., Subash, M., Carare, R.O., 2009. Cerebral amyloid angiopathy in the aetiology and immunotherapy of Alzheimer disease. Alzheimers Res Ther 1, 6. Wermke, M., Sorg, C., Wohlschlager, A.M., Drzezga, A., 2008. A new integrative model of cerebral activation, deactivation and default mode function in Alzheimer's disease. Eur J Nucl Med Mol Imaging 35 Suppl 1, S12-24. Wiley, C.A., Lopresti, B.J., Venneti, S., Price, J., Klunk, W.E., DeKosky, S.T., Mathis, C.A., 2009. Carbon 11-labeled Pittsburgh Compound B and carbon 11-labeled (R)-PK11195 positron emission tomographic imaging in Alzheimer disease. Arch Neurol 66, 60-67. Wirth, M., Villeneuve, S., Haase, C.M., Madison, C.M., Oh, H., Landau, S.M., Rabinovici, G.D.,

Jagust,

W.J.,

2013.

Associations

between

Alzheimer

disease

biomarkers,

neurodegeneration, and cognition in cognitively normal older people. JAMA Neurol 70, 1512-1519. Wolk, D.A., Price, J.C., Saxton, J.A., Snitz, B.E., James, J.A., Lopez, O.L., Aizenstein, H.J., Cohen, A.D., Weissfeld, L.A., Mathis, C.A., Klunk, W.E., DeKosky, S.T., 2009. Amyloid imaging in mild cognitive impairment subtypes. Ann Neurol 65, 557-568.

58

Yates, P.A., Sirisriro, R., Villemagne, V.L., Farquharson, S., Masters, C.L., Rowe, C.C., 2011. Cerebral microhemorrhage and brain {beta}-amyloid in aging and Alzheimer disease. Neurology 77, 48-54. Yokokura, M., Mori, N., Yagi, S., Yoshikawa, E., Kikuchi, M., Yoshihara, Y., Wakuda, T., Sugihara, G., Takebayashi, K., Suda, S., Iwata, Y., Ueki, T., Tsuchiya, K.J., Suzuki, K., Nakamura, K., Ouchi, Y., 2011. In vivo changes in microglial activation and amyloid deposits in brain regions with hypometabolism in Alzheimer's disease. Eur J Nucl Med Mol Imaging 38, 343-351.

59

Figure Captions

Figure 1: Left: Schematics showing the stages of A deposition in the human brain as proposed by Braak and Braak (Braak and Braak, 1997) Right: Representative surface projection PET images showing the regional brain distribution of

11

C-PIB in two asymptomatic healthy age-matched controls (HC), one with low and one

with high A burden, a subject classified as mild cognitive impairment (MCI) with significant A deposition in the brain, and an Alzheimer‟s disease (AD) patient with even a higher A burden in the brain. The pattern of

18

F-NAV4694 retention replicates the sequence of 

deposition described from post-mortem studies (Braak and Braak, 1997).

60