touchNEUROLOGY touchNEUROLOGY
Alzheimer's Disease & Dementia
Read Time: 9 mins

Positron Emission Tomography Imaging in Dementia

Copy Link
Published Online: Jun 4th 2011 European Neurological Review, 2008;3(2):109-112 DOI: http://doi.org/10.17925/ENR.2008.03.02.109
Authors: Karl Herholz
Quick Links:
Article
Article Information
Article:

Positron emission tomography (PET) utilises biologically active molecules in micromolar or nanomolar concentrations that have been labelled with short-lived positron-emitting isotopes. The physical characteristics of the isotopes and the molecular specificity of labelled molecules, combined with the high detection efficacy of modern PET scanners, provide sensitivity for in vivo measurement that is several orders of magnitude higher than with the other imaging techniques. While the very short half-lives of 15O (two minutes) and 11C (20 minutes) limit their use to fully equipped PET centres with a cyclotron and radiopharmaceutical laboratory, 18F-labelled tracers (half-life 110 minutes) can be produced in specialised cyclotron centres for regional distribution to hospitals running a PET scanner only.
Glucose is the main energy supply for the brain. Its metabolism maintains ion gradients and glutamate turnover and is closely coupled to neuronal function at rest and during functional activation.1 Its measurement by 18F-2- fluoro-2-deoxy-D-glucose (FDG) is based on phosphorylation of the tracer by hexokinase, which is the pivotal first step of that metabolic pathway. Typically, PET images are obtained 30–60 minutes after tracer injection, when FDG uptake is approximately proportional to glucose metabolism, and actual measurement times can be as short as five to 10 minutes. Under resting conditions (awake, but without external stimulation), normal grey matter displays two to four times higher glucose metabolism than white matter. There is a moderate reduction of cerebral glucose metabolism with age, mainly affecting the frontal association cortex.2 Significant regional reductions of glucose metabolism indicate impairment of synaptic function, and the technique is therefore applicable to all types of dementia.
There is growing interest in imaging amyloid deposits, the pathological hallmarks of Alzheimer’s disease (AD). There are now several PET tracers for in vivo amyloid imaging that allow longitudinal studies of amyloid deposition to clarify whether amyloid deposition is a cause or consequence in the pathophysiology of AD.3,4 Furthermore, the degeneration of major neurotransmitter systems can be demonstrated in vivo by using appropriate PET tracers. Impairment of the cholinergic and dopaminergic neurotransmission is of particular diagnostic interest. By these means, PET can detect early stages and differentiate between various types of dementia, and also monitor progression and the effect of therapeutic intervention.

Alzheimer’s Disease

Over more than 20 years, multiple studies have demonstrated that glucose metabolism and blood flow are impaired in temporal–parietal association cortices, with the angular gyrus usually being located at the centre of the metabolic impairment.5 The frontal association cortex may also be involved, but variably and usually to a lesser degree. The affected association cortices are those that become myelinated last during brain maturation and are also prone to cortical amyloid deposition.6 There may be a distinct hemispheric asymmetry, which usually corresponds to the predominant cognitive deficits (language impairment in the dominant and visuospatial disorientation in the sub-dominant hemisphere). In contrast to other dementia types, in AD glucose metabolism in the basal ganglia, primary motor and visual cortex and cerebellum is usually well preserved. This pattern generally reflects AD clinical symptoms, with impairment of memory and associative thinking, including higher-order sensory processing and planning of action, but relative preservation of primary motor and sensory function.
Voxel-based comparisons with normal reference samples clearly show that the posterior cingulate gyrus and the precuneus are also impaired at an early stage of AD.7 This is usually not directly obvious by mere inspection of FDG PET scans because metabolism in that area is above the cortical average in normal brain at a resting state,8 and with the beginning of impairment it returns to the level of the surrounding cortex but does not stand out as a hypometabolic lesion. Thus, this important diagnostic sign is easily missed by standard visual interpretation of FDG PET brain scans. On the background of sufficient numbers of FDG PET scans in normal controls it is becoming increasingly standard to base the interpretation of patient studies not merely on visual interpretation of the tracer distribution, but also on quantitative mapping with reference to an appropriate normal sample.9–15
The pattern of metabolic impairment can vary considerably among individual patients. It is typically more pronounced in patients with onset of the disease before 75 years of age than in patients who develop AD at later ages.16,17 The degree of metabolic impairment in frontal association cortices varies and is typically seen in more severely affected patients and in patients who are carriers of the apolipoprotein E4 allele.18 There are AD patients with very pronounced focal impairment of occipito-temporo-parietal association areas, which may correspond to the clinical syndrome of posterior cortical atrophy.19
Normal ageing and AD cause significant atrophy, including in those brain areas that are also hypometabolic on FDG PET.20 Regional atrophy will cause underestimation of regional glucose metabolism on PET scans due to partial volume effects, depending on scanner resolution. The magnitude of this effect has been estimated in several studies. They concluded that the metabolic reduction in posterior neocortical association areas cannot be explained completely by atrophy-related partial volume effects,21 while there is typically more severe hippocampal atrophy on MRI than apparent hypometabolism on PET scans.22
Longitudinal studies have demonstrated that the severity and extent of metabolic impairment in temporal and parietal cortex increases with dementia progression, and frontal reductions become more evident.23,24 The reduction of metabolism is in the order of 16–19% over three years in association cortices, which contrasts with an absence of significant decline in normal control subjects.25 Asymmetrical metabolic impairment and associated predominance of language or visuospatial impairment tends to persist during progression.26,27 Based on these observations, FDG PET can serve as a biomarker in therapeutic trials.28,29 When monitoring change due to disease progression over one year using standard neuropsychological testing by mini-mental state examination (MMSE) and Alzheimer’s Disease Assessment Scale Cognitive Subscale (ADAS-cog), one typically obtains coefficients of variance (COV) around 100% for these changes, whereas PET measurements are subject to about 50% COV, thus doubling t-scores and reducing the required sample size by about 75% at the same study power.30,31

Mild Cognitive Impairment

Mild cognitive impairment (MCI) can progress to AD, but MCI patients may also remain stable over many years, develop mixed or vascular dementia or even remit. There is considerable interest in whether PET would allow a reliable diagnosis of AD at this early stage before clinical manifestation of dementia. Impairment of regional cerebral metabolic rate of glucose consumption (rCMRglc) has been observed in individuals at high risk of AD due to family history of AD and possession of the apolipoprotein E (ApoE) ε4 allele,32,33 even at a completely asymptomatic stage, and this abnormality is seen decades before the likely onset of dementia.34 In middle-aged and elderly asymptomatic ApoE ε4-positive individuals, temporo-parietal and posterior cingulate rCMRglc declines by about 2% per year.35
The first study noting the predictive power of posterior cingulate metabolism in patients with severe memory deficits for predicting progression was performed by Minoshima et al. in 1997.7 This observation was followed by several studies indicating a high predictive power with sensitivity and specificity above 80% for prediction of rapid progression,36–41 which is clearly superior to ApoE ε4 testing. Mesial temporal metabolic impairment has been observed as a general feature in patients with memory impairment,42–44 and hippocampal metabolic impairment appears to predict development of MCI in cognitively normal subjects.45

Amyloid

The first tracer used to label Aβ-amyloid selectively in vivo was 11C-labelled thioflavin analogue, named for convenience Pittsburgh compound B (11CPIB).46 Dynamic scanning provides quantitation of binding potential, while a relatively simple and practical method of quantitating uptake in a clinical setting is based on scans obtained approximately 60 minutes after intravenous injection of the tracer, with the cerebellum as an unaffected reference region.47 Since its introduction, this tracer has been used by multiple research groups and has been consistently proved to provide high sensitivity in detecting amyloid plaques and vascular amyloid in the human brain in vivo.48–50 In normal subjects, some unspecific binding is observed, mainly in white matter – which probably is due to the compound’s lipophilicity – whereas in patients with AD, specific binding mostly in the frontal, temporal and parietal association cortices typically exceeds twice the background level. Follow-up studies with 11C-PIB in AD indicate that there is no further increase of tracer uptake during progression of the disease. 51
When used in tracer amounts, 11C-PIB is specific for amyloid and does not bind to neurofibrillary tangles.52 Its clinical specificity for AD is currently being studied and it has been shown that patients with frontotemporal dementia or semantic dementia do not show increased 11C-PIB binding.53,54 Findings in patients with MCI are heterogeneous: some show intensely increased binding, like AD patients, while others are within normal limits. First results from follow-up studies of up to 18 months indicate that some patients with increased binding may indeed progress to AD.55 As to be expected from previous post mortem findings, increased amyloid binding has also been observed in some cognitively normal elderly volunteers.56 It remains to be determined whether increased 11C-PIB uptake in elderly individuals who are cognitively normal represent false-positive findings or a pre-symptomatic stage of AD that could become clinically manifest up to a decade later.
An alternative 11C-labelled tracer is the stilbene derivative SB-13,57 which appears to have similar properties to 11C-PIB without obvious advantages. 18F-labelled tracers could have the practical advantage of becoming more widely available from commercial providers, even for PET centres without their own cyclotron. Recently, a series of stilbene derivatives have been labelled with 18F and evaluated as amyloid ligands, demonstrating satisfactory specific amyloid binding to non-specific binding in AD for compound AV-45.58 A related compound, 18F-BAY94-9172 (formerly known as AV-1), demonstrated 100% sensitivity and 90% specificity for AD in 15 patients with mild AD, 15 healthy elderly controls and five individuals with frontotemporal lobar degeneration.59
Another tracer, the anthracyclin derivative 18F-FDDNP, has different characteristics and binds to neurofibrillary tangles as well as amyloid plaques with less affinity and specificity than PIB.60,61 Binding to amyloid competes with some non-steroidal antiphlogistics.62 While providing a generally somewhat less favourable signal-to-background ratio, it also detects neurofibrillary tangles, which appear in the hippocampus in the earliest stages of AD63 and differentiates persons with mild cognitive impairment from those with Alzheimer’s disease and those with no cognitive impairment.64

Cholinergic System

Impairment of cholinergic neurotransmission is characteristic of AD, and may be even more severe in dementia with Lewy bodies (DLB).65 Tracers for imaging nicotinic receptors, which are of particular interest because of their largely pre-synaptic location, are 11C-nicotine66 and 18F-A-85380.67 Reduced binding has been observed in the cortex with 11C-nicotine,66,68 and mostly in the thalamus with 18F-A-8538069 in AD.
The most important degrading enzyme for acetylcholine in the human cortex is acetylcholine esterase (AChE), which is present in cholinergic axons and relatively few cholinoceptive neurons. As the cholinergic axons degenerate, AChE activity is also reduced.70 Labelled analogues of acetylcholine that are also substrates for AChE can be used to measure and image its activity in vivo. These are 11C-N-methyl-4-piperidyl-acetate (MP4A, also known as AMP),71 which is 94% specific for AChE in human brain, and 11C-N-methyl-4-piperidyl-propionate (MP4P, or PMP).72 There have been several studies measuring AChE activity, all of which found a reduction of cortical activity in AD73–76 and even in MCI,77 most severely affecting the temporal cortex. This technique has also been used to measure drug-induced AChE inhibition in AD patients, which for all currently available cholinesterase inhibitors at standard clinical dose is in the range of 30–40%.78–80

Fronto-temporal Lobar Degeneration

This group of diseases comprises fronto-temporal dementia (FTD) as the most frequent manifestation with mostly behavioural symptoms and also more focal degenerative processes such as progressive aphasia, progressive apraxia and semantic dementia with various histopathological features (including Pick bodies in some cases) but absence of amyloid plaques.81 In FTD, cerebral glucose metabolism is impaired mostly in the frontal cortex, especially in the mesial frontal cortex.82 Frontolateral and anterior temporal cortices are often also severely impaired, and this impairment is related to the clinical symptoms of aphasia or semantic memory deficits and may be very asymmetric.83,84 Milder metabolic impairment often involves posterior association cortices as well.85 The regional pattern of predominantly frontal impairment usually allows clear distinction from AD, although there may be overlap as AD can involve frontal regions as well and FTD may not spare the temporo-parietal cortex. It has recently been shown in a series of 45 patients with pathologically confirmed diagnosis that FDG PET can discriminate FTD from AD with more than 85% sensitivity and specificity.86
Semantic dementia, which is clinically characterised by a failure of semantic memory, typically shows severe metabolic impairment of the anterior parts of the left temporal lobe,87,88 often also extending into basolateral parts of the frontal lobe.89 The main metabolic deficits in progressive aphasia are typically located in the language areas of the left hemisphere.90 There is considerable overlap of metabolic deficits with FTD, especially as these diseases progress.

Dementia with Lewy Bodies

DLB is clinically characterised by fluctuating consciousness, possible parkinsonian motor features and impairment of visual perception, including hallucinations. The latter are the likely correlate of the reduction of glucose metabolism in the primary visual cortex that has been described with FDG PET in DLB, in addition to an impairment of posterior association areas, like in AD.91 In contrast, metabolic activity in the primary visual cortex is usually well preserved in AD, but in practice the distinction may be difficult as metabolic activity in that area is subject to considerable variability and depends on examination conditions (eyes open or eyes closed). A recent analysis of diagnostic discrimination reported a relatively low accuracy of 73% for discrimination between DLB and AD.92
A more reliable feature to differentiate DLB from AD is the impairment of dopamine synthesis and transport,93 which can be assessed with 18Ffluorodopa94 and tracers for dopamine transporters.95 A deficit of dopamine synthesis similar to PD has been found in DLB, even at a stage when parkinsonism may not yet be prominent,93,96 while it is normal in patients with AD. In contrast to the cholinergic impairment, which is severe in DLB but only mild in PD without dementia, the dopaminergic deficit does not appear to be related to dementia.97

Summary

Brain PET using FDG is a firmly established technique for demonstration of regional functional impairment in neurodegenerative disease. AD is associated with typical regional impairments of posterior cortical association areas, including the posterior cingulate gyrus, which are closely related to clinical symptoms. Thus, FDG PET facilitates very early diagnosis before clinical manifestation of dementia and can provide monitoring of progression and treatment effects. DLB shows metabolic impairment similar to AD but involves additional metabolic impairment of primary visual cortex. Predominant impairment of frontal and anterior temporal regions is seen in FTD: primary progressive aphasia and semantic dementia. New perspectives for specific molecular imaging have been opened by tracers for imaging amyloid that appear to be very sensitive even in the detection of pre-clinical AD cases. As amyloid deposition is also found in a proportion of cognitively normal elderly people, confirmation of its pathological significance remains to be demonstrated by long-term follow-up studies. Tracers for imaging nicotinic receptors and for measuring local acetylcholine esterase activity demonstrate cholinergic impairment in MCI, AD and DLB. The impairment of dopamine neurotransmission that is characteristic of DLB can be demonstrated by 18F-fluorodopa PET. ■

References

  1. Sokoloff L, Relation between physiological function and energy metabolism in the central nervous system, J Neurochem, 1977;29:13–26.
  2. Kuhl DE, Metter EJ, Riege WH, Phelps ME, Effects of human aging on patterns of local cerebral glucose utilization determined by the [18F]fluorodeoxyglucose method, J Cereb Blood Flow Metab, 1982;2:163–71.
  3. Hardy J, Selkoe DJ, The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics, Science, 2002;297:353–6.
  4. de la Torre JC, Pathophysiology of neuronal energy crisis in Alzheimer’s disease, Neurodegener Dis, 2008;5:126–32.
  5. Herholz K, PET studies in dementia, Ann Nucl Med, 2003;17: 79–89.
  6. Bartzokis G, Lu PH, Mintz J, Human brain myelination and amyloid beta deposition in Alzheimer’s disease, Alzheimers Dement, 2007;3:122–5.
  7. Minoshima S, Giordani B, Berent S, et al., Metabolic reduction in the posterior cingulate cortex in very early Alzheimer’s disease, Ann Neurol, 1997;42:85–94.
  8. Gusnard DA, Raichle ME, Raichle ME, Searching for a baseline: functional imaging and the resting human brain, Nat Rev Neurosci, 2001;2:685–94.
  9. Herholz K, Salmon E, Perani D, et al., Discrimination between Alzheimer Dementia and Controls by Automated Analysis of Multicenter FDG PET, NeuroImage, 2002;17:302–16.
  10. Burdette JH, Minoshima S, Van der Borght T, et al., Alzheimer disease: improved visual interpretation of PET images by using three-dimensional stereotaxic surface projections, Radiology, 1996;198:837–43.
  11. Ishii K, Willoch F, Minoshima S, et al., Statistical brain mapping of 18F-FDG PET in Alzheimer’s disease: validation of anatomic standardization for atrophied brains, J Nucl Med, 2001;42: 548–57.
  12. Azari NP, Pettigrew KD, Schapiro MB, et al., Early detection of Alzheimer's disease: a statistical approach using positron emission tomographic data, J Cereb Blood Flow Metab, 1993;13:438–47.
  13. Kippenhan JS, Barker WW, Nagel J, et al., Neural-network classification of normal and Alzheimer’s disease subjects using high-resolution and low-resolution PET cameras, J Nucl Med, 1994;35:7–15.
  14. Higdon R, Foster NL, Koeppe RA, et al., A comparison of classification methods for differentiating fronto-temporal dementia from Alzheimer’s disease using FDG-PET imaging, Stat Med, 2004;23:315–26.
  15. Ishii K, Kono AK, Sasaki H, et al., Fully automatic diagnostic system for early- and late-onset mild Alzheimer’s disease using FDG PET and 3D-SSP, Eur J Nucl Med Mol Imaging, 2006;33: 575–83.
  16. Sakamoto S, Ishii K, Sasaki M, et al., Differences in cerebral metabolic impairment between early and late onset types of Alzheimer’s disease, J Neurol Sci, 2002;200:27–32.
  17. Haense C, Herholz K, Heiss WD, Validation of an automated FDG PET analysis to discriminate patients with Alzheimer’s disease from normal subjects, J Nucl Med, 2008;49(Suppl. 1):34P.
  18. Mosconi L, Herholz K, Prohovnik I, et al., Metabolic interaction between ApoE genotype and onset age in Alzheimer's disease: implications for brain reserve, J Neurol Neurosurg Psychiatry, 2005;76:15–23.
  19. Nestor PJ, Caine D, Fryer TD, et al., The topography of metabolic deficits in posterior cortical atrophy (the visual variant of Alzheimer’s disease) with FDG-PET, J Neurol Neurosurg Psychiatry, 2003;74:1521–9.
  20. Fox NC, Schott JM, Imaging cerebral atrophy: normal ageing to Alzheimer’s disease, Lancet, 2004;363:392–4.
  21. Meltzer CC, Zubieta JK, Brandt J, et al., Regional hypometabolism in Alzheimer’s disease as measured by positron emission tomography after correction for effects of partial volume averaging, Neurology, 1996;47:454–61.
  22. Chetelat G, Desgranges B, Landeau B, et al., Direct voxel-based comparison between grey matter hypometabolism and atrophy in Alzheimer’s disease, Brain, 2008;131:60–71.
  23. Jagust WJ, Friedland RP, Budinger TF, et al., Longitudinal studies of regional cerebral metabolism in Alzheimer’s disease, Neurology, 1988;38:909–12.
  24. Mielke R, Herholz K, Grond M, et al., Clinical deterioration in probable Alzheimer’s disease correlates with progressive metabolic impairment of association areas, Dementia, 1994;5: 36–41.
  25. Smith GS, de Leon MJ, George AE, et al., Topography of crosssectional and longitudinal glucose metabolic deficits in Alzheimer's disease. Pathophysiologic implications, Arch Neurol, 1992;49:1142–50.
  26. Grady CL, Haxby JV, Schlageter NL, et al., Stability of metabolic and neuropsychological asymmetries in dementia of the Alzheimer type, Neurology, 1986;36:1390–92.
  27. Haxby JV, Grady CL, Koss E, et al., Longitudinal study of cerebral metabolic asymmetries and associated neuropsychological patterns in early dementia of the Alzheimer type, Arch Neurol, 1990;47:753–60.
  28. Heiss WD, Kessler J, Mielke R, et al., Long-term effects of phosphatidylserine, pyritinol, and cognitive training in Alzheimer’s disease. A neuropsychological, EEG, and PET investigation, Dementia, 1994;5:88–98.
  29. Kadir A, Andreasen N, Almkvist O, et al., Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer’s disease, Ann Neurol, 2008;63:621–31.
  30. Alexander GE, Chen K, Pietrini P, et al., Longitudinal PET Evaluation of Cerebral Metabolic Decline in Dementia: A Potential Outcome Measure in Alzheimer’s Disease Treatment Studies, Am J Psychiatry, 2002;159:738–45.
  31. Hirono N, Hashimoto M, Ishii K, et al., One-year change in cerebral glucose metabolism in patients with Alzheimer’s disease, J Neuropsychiatry Clin Neurosci, 2004;16:488–92.
  32. Small GW, Mazziotta JC, Collins MT, et al., Apolipoprotein E type 4 allele and cerebral glucose metabolism in relatives at risk for familial Alzheimer disease, JAMA, 1995;273:942–7.
  33. Reiman EM, Caselli RJ, Yun LS, et al., Preclinical evidence of Alzheimer's disease in persons homozygous for the epsilon 4 allele for apolipoprotein E, N Engl J Med, 1996;334:752–8.
  34. Reiman EM, Chen K, Alexander GE, 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;101:284–9.
  35. Small GW, Ercoli LM, Silverman DH, et al., Cerebral metabolic and cognitive decline in persons at genetic risk for Alzheimer’s disease, Proc Natl Acad Sci U S A, 2000;97:6037–42.
  36. Herholz K, Nordberg A, Salmon E, et al., Impairment of neocortical metabolism predicts progression in Alzheimer’s disease, Dement Geriatr Cogn Disord, 1999;10:494–504.
  37. Berent S, Giordani B, Foster N, et al., Neuropsychological function and cerebral glucose utilization in isolated memory impairment and Alzheimer’s disease, J Psychiatr Res, 1999;33:7–16.
  38. Arnaiz E, Jelic V, Almkvist O, et al., Impaired cerebral glucose metabolism and cognitive functioning predict deterioration in mild cognitive impairment, Neuroreport, 2001;12:851–5.
  39. Chetelat G, Desgranges B, de la Sayette V, et al., Mild cognitive impairment: Can FDG-PET predict who is to rapidly convert to Alzheimer’s disease?, Neurology, 2003;60:1374–7.
  40. Drzezga A, Grimmer T, Riemenschneider M, et al., Prediction of individual clinical outcome in MCI by means of genetic assessment and (18)F-FDG PET, J Nucl Med, 2005;46:1625–32.
  41. Anchisi D, Borroni B, Franceschi M, et al., Heterogeneity of brain glucose metabolism in mild cognitive impairment and clinical progression to Alzheimer disease, Arch Neurol, 2005;62:1728–33.
  42. Heiss WD, Pawlik G, Holthoff V, et al., PET correlates of normal and impaired memory functions, Cerebrovasc Brain Metab Rev, 1992;4:1–27.
  43. Nestor PJ, Fryer TD, Smielewski P, Hodges JR, Limbic hypometabolism in Alzheimer’s disease and mild cognitive impairment, Ann Neurol, 2003;54:343–51.
  44. Mosconi L, Tsui WH, DeSanti S, et al. Reduced Hippocampal metabolism in MCI and AD: Automated FDG-PET Image Analysis, Neurology, 2005;64:1860–67.
  45. de Leon MJ, Convit A, Wolf OT, et al., Prediction of cognitive decline in normal elderly subjects with 2-F-18-fluoro-2-deoxy-Dglucose positron-emission tomography (FDG PET), Proc Natl Acad Sci U S A, 2001;98:10966–71.
  46. William E, Klunk H, Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B, Ann Neurol, 2004;55:306–19.
  47. Price JC, Klunk WE, Lopresti BJ, et al., Kinetic modeling of amyloid binding in humans using PET imaging and Pittsburgh Compound- B, J Cereb Blood Flow Metab, 2005;25:1528–47.
  48. Edison P, Archer HA, Hinz R, et al., Amyloid, hypometabolism, and cognition in Alzheimer disease: an [11C]PIB and [18F]FDG PET study, Neurology, 2007;68:501–8.
  49. Kemppainen NM, Aalto S, Wilson IA, et al., Voxel-based analysis of PET amyloid ligand [11C]PIB uptake in Alzheimer disease, Neurology, 2006;67:1575–80.
  50. Nordberg A, PET imaging of amyloid in Alzheimer’s disease, Lancet Neurol, 2004;3:519–27.
  51. Engler H, Forsberg A, Almkvist O, et al., Two-year follow-up of amyloid deposition in patients with Alzheimer’s disease, Brain, 2006;129:2856–66.
  52. Klunk WE, Lopresti B, Nebes RD, et al., Development and Application of beta-Amyloid Imaging Agents in Alzheimer's Disease. In: Herholz K, Perani D, Morris CM (eds), The Dementias: Early Diagnosis and Evaluation, New York: Dekker, 2006.
  53. Rabinovici GD, Furst AJ, O’Neil JP, et al., 11C-PIB PET imaging in Alzheimer disease and frontotemporal lobar degeneration, Neurology, 2007;68:1205–12.
  54. Drzezga A, Grimmer T, Henriksen G, et al., Imaging of amyloid plaques and cerebral glucose metabolism in semantic dementia and Alzheimer’s disease, NeuroImage, 2008;39:619–33.
  55. Forsberg A, Engler H, Almkvist O, et al., PET imaging of amyloid deposition in patients with mild cognitive impairment, Neurobiol Aging, 2008;29:1456–65.
  56. Aizenstein HJ, Nebes RD, Saxton JA, et al., Amyloid Deposition is Frequent and Often is Not Associated With Significant Cognitive Impairment in The Elderly, Arch Neurol, in press.
  57. Verhoeff NP, Wilson AA, Takeshita S, et al., In Vivo Imaging of Alzheimer Disease {beta}-Amyloid With [11C]SB-13 PET, Am J Geriatr Psychiatry, 2004;12:584–95.
  58. Wong D, Rosenberg P, Zhou Y, et al., In vivo imaging of amyloid deposition in Alzheimer’s disease using the novel radioligand [18F] AV-45, J Nucl Med Meeting Abstracts, 2008;49:214P-c.
  59. Rowe CC, Ackerman U, Browne W, et al., Imaging of amyloid [beta] in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: proof of mechanism, Lancet Neurol, 2008;7:129–35.
  60. Shoghi-Jadid K, Small GW, Agdeppa ED, et al., Localization of neurofibrillary tangles and beta-amyloid plaques in the brains of living patients with Alzheimer disease, Am J Geriatr Psychiatry, 2002;10:24–35.
  61. Noda A, Murakami Y, Nishiyama S, et al., Amyloid imaging in aged and young macaques with [11C]PIB and [18F]FDDNP, Synapse, 2008;62:472–5.
  62. Agdeppa ED, Kepe V, Petri A, et al., In vitro detection of (S)- naproxen and ibuprofen binding to plaques in the Alzheimer's brain using the positron emission tomography molecular imaging probe 2-(1-{6-[(2-[18F]fluoroethyl)(methyl)amino]-2- naphthyl}ethylidene)malononitrile, Neuroscience, 2003;117: 723–30.
  63. Shin J, Lee SY, Kim SH, et al., Multitracer PET imaging of amyloid plaques and neurofibrillary tangles in Alzheimer’s disease, NeuroImage, 2008;43:236–44.
  64. Small GW, Kepe V, Ercoli LM, et al., PET of brain amyloid and tau in mild cognitive impairment, N Engl J Med, 2006;355:2652–63.
  65. Perry EK, Irving D, Kerwin JM, et al., Cholinergic transmitter and neurotrophic activities in Lewy body dementia: similarity to Parkinson’s and distinction from Alzheimer disease, Alzheimer Dis Assoc Disord, 1993;7:69–79.
  66. Nordberg A, Lundqvist H, Hartvig P, et al., Kinetic analysis of regional (S)(-)11C-nicotine binding in normal and Alzheimer brains—in vivo assessment using positron emission tomography, Alzheimer Dis Assoc Disord, 1995;9:21–7.
  67. Bottlaender M, Valette H, Roumenov D, et al., Biodistribution and radiation dosimetry of (18)f-fluoro-a-85380 in healthy volunteers, J Nucl Med, 2003;44:596–601.
  68. Kadir A, Almkvist O, Wall A, et al., PET imaging of cortical 11Cnicotine binding correlates with the cognitive function of attention in Alzheimer’s disease, Psychopharmacology (Berl), 2006;188:509–20.
  69. Sabri O, Kendziorra K, Wolf H, et al., Acetylcholine receptors in dementia and mild cognitive impairment, Eur J Nucl Med Mol Imaging, 2008;35(Suppl. 1):S30–45.
  70. Mesulam M, Giacobini E, Neuroanatomy of cholinesterases in the normal human brain and in Alzheimer’s disease. In: Giacobini E (ed.), Cholinesterases and cholinesterase inhibitors, London, UK: Martin Dunitz, 2000:121–37.
  71. Namba H, Irie T, Fukushi K, Iyo M, In vivo measurement of acetylcholinesterase activity in the brain with a radioactive acetylcholine analog, Brain Research, 1994;667:278–82.
  72. Kilbourn MR, Snyder SE, Sherman PS, Kuhl DE, In vivo studies of acetylcholinesterase activity using a labeled substrate, n-[C- 11]methylpiperdin-4-yl propionate ([C-11]PMP), Synapse, 1996;22:123–31.
  73. Kuhl DE, Koeppe RA, Minoshima S, et al., In vivo mapping of cerebral acetylcholinesterase activity in aging and Alzheimer’s disease, Neurology, 1999;52:691–9.
  74. Iyo M, Namba H, Fukushi K, et al., Measurement of acetylcholinesterase by positron emission tomography in the brains of healthy controls and patients with Alzheimers disease, Lancet, 1997;349:1805–9.
  75. Herholz K, Bauer B, Wienhard K, et al., In-vivo measurements of regional acetylcholine esterase activity in degenerative dementia: comparison with blood flow and glucose metabolism, J Neural Transm, 2000;12:1457–68.
  76. Rinne JO, Kaasinen V, Jarvenpaa T, et al., Brain acetylcholinesterase activity in mild cognitive impairment and early Alzheimer’s disease, J Neurol Neurosurg Psychiatry, 2003;74:113–15.
  77. Haense C, Hohmann C, Kalbe E, et al., Cortical Acetylcholine Esterase Activity in Healthy Controls, Mild Cognitive Impairment and Alzheimer’s Disease, Ann Neurol, 2008;64(Suppl. 12):S42.
  78. Bohnen NI, Kaufer DI, Hendrickson R, et al., Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease, J Neurol Neurosurg Psychiatry, 2005;76:315–19.
  79. Kaasinen V, Nagren K, Jarvenpaa T, et al., Regional effects of donepezil and rivastigmine on cortical acetylcholinesterase activity in Alzheimer’s disease, J Clin Psychopharmacol, 2002;22: 615–20.
  80. Kadir A, Darreh-Shori T, Almkvist O, et al., PET imaging of the in vivo brain acetylcholinesterase activity and nicotine binding in galantamine-treated patients with AD, Neurobiol Aging, 2008;29:1204–17.
  81. Snowden J, Neary D, Mann D, Frontotemporal lobar degeneration: clinical and pathological relationships, Acta Neuropathol, 2007;114:31–8.
  82. Salmon E, Garraux G, Delbeuck X, et al., Predominant ventromedial frontopolar metabolic impairment in frontotemporal dementia, NeuroImage, 2003;20:435–40.
  83. Diehl J, Grimmer T, Drzezga A, et al., Cerebral metabolic patterns at early stages of frontotemporal dementia and semantic dementia. A PET study, Neurobiol Aging, 2004;25:1051–6.
  84. Chawluk JB, Mesulam MM, Hurtig H, et al., Slowly progressive aphasia without generalized dementia: studies with positron emission tomography, Ann Neurol, 1986;19:68–74.
  85. Ishii K, Sakamoto S, Sasaki M, et al., Cerebral glucose metabolism in patients with frontotemporal dementia, J Nucl Med, 1998;39:1875–8.
  86. Foster NL, Heidebrink JL, Clark CM, et al., FDG-PET improves accuracy in distinguishing frontotemporal dementia and Alzheimer’s disease, Brain, 2007;130:2616–35.
  87. Hodges JR, Patterson K, Semantic dementia: a unique clinicopathological syndrome, Lancet Neurol, 2007;6:1004–14.
  88. Zahn R, Juengling F, Bubrowski P, et al., Hemispheric asymmetries of hypometabolism associated with semantic memory impairment in Alzheimer’s disease: a study using positron emission tomography with fluorodeoxyglucose-F18, Psychiatry Res, 2004;132:159–72.
  89. Desgranges B, Matuszewski V, Piolino P, et al., Anatomical and functional alterations in semantic dementia: a voxel-based MRI and PET study, Neurobiol Aging, 2007;28:1904–13.
  90. Perneczky R, Diehl-Schmid J, Pohl C, et al., Non-fluent progressive aphasia: cerebral metabolic patterns and brain reserve, Brain Res, 2007;1133:178–85.
  91. Minoshima S, Foster NL, Sima AA, et al., Alzheimer’s disease versus dementia with Lewy bodies: cerebral metabolic distinction with autopsy confirmation, Ann Neurol, 2001;50:358–65.
  92. Kono AK, Ishii K, Sofue K, et al., Fully automatic differential diagnosis system for dementia with Lewy bodies and Alzheimer’s disease using FDG-PET and 3D-SSP, Eur J Nucl Med Mol Imaging, 2007;34:1490–97.
  93. Hu XS, Okamura N, Arai H, et al., 18F-fluorodopa PET study of striatal dopamine uptake in the diagnosis of dementia with lewy bodies, Neurology, 2000;55:1575–7.
  94. Brooks DJ, Advances in imaging Parkinson’s disease, Curr Opin Neurol, 1997;10:327–31.
  95. Halldin C, Erixon-Lindroth N, Pauli S, et al., [(11)C]PE2I: a highly selective radioligand for PET examination of the dopamine transporter in monkey and human brain, Eur J Nucl Med Mol Imaging, 2003;30:1220–30.
  96. Tatsch K, Imaging of the dopaminergic system in differential diagnosis of dementia, Eur J Nucl Med Mol Imaging, 2008; 35(Suppl. 1):S51–7.
  97. Hilker R, Thomas A, Klein JC, et al., Dementia in Parkinson’s disease: functional imaging of cholinergic and dopaminergic pathways, Neurology, 2005;65:1716–22.

Further Resources

Share this Article
Related Content In Alzheimer's Disease & Dementia
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72