Elsevier

Ageing Research Reviews

Volume 30, September 2016, Pages 4-16
Ageing Research Reviews

Review
Neuroimaging biomarkers in Alzheimer’s disease and other dementias

https://doi.org/10.1016/j.arr.2016.01.004Get rights and content

Highlights

  • High Aβ burden in the brain of non-demented individuals is associated with greater risk of progression to Alzheimer’s disease (AD).

  • Aβ burden is associated with cognitive performance at the early stages of AD.

  • Several hypothesis have been proposed to explain the sequence of events leading to dementia.

  • While Aβ burden is associated with Aβ1-42 levels in CSF, the association with markers of neuronal injury is less clear.

  • While Aβ imaging allows differential diagnosis between AD and FTLD, this is not possible with DLB.

Abstract

In vivo imaging of β-amyloid (Aβ) has transformed the assessment of Aβ pathology and its changes over time, extending our insight into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional or global Aβ burden in the brain. This knowledge is essential for therapeutic trial recruitment and for the evaluation of anti-Aβ treatments. Although cross sectional evaluation of Aβ 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 Aβ 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 Aβ are required to confirm this hypothesis and further elucidate the role of Aβ deposition in the course of Alzheimer’s disease.

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 β-amyloid (Aβ) 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 Aβ, (Masters et al., 1985) surrounded by reactive gliosis and dystrophic neurites. Aβ is a 4 kDa 39–43 amino acid metalloprotein derived from the proteolytic cleavage of the amyloid precursor protein (APP), by β and γ-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 Aβ leading to its progressive accumulation is central to the pathogenesis of AD (Villemagne et al., 2006). The “Aβcentric theory” (Masters et al., 2006) postulates that Aβ 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 Aβ 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).

Section snippets

Aβ imaging in Alzheimer’s disease

The development of PET tracers for Aβ deposition has allowed in vivo assessment of Aβ pathology. Since the first study published more than 10 years ago (Klunk et al., 2004), several hundreds of Aβ PET studies have been performed, improving our knowledge on the topography, timing, propagation, rates of accumulation and covariates of Aβ accumulation in the living Human brain.

On visual inspection, cortical 11C-PiB retention is usually higher in AD than in cognitively intact controls. Quantitative

Aβ deposition in non demented individuals and its relation with cognition

About 25–35% of elderly subjects performing within normal limits on cognitive tests present with high cortical 11C-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 Aβ

Relation of Aβ imaging with other biomarkers

Another rapidly growing area is the exploration of the potential association between Aβ burden as measured by PET, and CSF biomarkers of Aβ deposition or neuroimaging biomarkers of neurodegeneration (Albert et al., 2011, Blennow et al., 2007, Clark et al., 2008, de Leon et al., 2006, 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).

Aβ imaging in other neurodegenerative conditions

Aβ 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 11C-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 11C-PiB retention

Aβ 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-Aβ therapy trials and providing a means to measure their impact on Aβ burden (Ostrowitzki et al., 2011, Rinne et al., 2010). While these studies represent one of the principal applications of Aβ imaging, they have brought forward an issue that needs to be addressed, namely that the advent of novel Aβ tracers with differing pharmacological and pharmacokinetics

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 and 90% (Beach et al., 2012). While clinical criteria, together with current

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

References (231)

  • B. Dubois et al.

    Revising the definition of Alzheimer’s disease: a new lexicon

    Lancet Neurol.

    (2010)
  • P. Edison et al.

    Microglia, amyloid, and cognition in Alzheimer’s disease: an [11C](R)PK11195-PET and [11C]PIB-PET study

    Neurobiol. Dis.

    (2008)
  • A. Forsberg et al.

    PET imaging of amyloid deposition in patients with mild cognitive impairment

    Neurobiol. Aging

    (2008)
  • N. Fox

    When, where, and how does Alzheimer’s disease start?

    Lancet Neurol.

    (2012)
  • T. Grimmer et al.

    Beta amyloid in Alzheimer’s disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid

    Biol. Psychiatry

    (2009)
  • H. Hampel et al.

    Total and phosphorylated tau protein as biological markers of Alzheimer’s disease

    Exp. Gerontol.

    (2010)
  • J. Hardy

    An ‘anatomical cascade hypothesis’ for Alzheimer’s disease

    Trends Neurosci.

    (1992)
  • J. Hardy

    Amyloid, the presenilins and Alzheimer’s disease

    Trends Neurosci.

    (1997)
  • J.R. Hodges et al.

    Semantic dementia: a unique clinicopathological syndrome

    Lancet Neurol.

    (2007)
  • O. Isacson et al.

    Alzheimer’s disease and Down’s syndrome: roles of APP, trophic factors and ACh

    Trends Neurosci.

    (2002)
  • C.R. Jack et al.

    Biomarker modeling of Alzheimer’s disease

    Neuron

    (2013)
  • C.R. Jack et al.

    Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers

    Lancet Neurol.

    (2013)
  • C.R. Jack et al.

    Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade

    Lancet Neurol.

    (2010)
  • K.A. Jellinger et al.

    Prevalence and pathogenic role of cerebrovascular lesions in Alzheimer disease

    J. Neurol. Sci.

    (2005)
  • H.J. Aizenstein et al.

    Frequent amyloid deposition without significant cognitive impairment among the elderly

    Arch. Neurol.

    (2008)
  • A. Altmann et al.

    Regional brain hypometabolism is unrelated to regional amyloid plaque burden

    Brain

    (2015)
  • R.A. Armstrong et al.

    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.

    (2000)
  • R.J. Bateman et al.

    Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo

    Nat. Med.

    (2006)
  • R.J. Bateman et al.

    Clinical and biomarker changes in dominantly inherited Alzheimer’s disease

    N. Engl. J. Med.

    (2012)
  • T.G. Beach et al.

    Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005–2010

    J. Neuropathol. Exp. Neurol.

    (2012)
  • P. Bourgeat et al.

    Beta-amyloid burden in the temporal neocortex is related to hippocampal atrophy in elderly subjects without dementia

    Neurology

    (2010)
  • H. Braak et al.

    Neuropathological stageing of Alzheimer-related changes

    Acta Neuropathol. (Berl.)

    (1991)
  • H. Braak et al.

    The pathological process underlying Alzheimer’s disease in individuals under thirty

    Acta Neuropathol.

    (2011)
  • R.L. Buckner et al.

    The brain’s default network: anatomy, function, and relevance to disease

    Ann. N. Y. Acad. Sci.

    (2008)
  • R.L. Buckner et al.

    Molecular, structural, and functional characterization of Alzheimer’s disease: evidence for a relationship between default activity, amyloid, and memory

    J. Neurosci.

    (2005)
  • N.J. Cairns et al.

    Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration

    Acta Neuropathol.

    (2007)
  • V. Camus et al.

    Using PET with (18)F-AV-45 (florbetapir) to quantify brain amyloid load in a clinical environment

    Eur. J. Nucl. Med. Mol. Imaging

    (2012)
  • S.F. Carter et al.

    Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG

    J. Nucl. Med.

    (2012)
  • G. Chetelat

    Alzheimer disease: Abeta-independent processes-rethinking preclinical AD

    Nat. Rev. Neurol.

    (2013)
  • G. Chetelat et al.

    Dissociating atrophy and hypometabolism impact on episodic memory in mild cognitive impairment

    Brain

    (2003)
  • G. Chetelat et al.

    FDG-PET measurement is more accurate than neuropsychological assessments to predict global cognitive deterioration in patients with mild cognitive impairment

    Neurocase

    (2005)
  • G. Chetelat et al.

    Relationship between atrophy and beta-amyloid deposition in Alzheimer disease

    Ann. Neurol.

    (2010)
  • G. Chetelat et al.

    Larger temporal volume in elderly with high versus low beta-amyloid deposition

    Brain

    (2010)
  • G. Chetelat et al.

    Accelerated cortical atrophy in cognitively normal elderly with high beta-amyloid deposition

    Neurology

    (2012)
  • C.M. Clark et al.

    Biomarkers for early detection of Alzheimer pathology

    Neurosignals

    (2008)
  • C. Cordonnier

    Brain microbleeds

    Pract. Neurol.

    (2010)
  • Z. Cselenyi et al.

    Clinical validation of 18F-AZD4694, an amyloid-beta-specific PET radioligand

    J. Nucl. Med.

    (2012)
  • J.L. Cummings et al.

    Alzheimer’s disease drug-development pipeline: few candidates, frequent failures

    Alzheimers Res. Ther.

    (2014)
  • J.L. Cummings et al.

    Alzheimer’s disease: etiologies, pathophysiology, cognitive reserve, and treatment opportunities

    Neurology

    (1998)
  • L. Davies et al.

    A4 amyloid protein deposition and the diagnosis of Alzheimer’s disease: prevalence in aged brains determined by immunocytochemistry compared with conventional neuropathologic techniques

    Neurology

    (1988)
  • Cited by (30)

    • Brain changes associated with sleep disruption in cognitively unimpaired older adults: A short review of neuroimaging studies

      2021, Ageing Research Reviews
      Citation Excerpt :

      Of note, SDB has been related to both reduced (Huang et al., 2019; Tahmasian et al., 2016; Weng et al., 2014) and greater (Cross et al., 2018; Rosenzweig et al., 2013) MTL volume. It seems crucial to better characterize sleep parameters associated with MTL atrophy, as medial temporal areas are known to be affected by tau pathology and atrophied since AD pre-dementia stages (Braak and Braak, 1991; Villemagne and Chételat, 2016). In addition, beyond the MTL, tau pathology also affects the brainstem, which is involved in sleep-wake regulation and REM sleep generation (Horner and Peever, 2017).

    • Magnetic resonance imaging relaxation time in Alzheimer's disease

      2018, Brain Research Bulletin
      Citation Excerpt :

      Research is increasingly focusing on biomarkers for early diagnosis, disease progression monitoring and potential treatment response predictions. Neuroimaging biomarkers play a crucial role in this field, including regional structural alterations on structural magnetic resonance imaging (MRI), metabolism alterations on 18F-fluorodeoxyglucose (FDG) Positron Emission Computed Tomography (PET), detection of amyloid plaque on amyloid PET, and brain function alterations on blood oxygen level dependent (BOLD) −functional MRI (Dustin et al., 2016; Pini et al., 2016; Villemagne and Chetelat, 2016). In contrast to conventional MRI, quantitative MRI uses of MRI parameters to evaluate and determine the fundamental biologic properties of tissue.

    • Longitudinal whole-brain atrophy and ventricular enlargement in nondemented Parkinson's disease

      2017, Neurobiology of Aging
      Citation Excerpt :

      This runs counter to previous studies, where reductions in CSF α-synuclein have been shown to correlate with functional deficits in motor networks (Campbell et al., 2015) and frontal atrophy in nondemented PD (Compta et al., 2014). Interestingly, Compta et al. observed a reverse relationship in the PD-D group, where increased CSF α-synuclein was associated with atrophy in the parahippocampal gyrus and precuneus, both of which are commonly implicated in preclinical and established stages of dementia (Mak et al., 2017; Villemagne and Chételat, 2016). However, the correlations did not persist after adjusting for CSF tau, suggesting that any effects of CSF α-synuclein may be mediated through tau (Compta et al., 2014).

    • In vivo tau PET imaging in dementia: Pathophysiology, radiotracer quantification, and a systematic review of clinical findings

      2017, Ageing Research Reviews
      Citation Excerpt :

      Furthermore, the regions with the highest burden of tau in the DLB group were in the occipital visual association cortices, suggesting an atypical profile of tau progression that deviates from the classical Braak staging. Advances in neuroimaging methods and intra-individual registration across multimodal datasets have enabled the precise quantification of both subtle and widespread patterns of downstream neurodegeneration across the dementia spectrum, from normal aging to preclinical stages and established dementia (see Pini et al., 2016; Villemagne and Chételat, 2016 for a series of reviews). Briefly, there are characteristic profiles of grey and white matter changes that most likely reflect underlying differences in pathophysiology among different conditions (Mak et al., 2014; Whitwell et al., 2007).

    View all citing articles on Scopus
    View full text