G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS Maturitas xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Maturitas journal homepage: www.elsevier.com/locate/maturitas

Review

Neuroimaging in dementia Vyara Valkanova, Klaus P. Ebmeier ∗ Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK

a r t i c l e

i n f o

Article history: Available online xxx Keywords: Dementia Alzheimer’s disease Lewy body dementia Fronto-temporal dementia Vascular dementia PET

a b s t r a c t Over the last few years, advances in neuroimaging have generated biomarkers, which increase diagnostic certainty, provide valuable information about prognosis, and suggest a particular pathology underlying the clinical dementia syndrome. We aim to review the evidence for use of already established imaging modalities, along with selected techniques that have a great potential to guide clinical decisions in the future. We discuss structural, functional and molecular imaging, focusing on the most common dementias: Alzheimer’s disease, fronto-temporal dementia, dementia with Lewy bodies and vascular dementia. Finally, we stress the importance of conducting research using representative cohorts and in a naturalistic set up, in order to build a strong evidence base for translating imaging methods for a National Health Service. If we assess a broad range of patients referred to memory clinic with a variety of imaging modalities, we will make a step towards accumulating robust evidence and ultimately closing the gap between the dramatic advances in neurosciences and meaningful clinical applications for the maximum benefit of our patients. © 2014 Elsevier Ireland Ltd. All rights reserved.

Contents 1. 2.

3.

4.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Structural imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Alzheimer’s dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Fronto-temporal dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Dementia with Lewy bodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. Vascular dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Functional and molecular imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Alzheimer’s dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1.1. Perfusion SPECT and FDG-PET imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1.2. Amyloid imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Fronto-temporal dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3. Dementia with Lewy bodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4. Vascular dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Provenance and peer review . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00 00

Abbreviations: 123-I-FP-CIT, 123-I labelled ioflupane (DatSCAN® ); AD, Alzheimer’s dementia; bv-FTD, behavioural variant fronto-temporal dementia; CERAD, consortium to establish a registry for Alzheimer’s disease; CT, (X-ray) computed tomography; FTD, fronto-temporal dementia; LBD, dementia with Lewy bodies; lv-P, palogopenic variant PPA; MCI, mild cognitive impairment; MRI, magnetic resonance imaging; MTA, medial temporal lobe atrophy; NICE, National Institute for Health and Care Excellence; nv-PPA, non-fluent PPA; PET, positron emission tomography; PiB, Pittsburgh compound B; PPA, primary progressive aphasia; SPECT, single photon emission computed tomography; sv-PPA, semantic variant PPA; VBM, voxel-based morphometry; VD, vascular dementia; WMH, white matter magnetic resonance hyperintensities. ∗ Corresponding author. Tel.: +44 1865 226469; fax: +44 1865 793101. E-mail address: [email protected] (K.P. Ebmeier). http://dx.doi.org/10.1016/j.maturitas.2014.02.016 0378-5122/© 2014 Elsevier Ireland Ltd. All rights reserved.

Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

2

1. Introduction

an MTA score of 0 converted to AD, compared with more than 75% with baseline MTA score of 3.

Over the last few years, advances in neuroimaging have generated biomarkers, which increase diagnostic certainty, provide valuable information about prognosis, and suggest a particular pathology underlying the clinical dementia syndrome. This is an advance over the role imaging plays in simply excluding rare causes of dementia, such as mass lesions [1,2]. Although the use of specific biomarkers is generally limited to research studies, it is likely to translate into clinical practice with increased standardization and access to biomarkers. We aim to review the evidence for established imaging modalities, along with selected techniques that have the potential to guide clinical decisions in the future, focusing on the most common dementias: Alzheimer’s disease (AD), fronto-temporal dementia (FTD), dementia with Lewy bodies (DLB) and vascular dementia (VD). Discussion of other dementia syndromes and the theory behind the various imaging modalities is beyond the scope of this review. 2. Structural imaging 2.1. Alzheimer’s dementia In many centres, X-ray computed tomography is the standard investigation to exclude any space occupying and mass lesions. In fact, some authors have used this technique to estimate hippocampal atrophy and thus provide added information supporting the diagnosis of Alzheimer’s dementia [3]. However, the structural imaging technique most widely used in clinical research is T1-weighted magnetic resonance imaging (MRI). The most characteristic feature of AD is early, localized medial temporal lobe atrophy (MTA) affecting primarily the hippocampus and entorhinal cortex [4–8]. MRI evidence of disproportionate MTA has been incorporated into revised diagnostic criteria as a topographical marker of downstream neuronal injury [1,2]. In clinical practice visual assessment is used most often. One of the more widely validated rating scales is the Scheltens’ MTA rating scale [9], which assesses hippocampal atrophy according to the width of the choroid fissure, width of the temporal horn, and height of the hippocampus, using a 0–4 severity scale. Visual inspection differentiates mild AD from normal ageing with a sensitivity and specificity of 80–85% [9–11]. The specificity of MTA decreases with age because of age-related hippocampal atrophy [12]. In research studies, mainly volumetric techniques are used and they appear to correlate well with both neuropathological disease progression [13–15] and the degree of cognitive impairment [16,17]. Although MTA has been found in other dementias, including FTD, VD and PD, it is less severe there than in AD when matched for clinical severity [18,19]. Hippocampal atrophy develops gradually starting about 5 years before diagnosis of AD, while 3 years prior to diagnosis hippocampal volumes are reduced by 10% [19]. In atypical forms of AD, in younger patients, or early in the disease course it is possible to have greater atrophy of the parietal lobes and less MTA [20–25]. A rating scale based on the degree of widening of the posterior cingulate, parietal and parieto-occipital sulci has been developed [26]. Particularly in individuals with MCI, structural imaging provides valuable information about prognosis. Although the significant overlap in volumetric measures between patients with MCI and controls sets some limitations, a recent meta-analysis of 6 VBM studies found that decreased grey matter in the left hippocampus and parahippocampal gyrus was associated with conversion from MCI to AD [27]. Further, Lehmann et al. [28] found that the probability of converting from MCI to AD increased with greater baseline MTA scores; e.g. after three years, fewer than 40% of patients with Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

2.2. Fronto-temporal dementia FTD encompasses a heterogeneous group of conditions and can be broadly divided into behavioural variant fronto-temporal dementia (bv-FTD) and primary progressive aphasia (PPA). Bv-FTD is associated with predominant atrophy in the frontal and paralimbic areas, including the anterior cingulate cortex, as well as orbitofrontal and medial frontal cortices and subcortical structures [29–32]. Ratings of orbitofrontal atrophy in conjunction with an executive function test classified 92% of patients correctly into bvFTD or AD [33]. PPA is itself categorized into three clinical phenotypes: semantic variant PPA (sv-PPA), non-fluent PPA (nv-PPA) and logopenic variant PPA (lv-PPA). In semantic variant PPA, there is typically atrophy of the anterior and inferior temporal lobes, including the fusiform gyrus, while the non-fluent variant PPA is characterized by perisylvian atrophy and involvement of the anterior insula [31,34,35]. The logopenic variant PPA is commonly associated with Alzheimer’s type pathology and the atrophy involves the posterior temporal cortex and inferior parietal lobule [36]. In PPA the atrophy is more often asymmetrical, with the left side being more affected [31]. In addition to distinct patterns of regional atrophy that are associated with each clinical phenotype, rate of atrophy was found to be twice as great in FTD and SD, compared with AD [37]. However, in the early stages, functional imaging may be more useful because the structural scan can be normal [32,38]. The most recently published guidelines for FTD proposed, in addition to a clinical diagnosis, evidence for abnormalities on either structural or functional brain imaging as a criterion for establishing a diagnosis of ‘probable’ FTD [36,39]. 2.3. Dementia with Lewy bodies DLB is associated with diffuse atrophy, which is greater than in controls, but less than in patients with AD. Relatively focused dorsal meso-pontine grey matter atrophy, with a relative sparing of the medial temporal lobes, supports a clinical diagnosis of DLB rather than AD [40–42]. 2.4. Vascular dementia Evidence from structural neuroimaging is mandatory for diagnosing vascular dementia (VD), with MRI being the preferred modality. Small-vessel disease is the commonest cause of VD [43], although large artery ischaemic disease may lead to VD, as well. Small-vessel disease is usually defined as lesions involving >25% of the white matter. Signs of small vessel disease on MRI include white matter magnetic resonance hyperintensities (WMH), recent small subcortical infarcts, lacunes, prominent perivascular spaces, cerebral microbleeds and atrophy. Their defining features are summarized in an excellent paper aiming to provide standards for reporting vascular changes on neuroimaging (STRIVE [44]). Although white matter changes can be detected on X-ray CT scans, the tissue contrast tends to be discrete, and this method is less sensitive than MRI. White matter lesions appear as bilateral, mostly symmetrical hyperintensities on T2-weighted MRI. They become more common with advancing age and are found in 10–90% of cognitively normal elderly depending on the study. Although WMH are strongly associated with vascular risk factors, as well as predicting an increased risk of stroke and dementia [45], they are clinically and pathologically heterogeneous [46]. WMH are not specific to vascular dementia and can be found in a variety of other conditions Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

including leucodystrophies, leucoencephalopathies, inflammatory conditions (multiple sclerosis) and infections [47]. The relationship between vascular damage and cognition is not fully understood, but it is increasingly recognized that whether cognitive deficits develop in the presence of vascular damage depends on the balance between factors conferring resilience (education, premorbid IQ) and risk (age, vascular risk factors, and lifestyle). However, as WMH become more extensive they are more likely to be clinically significant [48]. The severity of white matter disease can be graded visually using established scales such as the Fazekas scale [49]. Large irregular periventricular or confluent deep WMH (Fazekas grade 3) are characteristic of VD. Silent infarcts and lacunes are also common in healthy elderly people (between 20 and 50%), but they are associated with an increased risk of future dementia and stroke [50]. Although MRI is more sensitive than CT for detecting ischaemic injury, it was negative in up to 30% of patients with symptomatic lacunar stroke syndromes, suggesting that it cannot detect all infarcts [51]. Up to 65% of patients with VD and 10–20% of healthy elderly have microbleeds which on T2*-gradient echo image appear as focal hypointensities (typically 2–5 mm, but up to 10 mm) [52,53]. Microbleeds related to amyloid angiopathy most commonly have cortico-subcortical distribution, while microbleeds associated with hypertension are located centrally. The two conditions often coexist [54–56]. Patients with vascular dementia also have global atrophy, probably due to subclinical diffuse ischaemia, or focal atrophy corresponding to an area of infarction [57,58]. Finally, it is increasingly recognized that mixed pathologies are common. VD and AD share risk factors [59,60] and can have a synergistic relationship [61]. This implies that even when extensive vascular damage is suggested by a structural scan, the presence of mixed pathology should be considered.

3. Functional and molecular imaging 3.1. Alzheimer’s dementia 3.1.1. Perfusion SPECT and FDG-PET imaging Functional and molecular imaging has an important role in the neuroimaging of dementias. Changes in function and molecular composition of brain tissue typically precede atrophy detectable by structural imaging. Jack et al. [62] have suggested the dynamic biomarkers model of AD: at first markers of amyloidosis became positive, followed by markers of cortical hypometabolism on FDGPET, and last, markers of brain atrophy. The characteristic AD patterns of abnormal brain perfusion or metabolism are found in the association cortex while primary sensory-motor cortex is relatively preserved [63], with a ratio of association cortex activity over primary sensory-motor cortex activity being suggested as a diagnostic index for AD [64]. Early in the disease course, SPECT and FDG-PET show reduction in posterior cingulate and precuneus perfusion or metabolism, followed by bilateral posterior temporo-parietal reductions and involvement of the frontal areas in advanced disease [63–65]. The findings in functional scans correlate with CSF biomarkers [66], as well as with cognitive measures [67]. Perfusion abnormalities in AD-related regions are predictive of conversion from MCI to AD [68–70]. Hypometabolism was found even in cognitively normal individuals at increased risk of AD [71,72]. An autopsy-confirmed study found that SPECT improves the accuracy of the diagnoses, with a positive scan in patients with possible AD increasing the likelihood of pathologically confirmed AD from 67% to 84% [73]. A recent meta-analysis of 11 studies using SPECT and 20 studies using PET found pooled sensitivity of 80% and specificity of 85% for SPECT to differentiate patients with AD from controls, while PET showed pooled sensitivity of 90% and specificity Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

3

of 89% [74]. These results suggest that PET is more sensitive than clinical assessment in detecting AD (90% vs 81%), while both SPECT and PET are more specific than clinical criteria (85% and 89% vs 70%) [2]. However a prospective community-based study of 102 individuals with early-onset dementia found PET to have considerably lower sensitivity and specificity for AD (78% and 81%, respectively) than reported in the meta-analysis [75]. Although the results could be explained by sample characteristics (e.g. early-onset dementia), or the lack of pathological confirmation, this study highlights the significance of ecologically more valid research with ‘real’ world samples. It is also important to investigate the added value of imaging markers above a standard work-up, as well as how imaging markers perform in combination. A study of 154 memory clinic patients found that combined imaging (PiB and FDG-PET) led to change in 23% of the initial clinical diagnoses, while the diagnosis established after PET remained unchanged in 96% of cases after two years. Combined PiB and FDG-PET contributed to diagnosis in 104 cases, followed by PiB only (29 cases) and FDG-PET only (11 cases) [76]. While both PET and SPECT are good enough to assist in the differential diagnosis of dementia [63], SPECT is currently more widely available. This has been translated into guidelines, with NICE stating that ‘HMPAO-SPECT should be used to help differentiate between AD, VD and FTD if the diagnosis is in doubt’ [77]. 18F-FDGPET is included as a topographical biomarker of neuronal injury in the revised international diagnostic criteria for AD [1,2]. A recent review concluded that although studies suggest superiority of PET over SPECT, the evidence base for this is quite limited [78]. 3.1.2. Amyloid imaging Amyloid imaging provides direct evidence of the presence of Alzheimer’s type pathology. A␤ plaque markers become positive years before clinical symptoms and reach a plateau by the time these appear [62]. The best characterized amyloid PET tracer is Pittsburgh compound B (PiB) which is labelled with C-11 and binds mostly to fibrillar ␤-amyloid. Amyloid PET with PiB has very high sensitivity for detecting amyloid deposition. More than 90% of patients with AD show increased cortical binding [79], and false–negative cases are reported only rarely [80]. As a result, it has been included in the revised guidelines as a pathophysiological marker of AD [1,2]. Its positive predictive value is low, as some healthy elderly people show higher cortical binding. The reported proportions vary, but frequencies of 15–30% are most commonly reported. A␤ deposition is affected by the imaging method, the definition of what constitutes a positive result, and by the two main risk factors for AD, namely age and ApoE genotype [81,82]. The frequency of amyloid-positive scans increased from 0% below the age of 50 to 30% at age 80 [83]. Non-carriers of the allele were less than half as likely to have a positive scan as ApoE4 carriers (21% vs 49%) [84]. Independently of the effect of ApoE4, maternal familial history of AD has also been associated with higher cortical binding [85,86]. A major disadvantage for the clinical application of PiB PET is the short half-life of C-11, which limits its use to centres with an on-site cyclotron. However, over the last years tracers labelled with 18F such as Flumetamol, Florbetapir and Florbetaben have been developed. 18F has a longer half-life (110 min), allowing central production and distribution, which is already making amyloid imaging more widely available [87]. Clinical trials have demonstrated that the new 18F-compounds have properties similar to PiB. Comparative studies within the same subjects show excellent correlation between PiB and each of them [88–90], as well as similar effect sizes for distinguishing AD from healthy controls [89,91]. In a prospective phase 3 study with Florbetapir, the PET scan results of 59 patients were dichotomized and Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

4

compared to their dichotomized CERAD neuritic amyloid plaque scores obtained from autopsy within 1 to 2 years of PET imaging. The study included healthy subjects, as well as patients with AD, VD and FTD, with a varying degree of cognitive impairment and found a sensitivity and specificity of PET for detection of moderate to frequent plaques of 92–96% and 100%, respectively [92]. Amyloid imaging has very good predictive value for conversion to AD in patients with MCI, as demonstrated in studies with up to 3 years follow-up [93–95]. Negative scans in individuals with MCI do not discount progression, as other neurodegenerative diseases without amyloidosis, such as FTD, cannot be excluded. Amyloid imaging is also excellent in differentiating AD from non-amyloid dementias such as FTD [96–98], and there is no uptake in most cases with PD dementia [99]. However positive scans are common in DLB [98,100,101] which is consistent with pathological findings [102]. This implies that amyloid imaging may be most useful in atypical presentations or in early-onset dementia, when the prevalence of AD equals that of FTD and the frequency of amyloid deposition in controls is low. Positive scan in cases with extensive vascular damage can confirm the presence of mixed pathology, which would have implications for treatment [103]. For instance, 31% of cases who met DSM-IV criteria for VD and had extensive WMH without large-vessel stroke or macro-haemorrhage were PiB positive [104]. 3.2. Fronto-temporal dementia The patterns seen on functional imaging in FTD tend to be similar to those seen on structural scans, but they are visible earlier in the disease course [105]. A study of 134 patients with suspected FTD found that SPECT/PET increased sensitivity of consensus criteria from 36.5% to 90.5%, when clinical diagnosis after 2 years was used as a gold standard [106]. Another study with pathologically confirmed diagnoses of FTD or AD also showed that PET increases diagnostic accuracy beyond clinical assessment [107]. Amyloid imaging in FTD confirms the lack of amyloid deposition in this condition, with patients showing values of cortical PiB retention close to those found in controls [108]. 3.3. Dementia with Lewy bodies There are few publications for perfusion imaging in DLB, but the typical pattern involves occipital reductions, including the primary visual cortices [109–111]. Although parieto-temporal reductions are observed in both AD and DLB, occipital hypometabolism differentiated between the two conditions with 90% sensitivity and 71–80% specificity [65,109]. Amyloid imaging in DLB shows levels of cortical PiB retention similar to those observed in AD, with higher occipital retention [100,112]. The most reliable imaging biomarker of DLB is 123-I-FP-CIT (ioflupane) SPECT of the dopamine transporter. Ioflupane shows asymmetrical (anterior-posterior) striatal uptake in DLB, with larger decrease of uptake in putamen than in caudate nucleus. In contrast, age-related nigro-striatal degeneration affects both the putamen and caudate nucleus, and the putamen-to-caudate ratio is preserved [113]. Normal ioflupane scans in patients with DLB are not common, but have been reported; it is possible that these patients have exclusive cortical involvement with relative sparing of basal ganglia [114]. Compared with autopsy results, dopamine transporter imaging is sensitive and specific (88% and 100%, respectively), whereas clinical diagnosis had a sensitivity of 75% and specificity of only 42% [115]. Abnormal ioflupane scans may be particularly valuable in the group of patients with possible DLB, as this seems to be an unstable diagnosis. A multi-centre study found that only 41% of patients with ‘possible’ DLB have this diagnosis at 12 months Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

follow up, compared with 93% of patients diagnosed with ‘probable’ DLB; scan results can help to classify them correctly [116]. Ioflupane scans have also an important role in the differential diagnosis of DLB from AD [117]. A recent meta-analysis of four 123I-FP-CIT SPECT studies, including a total of 419 patients, reported a pooled sensitivity of 86.5% and a specificity of 93.6% for the differentiation of DLB from non-DLB, predominantly AD [118]. However abnormal ioflupane binding is less reliable in differentiating DLB from FTD, because about 30% of FTD patients were have positive ioflupane scans [119]. Ioflupane scans also cannot differentiate between syndromes characterized by dopaminergic loss, including DLB, Parkinson’s disease, multiple system atrophy or progressive supranuclear palsy. Reflecting the increasing evidence for the clinical utility of ioflupane SPECT, it has been incorporated in revised diagnostic criteria for DLB [120]. It has also been recommended by NICE to help with the diagnosis of DLB when the diagnosis is in doubt [77].

3.4. Vascular dementia In patients with VD and no infarcts on structural imaging, functional scans typically show scattered areas of reduced perfusion or metabolism. The lesions are often multiple, asymmetrical or localized in ‘watershed’ regions of the brain [63]. Compared with AD, in VD there is more pronounced hypometabolism in subcortical areas and primary sensorimotor cortex whereas the association areas are typically less affected [121].

4. Conclusion At present, neuroimaging is most likely to be used in the differential diagnosis of dementia, as well as to aid in establishing a prognosis. When disease-modifying treatments become available, this is likely to change, and imaging methods can be used to screen patients, with function-based and ligand-based techniques having a particular role in the detection of early changes. The future of neuroimaging will involve incorporating new modalities into routine clinical practice. However, introducing imaging methods as routine in a National Health Service requires a strong evidence base for their clinical utility and added value. When considering the clinical utility of neuroimaging, it is important to realize that mixed pathologies commonly co-exist in the same individual, and there is a substantial overlap of pathological changes even with health. This will impose limitations on the diagnostic performance of any test and implies that a combination of imaging biomarkers will be most useful. Therefore, studies investigating how different modalities perform in combination will be particularly useful. In addition, strong evidence can only come from studies using representative cohorts and conducted in a naturalistic clinical environment. Currently, studies are conducted mainly in highly specialized centres and often include subjects from clear-cut diagnostic groups, which limits the generalizability of results. Establishment of pilot services is crucial, because each clinical context is associated with characteristic variables, such as demographics of the sample, a priori probability of dementia types, frequency of atypical presentations, and clinical expertise of health care professionals; all these factors have an impact on the value that imaging methods add above routine clinical assessment. If we assess a broad range of patients referred to memory clinic with a variety of imaging modalities, we will make a step towards accumulating reliable evidence and ultimately closing the gap between the dramatic advances in neurosciences and meaningful clinical applications for the maximum benefit of our patients.

Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

Contributors Vyara Valkanova – first draft (part), revisions (whole). Klaus P. Ebmeier – first draft (part), revisions (whole) Competing interest Vyara Valkanova – none; Klaus P. Ebmeier – reports consultation fees received from Lily in relation to Amyvid TM Funding Klaus P. Ebmeier – UK Medical Research Council (G1001354), the Gordon Edward Small’s Charitable Trust (SC008962), and the HDH Wills 1965 Charitable Trust. Provenance and peer review Commissioned and externally peer reviewed. References [1] Dubois B, Feldman HH, Jacova C, Cummings JL, Dekosky ST, Barberger-Gateau P, et al. Revising the definition of Alzheimer’s disease: a new lexicon. Lancet Neurol 2010;9(11):1118–27. [2] McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack Jr CR, Kawas CH, et al. 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 2011;7(3):263–9. [3] Jobst KA, Smith AD, Szatmari M, Molyneux A, Esiri ME, King E, et al. Detection in life of confirmed Alzheimer’s disease using a simple measurement of medial temporal lobe atrophy by computed tomography. Lancet 1992;340(8829):1179–83. [4] Chan D, Fox NC, Scahill RI, Crum WR, Whitwell JL, Leschziner G, et al. Patterns of temporal lobe atrophy in semantic dementia and Alzheimer’s disease. Ann Neurol 2001;49(4):433–42. [5] Scheltens P, Fox N, Barkhof F, De Carli C. Structural magnetic resonance imaging in the practical assessment of dementia: beyond exclusion. Lancet Neurol 2002;1(1):13–21. [6] Killiany RJ, Hyman BT, Gomez-Isla T, Moss MB, Kikinis R, Jolesz F, et al. MRI measures of entorhinal cortex vs hippocampus in preclinical AD. Neurology 2002;58(8):1188–96. [7] Frisoni GB, Fox NC, Jack Jr CR, Scheltens P, Thompson PM. The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol 2010;6(2):67–77. [8] van der Flier WM, Pijnenburg YA, Fox NC, Scheltens P. Early-onset versus lateonset Alzheimer’s disease: the case of the missing APOE varepsilon4 allele. Lancet Neurol 2011;10(3):280–8. [9] Scheltens P, Leys D, Barkhof F, Huglo D, Weinstein HC, Vermersch P, et al. Atrophy of medial temporal lobes on MRI in probable Alzheimer’s disease and normal ageing: diagnostic value and neuropsychological correlates. J Neurol Neurosurg Psychiatry 1992;55(10):967–72. [10] Duara R, Loewenstein DA, Potter E, Appel J, Greig MT, Urs R, et al. Medial temporal lobe atrophy on MRI scans and the diagnosis of Alzheimer disease. Neurology 2008;71(24):1986–92. [11] Burton EJ, Barber R, Mukaetova-Ladinska EB, Robson J, Perry RH, Jaros E, et al. Medial temporal lobe atrophy on MRI differentiates Alzheimer’s disease from dementia with Lewy bodies and vascular cognitive impairment: a prospective study with pathological verification of diagnosis. Brain 2009;132(Pt 1):195–203. [12] van de Pol LA, Hensel A, Barkhof F, Gertz HJ, Scheltens P, van der Flier WM. Hippocampal atrophy in Alzheimer disease: age matters. Neurology 2006;66(2):236–8. [13] Gosche KM, Mortimer JA, Smith CD, Markesbery WR, Snowdon DA. Hippocampal volume as an index of Alzheimer neuropathology: findings from the Nun Study. Neurology 2002;58(10):1476–82. [14] Jack Jr CR, Dickson DW, Parisi JE, Xu YC, Cha RH, O’Brien PC, et al. Antemortem MRI findings correlate with hippocampal neuropathology in typical aging and dementia. Neurology 2002;58(5):750–7. [15] Vemuri P, Whitwell JL, Kantarci K, Josephs KA, Parisi JE, Shiung MS, et al. Antemortem MRI based STructural Abnormality iNDex (STAND)-scores correlate with postmortem Braak neurofibrillary tangle stage. Neuroimage 2008;42(2):559–67. [16] Hua X, Leow AD, Lee S, Klunder AD, Toga AW, Lepore N, et al. 3D characterization of brain atrophy in Alzheimer’s disease and mild cognitive impairment using tensor-based morphometry. Neuroimage 2008;41(1):19–34. [17] Vemuri P, Wiste HJ, Weigand SD, Shaw LM, Trojanowski JQ, Weiner MW, et al. MRI and CSF biomarkers in normal, MCI, and AD subjects: diagnostic discrimination and cognitive correlations. Neurology 2009;73(4):287–93.

Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

5

[18] van de Pol L, Gertz HJ, Scheltens P, Wolf H. Hippocampal atrophy in subcortical vascular dementia. Neurodegener Dis 2011;8(6):465–9. [19] Johnson KA, Fox NC, Sperling RA, Klunk WE. Brain imaging in Alzheimer disease. Cold Spring Harb Perspect Med 2012;2(4):a006213. [20] Galton CJ, Patterson K, Xuereb JH, Hodges JR. Atypical and typical presentations of Alzheimer’s disease: a clinical, neuropsychological, neuroimaging and pathological study of 13 cases. Brain 2000;123(Pt 3):484–98. [21] Frisoni GB, Geroldi C, Binetti G, Zanetti O. The heterogeneity and natural history of mild cognitive impairment. Arch Neurol 2005;62(1):163, author reply-4. [22] Ishii K, Kawachi T, Sasaki H, Kono AK, Fukuda T, Kojima Y, et al. Voxel-based morphometric comparison between early- and late-onset mild Alzheimer’s disease and assessment of diagnostic performance of z score images. AJNR Am J Neuroradiol 2005;26(2):333–40. [23] Frisoni GB, Pievani M, Testa C, Sabattoli F, Bresciani L, Bonetti M, et al. The topography of grey matter involvement in early and late onset Alzheimer’s disease. Brain 2007;130(Pt 3):720–30. [24] Karas G, Scheltens P, Rombouts S, van Schijndel R, Klein M, Jones B, et al. Precuneus atrophy in early-onset Alzheimer’s disease: a morphometric structural MRI study. Neuroradiology 2007;49(12):967–76. [25] Josephs KA, Whitwell JL, Duffy JR, Vanvoorst WA, Strand EA, Hu WT, et al. Progressive aphasia secondary to Alzheimer disease vs FTLD pathology. Neurology 2008;70(1):25–34. [26] Koedam EL, Lehmann M, van der Flier WM, Scheltens P, Pijnenburg YA, Fox N, et al. Visual assessment of posterior atrophy development of a MRI rating scale. Eur Radiol 2011;21(12):2618–25. [27] Ferreira LK, Diniz BS, Forlenza OV, Busatto GF, Zanetti MV. Neurostructural predictors of Alzheimer’s disease: a meta-analysis of VBM studies. Neurobiol Aging 2011;32(10):1733–41. [28] Lehmann M, Koedam EL, Barnes J, Bartlett JW, Barkhof F, Wattjes MP, et al. Visual ratings of atrophy in MCI: prediction of conversion and relationship with CSF biomarkers. Neurobiol Aging 2013;34(1):73–82. [29] Lindberg O, Ostberg P, Zandbelt BB, Oberg J, Zhang Y, Andersen C, et al. Cortical morphometric subclassification of frontotemporal lobar degeneration. AJNR Am J Neuroradiol 2009;30(6):1233–9. [30] Davatzikos C, Resnick SM, Wu X, Parmpi P, Clark CM. Individual patient diagnosis of AD and FTD via high-dimensional pattern classification of MRI. Neuroimage 2008;41(4):1220–7. [31] Seelaar H, Rohrer JD, Pijnenburg YA, Fox NC, van Swieten JC. Clinical, genetic and pathological heterogeneity of frontotemporal dementia: a review. J Neurol Neurosurg Psychiatry 2011;82(5):476–86. [32] Piguet O, Hornberger M, Mioshi E, Hodges JR. Behavioural-variant frontotemporal dementia: diagnosis, clinical staging, and management. Lancet Neurol 2011;10(2):162–72. [33] Hornberger M, Savage S, Hsieh S, Mioshi E, Piguet O, Hodges JR. Orbitofrontal dysfunction discriminates behavioral variant frontotemporal dementia from Alzheimer’s disease. Dement Geriatr Cogn Disord 2010;30(6):547–52. [34] Rohrer JD, Warren JD, Modat M, Ridgway GR, Douiri A, Rossor MN, et al. Patterns of cortical thinning in the language variants of frontotemporal lobar degeneration. Neurology 2009;72(18):1562–9. [35] Kipps CM, Davies RR, Mitchell J, Kril JJ, Halliday GM, Hodges JR. Clinical significance of lobar atrophy in frontotemporal dementia: application of an MRI visual rating scale. Dement Geriatr Cogn Disord 2007;23(5):334–42. [36] Gorno-Tempini ML, Dronkers NF, Rankin KP, Ogar JM, Phengrasamy L, Rosen HJ, et al. Cognition and anatomy in three variants of primary progressive aphasia. Ann Neurol 2004;55(3):335–46. [37] Krueger CE, Dean DL, Rosen HJ, Halabi C, Weiner M, Miller BL, et al. Longitudinal rates of lobar atrophy in frontotemporal dementia, semantic dementia, and Alzheimer’s disease. Alzheimer Dis Assoc Disord 2010;24(1):43–8. [38] Perry RJ, Graham A, Williams G, Rosen H, Erzinclioglu S, Weiner M, et al. Patterns of frontal lobe atrophy in frontotemporal dementia: a volumetric MRI study. Dement Geriatr Cogn Disord 2006;22(4):278–87. [39] Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain 2011;134(Pt 9):2456–77. [40] Whitwell JL, Weigand SD, Shiung MM, Boeve BF, Ferman TJ, Smith GE, et al. Focal atrophy in dementia with Lewy bodies on MRI: a distinct pattern from Alzheimer’s disease. Brain 2007;130(Pt 3):708–19. [41] Vemuri P, Simon G, Kantarci K, Whitwell JL, Senjem ML, Przybelski SA, et al. Antemortem differential diagnosis of dementia pathology using structural MRI: differential-STAND. Neuroimage 2011;55(2):522–31. [42] Kantarci K, Ferman TJ, Boeve BF, Weigand SD, Przybelski S, Vemuri P, et al. Focal atrophy on MRI and neuropathologic classification of dementia with Lewy bodies. Neurology 2012;79(6):553–60. [43] Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 2010;9(7):689–701. [44] Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, Frayne R, et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol 2013;12(8):822–38. [45] Debette S, Markus HS. The clinical importance of white matter hyperintensities on brain magnetic resonance imaging: systematic review and meta-analysis. BMJ 2010;341:c3666. [46] Gouw AA, Seewann A, van der Flier WM, Barkhof F, Rozemuller AM, Scheltens P, et al. Heterogeneity of small vessel disease: a systematic review of MRI and histopathology correlations. J Neurol Neurosurg Psychiatry 2011;82(2):126–35.

Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7 6

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

[47] Harper L, Barkhof F, Scheltens P, Schott JM, Fox NC. An algorithmic approach to structural imaging in dementia. J Neurol Neurosurg Psychiatry 2013. [48] Dufouil C, Godin O, Chalmers J, Coskun O, MacMahon S, Tzourio-Mazoyer N, et al. Severe cerebral white matter hyperintensities predict severe cognitive decline in patients with cerebrovascular disease history. Stroke 2009;40(6):2219–21. [49] Fazekas F, Chawluk JB, Alavi A, Hurtig HI, Zimmerman RA. MR signal abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. AJR Am J Roentgenol 1987;149(2):351–6. [50] Vermeer SE, Longstreth Jr WT, Koudstaal PJ. Silent brain infarcts: a systematic review. Lancet Neurol 2007;6(7):611–9. [51] Doubal FN, Dennis MS, Wardlaw JM. Characteristics of patients with minor ischaemic strokes and negative MRI: a cross-sectional study. J Neurol Neurosurg Psychiatry 2011;82(5):540–2. [52] Poels MM, Vernooij MW, Ikram MA, Hofman A, Krestin GP, van der Lugt A, et al. Prevalence and risk factors of cerebral microbleeds: an update of the Rotterdam scan study. Stroke 2010;41(10 Suppl.):S103–6. [53] Cordonnier C, van der Flier WM, Sluimer JD, Leys D, Barkhof F, Scheltens P. Prevalence and severity of microbleeds in a memory clinic setting. Neurology 2006;66(9):1356–60. [54] Lee SH, Kim SM, Kim N, Yoon BW, Roh JK. Cortico-subcortical distribution of microbleeds is different between hypertension and cerebral amyloid angiopathy. J Neurol Sci 2007;258(1-2):111–4. [55] Yamada S, Saiki M, Satow T, Fukuda A, Ito M, Minami S, et al. Periventricular and deep white matter leukoaraiosis have a closer association with cerebral microbleeds than age. Eur J Neurol 2012;19(1):98–104. [56] Cordonnier C, van der Flier WM. Brain microbleeds and Alzheimer’s disease: innocent observation or key player? Brain 2011;134(Pt 2):335–44. [57] Logue MW, Posner H, Green RC, Moline M, Cupples LA, Lunetta KL, et al. Magnetic resonance imaging-measured atrophy and its relationship to cognitive functioning in vascular dementia and Alzheimer’s disease patients. Alzheimers Dement 2011;7(5):493–500. [58] Seo SW, Lee JM, Im K, Park JS, Kim SH, Kim ST, et al. Cardiovascular risk factors cause cortical thinning in cognitively impaired patients: relationships among cardiovascular risk factors, white matter hyperintensities, and cortical atrophy. Alzheimer Dis Assoc Disord 2012;26(2):106–12. [59] Kivipelto M, Helkala EL, Laakso MP, Hanninen T, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ 2001;322(7300):1447–51. [60] Dichgans M, Zietemann V. Prevention of vascular cognitive impairment. Stroke 2012;43(11):3137–46. [61] Leys D, Henon H, Mackowiak-Cordoliani MA, Pasquier F. Poststroke dementia. Lancet Neurol 2005;4(11):752–9. [62] Jack Jr CR, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol 2010;9(1):119–28. [63] Ebmeier KP. Is there Still a Place for Perfusion SPECT in the Diagnosis of Dementia? Open Nucl Med J 2010;2:40–5. [64] Herholz K, Nordberg A, Salmon E, Perani D, Kessler J, Mielke R, et al. Impairment of neocortical metabolism predicts progression in Alzheimer’s disease. Dement Geriatr Cogn Disord 1999;10(6):494–504. [65] Mosconi L, Tsui WH, Herholz K, Pupi A, Drzezga A, Lucignani G, et al. Multicenter standardized 18F-FDG PET diagnosis of mild cognitive impairment, Alzheimer’s disease, and other dementias. J Nucl Med 2008;49(3):390–8. [66] Ceravolo R, Borghetti D, Kiferle L, Tognoni G, Giorgetti A, Neglia D, et al. CSF phosporylated TAU protein levels correlate with cerebral glucose metabolism assessed with PET in Alzheimer’s disease. Brain Res Bull 2008;76(1-2):80–4. [67] Arlt S, Brassen S, Jahn H, Wilke F, Eichenlaub M, Apostolova I, et al. Association between FDG uptake, CSF biomarkers and cognitive performance in patients with probable Alzheimer’s disease. Eur J Nucl Med Mol Imaging 2009;36(7):1090–100. [68] Drzezga A, Grimmer T, Riemenschneider M, Lautenschlager N, Siebner H, Alexopoulus P, et al. Prediction of individual clinical outcome in MCI by means of genetic assessment and (18)F-FDG PET. J Nucl Med 2005;46(10):1625–32. [69] Borroni B, Anchisi D, Paghera B, Vicini B, Kerrouche N, Garibotto V, et al. Combined 99mTc-ECD SPECT and neuropsychological studies in MCI for the assessment of conversion to AD. Neurobiol Aging 2006;27(1):24–31. [70] Nobili F, De Carli F, Frisoni GB, Portet F, Verhey F, Rodriguez G, et al. SPECT predictors of cognitive decline and Alzheimer’s disease in mild cognitive impairment. J Alzheimers Dis 2009;17(4):761–72. [71] Reiman EM, Chen K, Alexander GE, Caselli RJ, Bandy D, Osborne D, et al. Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer’s dementia. Proc Natl Acad Sci U S A 2004;101(1):284–9. [72] Mosconi L, Brys M, Switalski R, Mistur R, Glodzik L, Pirraglia E, et al. Maternal family history of Alzheimer’s disease predisposes to reduced brain glucose metabolism. Proc Natl Acad Sci U S A 2007;104(48):19067–72. [73] Jagust W, Thisted R, Devous Sr MD, Van Heertum R, Mayberg H, Jobst K, et al. SPECT perfusion imaging in the diagnosis of Alzheimer’s disease: a clinicalpathologic study. Neurology 2001;56(7):950–6. [74] Bloudek LM, Spackman DE, Blankenburg M, Sullivan SD. Review and metaanalysis of biomarkers and diagnostic imaging in Alzheimer’s disease. J Alzheimers Dis 2011;26(4):627–45. [75] Panegyres PK, Rogers JM, McCarthy M, Campbell A, Wu JS. Fluorodeoxyglucose-positron emission tomography in the differential diagnosis of early-onset dementia: a prospective, community-based study. BMC Neurol 2009;9:41.

Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

[76] Ossenkoppele R, Prins ND, Pijnenburg YA, Lemstra AW, van der Flier WM, Adriaanse SF, et al. Impact of molecular imaging on the diagnostic process in a memory clinic. Alzheimers Dement 2013;9(4):414–21. [77] National Institute of Health and Care Excellence. A NICE–SCIE guideline on supporting people with dementia and their carers in health and social care (clinical guideline number 42). London: The British Psychological Society & Gaskell; 2011. [78] Davison CM, O’Brien JT. A comparison of FDG-PET and blood flow SPECT in the diagnosis of neurodegenerative dementias: a systematic review. Int J Geriatr Psychiatry 2013. [79] Rabinovici GD, Jagust WJ. Amyloid imaging in aging and dementia: testing the amyloid hypothesis in vivo. Behav Neurol 2009;21(1):117–28. [80] Cairns NJ, Ikonomovic MD, Benzinger T, Storandt M, Fagan AM, Shah AR, et al. Absence of Pittsburgh compound B detection of cerebral amyloid beta in a patient with clinical, cognitive, and cerebrospinal fluid markers of Alzheimer disease: a case report. Arch Neurol 2009;66(12):1557–62. [81] Chetelat G, La Joie R, Villain N, Perrotin A, de La Sayette V, Eustache F, et al. Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer’s disease. Neuroimage Clin 2013;2:356–65. [82] Mielke MM, Wiste HJ, Weigand SD, Knopman DS, Lowe VJ, Roberts RO, et al. Indicators of amyloid burden in a population-based study of cognitively normal elderly. Neurology 2012;79(15):1570–7. [83] Morris JC, Roe CM, Xiong C, Fagan AM, Goate AM, Holtzman DM, et al. APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann Neurol 2010;67(1):122–31. [84] Rowe CC, Ellis KA, Rimajova M, Bourgeat P, Pike KE, Jones G, et al. Amyloid imaging results from the Australian imaging, biomarkers and lifestyle (AIBL) study of aging. Neurobiol Aging 2010;31(8):1275–83. [85] Reiman EM, Chen K, Liu X, Bandy D, Yu M, Lee W, et al. 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 2009;106(16):6820–5. [86] Mosconi L, Rinne JO, Tsui WH, Berti V, Li Y, Wang H, et al. Increased fibrillar amyloid-{beta} burden in normal individuals with a family history of lateonset Alzheimer’s. Proc Natl Acad Sci U S A 2010;107(13):5949–54. [87] Herholz K, Ebmeier K. Clinical amyloid imaging in Alzheimer’s disease. Lancet Neurol 2011;10(7):667–70. [88] Vandenberghe R, Van Laere K, Ivanoiu A, Salmon E, Bastin C, Triau E, et al. 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann Neurol 2010;68(3):319–29. [89] Villemagne VL, Mulligan RS, Pejoska S, Ong K, Jones G, O’Keefe G, et al. Comparison of 11C-PiB and 18F-florbetaben for Abeta imaging in ageing and Alzheimer’s disease. Eur J Nucl Med Mol Imaging 2012;39(6): 983–9. [90] Landau SM, Breault C, Joshi AD, Pontecorvo M, Mathis CA, Jagust WJ, et al. Amyloid-beta imaging with Pittsburgh compound B and florbetapir: comparing radiotracers and quantification methods. J Nucl Med 2013;54(1):70–7. [91] Wolk DA, Zhang Z, Boudhar S, Clark CM, Pontecorvo MJ, Arnold SE. Amyloid imaging in Alzheimer’s disease: comparison of florbetapir and Pittsburgh compound-B positron emission tomography. J Neurol Neurosurg Psychiatry 2012;83(9):923–6. [92] Clark CM, Pontecorvo MJ, Beach TG, Bedell BJ, Coleman RE, Doraiswamy PM, et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-beta plaques: a prospective cohort study. Lancet Neurol 2012;11(8):669–78. [93] Forsberg A, Engler H, Almkvist O, Blomquist G, Hagman G, Wall A, et al. PET imaging of amyloid deposition in patients with mild cognitive impairment. Neurobiol Aging 2008;29(10):1456–65. [94] Okello A, Koivunen J, Edison P, Archer HA, Turkheimer FE, Nagren K, et al. Conversion of amyloid positive and negative MCI to AD over 3 years: an 11CPIB PET study. Neurology 2009;73(10):754–60. [95] Nordberg A, Carter SF, Rinne J, Drzezga A, Brooks DJ, Vandenberghe R, et al. A European multicentre PET study of fibrillar amyloid in Alzheimer’s disease. Eur J Nucl Med Mol Imaging 2013;40(1):104–14. [96] Rabinovici GD, Furst AJ, O’Neil JP, Racine CA, Mormino EC, Baker SL, et al. 11CPIB PET imaging in Alzheimer disease and frontotemporal lobar degeneration. Neurology 2007;68(15):1205–12. [97] Rabinovici GD, Rosen HJ, Alkalay A, Kornak J, Furst AJ, Agarwal N, et al. Amyloid vs FDG-PET in the differential diagnosis of AD and FTLD. Neurology 2011;77(23):2034–42. [98] Rowe CC, Ng S, Ackermann U, Gong SJ, Pike K, Savage G, et al. Imaging betaamyloid burden in aging and dementia. Neurology 2007;68(20):1718–25. [99] Maetzler W, Reimold M, Liepelt I, Solbach C, Leyhe T, Schweitzer K, et al. [11C]PIB binding in Parkinson’s disease dementia. Neuroimage 2008;39(3):1027–33. [100] Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, et al. Microglia, amyloid, and cognition in Alzheimer’s disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol Dis 2008;32(3):412–9. [101] Villemagne VL, Ong K, Mulligan RS, Holl G, Pejoska S, Jones G, et al. Amyloid imaging with (18)F-florbetaben in Alzheimer disease and other dementias. J Nucl Med 2011;52(8):1210–7. [102] Harding AJ, Halliday GM. Cortical Lewy body pathology in the diagnosis of dementia. Acta Neuropathol 2001;102(4):355–63. [103] Vandenberghe R, Adamczuk K, Dupont P, Laere KV, Chetelat G. Amyloid PET in clinical practice: Its place in the multidimensional space of Alzheimer’s disease. Neuroimage Clin 2013;2:497–511.

Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

G Model MAT-6131; No. of Pages 7

ARTICLE IN PRESS V. Valkanova, K.P. Ebmeier / Maturitas xxx (2014) xxx–xxx

[104] Lee JH, Kim SH, Kim GH, Seo SW, Park HK, Oh SJ, et al. Identification of pure subcortical vascular dementia using 11C-Pittsburgh compound B. Neurology 2011;77(1):18–25. [105] Rohrer JD, Rosen HJ. Neuroimaging in frontotemporal dementia. Int Rev Psychiatry 2013;25(2):221–9. [106] Mendez MF, Shapira JS, McMurtray A, Licht E, Miller BL. Accuracy of the clinical evaluation for frontotemporal dementia. Arch Neurol 2007;64(6):830–5. [107] Foster NL, Heidebrink JL, Clark CM, Jagust WJ, Arnold SE, Barbas NR, et al. FDG-PET improves accuracy in distinguishing frontotemporal dementia and Alzheimer’s disease. Brain 2007;130(Pt 10):2616–35. [108] Engler H, Santillo AF, Wang SX, Lindau M, Savitcheva I, Nordberg A, et al. In vivo amyloid imaging with PET in frontotemporal dementia. Eur J Nucl Med Mol Imaging 2008;35(1):100–6. [109] Minoshima S, Foster NL, Sima AA, Frey KA, Albin RL, Kuhl DE. Alzheimer’s disease versus dementia with Lewy bodies: cerebral metabolic distinction with autopsy confirmation. Ann Neurol 2001;50(3):358–65. [110] Ishii K, Soma T, Kono AK, Sofue K, Miyamoto N, Yoshikawa T, et al. Comparison of regional brain volume and glucose metabolism between patients with mild dementia with lewy bodies and those with mild Alzheimer’s disease. J Nucl Med 2007;48(5):704–11. [111] Sinha N, Firbank M, O’Brien JT. Biomarkers in dementia with Lewy bodies: a review. Int J Geriatr Psychiatry 2012;27(5):443–53. [112] Gomperts SN, Rentz DM, Moran E, Becker JA, Locascio JJ, Klunk WE, et al. Imaging amyloid deposition in Lewy body diseases. Neurology 2008;71(12):903–10.

Please cite this article in press as: Valkanova http://dx.doi.org/10.1016/j.maturitas.2014.02.016

V,

7

[113] Tatsch K, Poepperl G. Nigrostriatal dopamine terminal imaging with dopamine transporter SPECT: an update. J Nucl Med 2013;54(8):1331–8. [114] Siepel FJ, Rongve A, Buter TC, Beyer MK, Ballard CG, Booij J, et al. (123I)FP-CIT SPECT in suspected dementia with Lewy bodies: a longitudinal case study. BMJ Open 2013;3(4.). [115] Walker Z, Jaros E, Walker RW, Lee L, Costa DC, Livingston G, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry 2007;78(11):1176–81. [116] O’Brien JT, McKeith IG, Walker Z, Tatsch K, Booij J, Darcourt J, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry 2009;194(1):34–9. [117] Warr L, Walker Z. Identification of biomarkers in Lewy-body disorders. Q J Nucl Med Mol Imaging 2012;56(1):39–54. [118] Papathanasiou ND, Boutsiadis A, Dickson J, Bomanji JB. Diagnostic accuracy of 123 I-FP-CIT (DaTSCAN) in dementia with Lewy bodies: a meta-analysis of published studies. Parkinsonism Relat Disord 2012;18(3):225–9. [119] Morgan S, Kemp P, Booij J, Costa DC, Padayachee S, Lee L, et al. Differentiation of frontotemporal dementia from dementia with Lewy bodies using FP-CIT SPECT. J Neurol Neurosurg Psychiatry 2012;83(11):1063–70. [120] McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology 2005;65(12):1863–72. [121] Heiss WD, Zimmermann-Meinzingen S. PET imaging in the differential diagnosis of vascular dementia. J Neurol Sci 2012;322(1-2):268–73.

Ebmeier

KP.

Neuroimaging

in

dementia.

Maturitas

(2014),

Neuroimaging in dementia.

Over the last few years, advances in neuroimaging have generated biomarkers, which increase diagnostic certainty, provide valuable information about p...
1MB Sizes 0 Downloads 3 Views