Clinical Biomarker For Alzheimer
Clinical Biomarker For Alzheimer
Clinical Biomarker For Alzheimer
Biomarkers
for Preclinical
Alzheimer’s
Disease
Neuromethods
Series Editor
Wolfgang Walz
University of Saskatchewan
Saskatoon, SK, Canada
Edited by
Robert Perneczky
Department of Psychiatry and Psychotherapy,
Ludwig-Maximilians-Universität München,
Munich, Germany
Editor
Robert Perneczky
Department of Psychiatry and Psychotherapy
Ludwig-Maximilians-Universität München
Munich, Germany
Experimental life sciences have two basic foundations: concepts and tools. The Neuromethods
series focuses on the tools and techniques unique to the investigation of the nervous system
and excitable cells. It will not, however, shortchange the concept side of things as care has
been taken to integrate these tools within the context of the concepts and questions under
investigation. In this way, the series is unique in that it not only collects protocols but also
includes theoretical background information and critiques which led to the methods and
their development. Thus it gives the reader a better understanding of the origin of the
techniques and their potential future development. The Neuromethods publishing program
strikes a balance between recent and exciting developments like those concerning new ani-
mal models of disease, imaging, in vivo methods, and more established techniques, includ-
ing, for example, immunocytochemistry and electrophysiological technologies. New
trainees in neurosciences still need a sound footing in these older methods in order to apply
a critical approach to their results.
Under the guidance of its founders, Alan Boulton and Glen Baker, the Neuromethods
series has been a success since its first volume published through Humana Press in 1985.
The series continues to flourish through many changes over the years. It is now published
under the umbrella of Springer Protocols. While methods involving brain research have
changed a lot since the series started, the publishing environment and technology have
changed even more radically. Neuromethods has the distinct layout and style of the Springer
Protocols program, designed specifically for readability and ease of reference in a laboratory
setting.
The careful application of methods is potentially the most important step in the process
of scientific inquiry. In the past, new methodologies led the way in developing new disci-
plines in the biological and medical sciences. For example, Physiology emerged out of
Anatomy in the nineteenth century by harnessing new methods based on the newly discov-
ered phenomenon of electricity. Nowadays, the relationships between disciplines and meth-
ods are more complex. Methods are now widely shared between disciplines and research
areas. New developments in electronic publishing make it possible for scientists that
encounter new methods to quickly find sources of information electronically. The design of
individual volumes and chapters in this series takes this new access technology into account.
Springer Protocols makes it possible to download single protocols separately. In addition,
Springer makes its print-on-demand technology available globally. A print copy can there-
fore be acquired quickly and for a competitive price anywhere in the world.
v
Preface
The constantly increasing number of individuals with dementia due to Alzheimer’s disease
(AD) poses a significant financial and emotional burden on the affected families and the
global society. Therapeutic strategies designed to treat symptoms or alter the disease course
have so far failed to make a positive impact, despite billions of US dollars of R&D invest-
ments and massive efforts of the industry and academic communities. These failures in
treatment have led many to believe that symptomatic AD, including its earliest clinical
stages, is resistant to drug interventions.
The recently increased focus on biomarkers to diagnose early disease is fuelled by the
hope of identifying a therapeutic window, in which the brain is still largely intact and there-
fore amenable to treatment effects. The ability of biomarkers to adequately define the pre-
clinical (i.e., at risk of AD dementia) disease stage may ultimately allow novel or repurposed
drugs to finally achieve clinically meaningful results for the affected individuals and to help
to prevent dementia and associated disability.
This book discusses the usefulness of established biomarkers (imaging, fluid, and genetic)
to detect preclinical AD, providing detailed protocols for state-of-the-art biomarker mea-
surement and analysis. It is also explained how cutting-edge technology is used to develop
novel improved biomarkers for the earliest, presymptomatic stages of AD. Our book aims to
provide a comprehensive overview specifically of biomarkers for the earliest detectable dis-
ease stages. All previous publications cover biomarkers for clinical, symptomatic AD.
This publication comprises five parts: In Part I (Chaps. 1 and 2) we explain why AD is
one of the major challenges for the global societies and healthcare systems, which highlights
the urgent need for improved approaches for early diagnosis enabling disease prevention. In
Part II (Chaps. 3–5) clinical and research concepts are presented, which are important to
improve early recognition of AD. Part III (Chaps. 6–10) provides a comprehensive over-
view of methods currently used in the AD diagnostic work-up, and it is explained how these
methods can be applied to preclinical disease. In Part IV (Chaps. 11–15) cutting-edge tech-
nology innovations and their value for early AD diagnosis are discussed. Finally, in Part V
(Chaps. 16–18), we consider important ethical considerations in relation to biomarker-based
early diagnosis, and we also discuss the meaningfulness of biomarker endpoints in clinical
AD research.
The book is targeted at individuals with an interest in the use of advanced biomarker
strategies to significantly improve the early diagnosis of AD, and thereby to accelerate the
development of effective, disease-modifying drugs. This includes researchers, clinicians,
and those interested in regulatory and medical affairs, both from academia and industry. We
wish to present biomarker development approaches as a strategy for the study of AD with
the hope and expectation that the results will translate into more effective diagnosis and
treatment and improved public health policies. We expect this book to complement other
excellent volumes and monographs on AD that cover basic science or clinical aspects of the
disease.
vii
Contents
Series Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
ix
x Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 271
Contributors
Abstract
Dementia is the principal cause of functional dependence and institutionalization in the elderly and has already
posed tremendous economic burden on the aging society. Thus, dementia has been recognized by the World
Health Organization as a global public health priority. Here, we briefly introduce the conceptual and method-
ological issues of the cost-of-illness studies (COI) of dementia, summarize the major literature regarding the
economic costs of dementia, and identify some key issues that need be addressed in future COI studies of
dementia. We identified 17 COI studies that estimated the costs of dementia in different countries, in which the
annual total costs per patient with dementia varied from US$2935 to US$64168. Differences in methodology,
data sources, cost categories assessed, and severity of patients employed in the COI studies are likely to contrib-
ute to the substantial variations in cost estimates of dementia. Thus, to increase the comparability of findings
from COI studies, the methodological issues need to be harmonized and standardized, and consensus on the
methodological principles in the COI studies would be essential. Further well-designed COI studies of demen-
tia could provide critical evidence to help develop health policy and improve allocation of healthcare resources.
Key words Dementia, Aging, Care Costs, Economic Burden, Societal Burden, Cost-of-Illness Studies
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_1, © Springer Science+Business Media, LLC 2018
3
4 Junfang Xu and Chengxuan Qiu
3.1 The Cost-of- The COI approaches are usually used to estimate dementia costs
Illness Approaches where epidemiological (e.g., number of patients with dementia)
and economic or cost data are integrated. The prevalence-based
and incidence-based approaches are the two main types of COI
studies [11]. The prevalence-based approach is based on the preva-
lence of a disease at a given year to estimate economic burden of
the disease. The total costs are estimated by multiplying the total
number of patients with dementia in a given year with the mean
cost per patient in the same year [11]. In the incidence-based
approach, the total costs are calculated by multiplying the total
number of patients with newly diagnosed (incident) dementia in a
given year with the lifetime costs of these patients [12]. The
prevalence-based approach has been frequently used to estimate
dementia costs for two reasons. First, most cost data of dementia
available in the real world cover costs of a short period of time
rather than the lifetime. Second, dementia, and Alzheimer’s dis-
ease in particular, is an irreversible chronic health condition with
an insidious onset, and prevalence could measure burden of the
disease in the society [13].
3.2 Estimation The economic burden of dementia can be analyzed from three
of Dementia Costs perspectives: family, healthcare provider, and the society [5]. The
from Different economic burden from a societal perspective refers to the costs of
Perspectives all resources consumed or lost due to dementia, irrespective of
who the payer is [14]. In this regard, costs from the societal per-
spective are most relevant for decision makers whose main interest
is the welfare of the society as a whole. A healthcare provider may
be concerned with only the costs of a hospital or nursing home in
order to optimize healthcare delivery of the hospital or nursing
home within a given budget [14]. The costs from a family perspec-
tive include those paid by the family due to the treatment or care
for dementia [14]. The cost components of different perspectives
can be varied, but it is important to include each cost item only
once regardless of the perspectives to avoid double counting.
3.3 Cost There are four cost components in COI studies of dementia [14]:
Components in Cost- (a) direct medical costs, (b) direct nonmedical costs, (c) indirect
of-Illness Studies costs, and (d) intangible costs (Fig. 1). Direct medical costs include
of Dementia all healthcare expenditures used for dementia, including goods and
service costs that are related to diagnosis and treatments of dementia
such as hospitalization and outpatient services. Direct nonmedical
6 Junfang Xu and Chengxuan Qiu
3.4 Estimation The bottom-up approach and top-down approach are the two
of Costs of the Main methods that are commonly used to estimate the direct costs of
Cost Components dementia [11]. In the bottom-up approach, cost data by individual
cost items are collected through questionnaire survey or review of
3.4.1 Direct Costs
medical records. Then, estimates of the total costs can be obtained
by multiplying the mean cost per patient with the number of
patients derived from prevalence studies. The top-down method
uses national or regional statistics on the total costs of diseases to
separate out the costs of a certain disease (e.g., dementia) [11].
Worldwide Costs of Dementia 7
3.4.2 Informal Care Informal care provided by family members, relatives, or friends has
Costs a significant impact on the societal costs of dementia [18]. Informal
caregiving, in the form of assistance with basic activities of daily
living (ADLs), is an important component of the support required
by patients with dementia, yet it is unclear how to attribute the
monetary cost to informal caregiver’s time. Indeed, it is compli-
cated to quantify the informal care costs owing to lack of consensus
in methodological issues. First, quantifying caregiver’s time can be
problematic. The time spending on informal care often depends on
the needs of care in self-care ADLs and instrumental ADLs. The
cost is much lower when only considering the cares in basic ADLs
compared with cares in both basic and instrumental ADLs, as well
as time spending in supervision [19]. Moreover, dementia patient
and the caregiver may both involve the same activities such as
shopping together [20]. Second, estimating informal care cost is
also complicated. Most of the previous studies have applied two
different methods to assessing informal care costs [20–22]: the
opportunity cost approach and the replacement cost approach. To
estimate the opportunity cost, the best alternative use of the care-
giver’s time is taken to indicate the value of that time spent in care.
If a caregiver gives up paid work to care, the lost earnings are esti-
mated. Quantifying the costs of caregiver’s time for retired people
can be challenging because there are no obvious market prices [16].
The replacement cost approach assumes that the informal caregiver’s
inputs should be calculated according to the cost of replacing their
care inputs with those of a professional caregiver [16].
3.4.3 Indirect Costs The appropriate method for estimating indirect costs remains
debatable [5]. The common approaches are the human capital, the
willingness-to-pay, and the friction cost methods [23]. In the
human capital approach, the average earnings by age and gender
are combined with time lost from work or years of working life lost
due to premature death to estimate unrealized earnings [24].
However, human capital approach with average wage may be sub-
ject to capital market imperfections. The willingness-to-pay
approach, in contrast, incorporates both lost productivity and the
8 Junfang Xu and Chengxuan Qiu
t
where Ca denotes the cost in present year a, Ct represents the
cost in year t, r refers to the discount rate, and n implies the time
interval between year a and year t [5].
In addition, there are always uncertainties when estimating
costs of a disease because COI studies are often based on a set of
assumptions and data sources [28]. Sensitivity analysis can be car-
ried out to assess the impact of variations of certain factors on the
cost estimates. In the sensitivity analysis of dementia costs, we
could assess the impact on cost estimates of variations in factors
such as prevalence, medical costs, average wages, productivity
weight, time spending on informal care, and the proportion of
patients with dementia who receive informal and formal care [24,
29–31].
3.5 Sources The sources of cost data in COI studies include the following.
of Cost Data First, we can collect cost data through questionnaire survey that
includes costs by individual items, where direct and indirect costs
can be estimated. When designing the questionnaire, cost indica-
tors can be relatively comprehensive. However, data collected
through the survey may be subject to recall bias. Accurate and
precise data may be obtained through follow-up survey, but it can
be expensive and time consuming [23]. Moreover, the questions
and guides in the questionnaire may vary across studies, thus
resulting in inaccuracies of the data [23]. Second, cost data can be
derived from electronic medical records, which usually include
accurate and reliable information on medical expenses. However,
medical records often include data of direct medical costs, and the
costs may vary depending on available facilities of hospitals or medical
institutes. Thus, cost data from certain medical institutes may not
be representative of the target population [23]. Finally, relevant
data needed for cost estimates can also be obtained from literature
or government statistics. This is usually convenient and timesaving,
but it may not capture all the utilizations, and the representativeness
should also be kept in mind [32].
Worldwide Costs of Dementia 9
5 Future Perspectives
Table 1
A brief summary of literature regarding economic costs of dementia, 2000–2017
Acknowledgment
References
1. American Psychiatric Association (2000) 4. Quentin W, Riedel-Heller S, Luppa M,
Diagnostic and statistical manual of mental Rudolph A, König HH (2010) Cost of illness
disorders, 4th edn. American Psychiatric studies of dementia: a systematic review focus-
Association, Washington DC ing on stage dependency of costs. Acta Psyciatr
2. Alzheimer’s Disease International (ADI) Scand 121:243–259
(2015) World Alzheimer report 2015, the 5. Costa N, Derumeaux H, Rapp T et al (2012)
global impact of dementia: an analysis of preva- Methodological considerations in cost of illness
lence, incidence, cost and trends. Alzheimer’s studies on Alzheimer disease. Health Econ Rev
Disease International (ADI), London. http:// 15:18
www.worldalzreport2015.org/downloads/ 6. McKhann G, Drachman D, Folstein M,
world-alzheimer-report-2015.pdf. Accessed 10 Katzman R, Price D, Stadlan EM (1984)
Jun 2017 Clinical diagnosis of Alzheimer’s disease: report
3. World Health Organization and Alzheimer’s of the NINCDS-ADRDA work group under
Disease International (2012) Dementia: a pub- the auspices of Department of Health and
lic health priority. World Health Organization, Human Services Task Force on Alzheimer's
Geneva. http://apps.who.int/iris/bitstr disease. Neurology 34:939–944
eam/10665/75263/1/9789241564458_ 7. Hope T, Keene J, Gedling K, Fairburn CG,
eng.pdf?ua=1. Accessed 10 June 2017 Jacoby R (1998) Predictors of institutionaliza-
12 Junfang Xu and Chengxuan Qiu
tion for people with dementia living at home 21. McDaid D (2001) Estimating the costs of
with a carer. Int J Geriatr Psychiatry 13: informal care for people with Alzheimer’s dis-
682–690 ease: methodological and practical challenges.
8. Small GW, McDonnell DD, Brooks RL, Int J Geriatr Psychiatry 16:400–405
Papadopoulos G (2002) The impact of symp- 22. Nordberg G, Wimo A, Jönsson L et al (2007)
tom severity on the cost of Alzheimer disease. Time use and costs of institutionalised elderly
J Am Geriatric Soc 50:321–327 persons with or without dementia: results from
9. Mauskopf J, Racketa J, Sherrill E (2010) the Nordanstig cohort in the Kungsholmen
Alzheimer’s disease: the strength of association project: a population based study in Sweden.
of costs with different measures of disease Int J Geriatr Psychiatry 22:639–648
severity. J Nutr Health Aging 14:655–663 23. Xu JF (2016) Study on the economic burden
10. Herman N, Tam DY, Balshaw R, Sambrook R, of mental disorders and the influence of health
Lesnikova N, Lanctôt KL (2010) The relation insurance-based on patients in Shandong prov-
between disease severity and cost of caring for ince. Shandong University, Jinan
patients with Alzheimer disease in Canada. Can 24. Xu J, Wang J, Wimo A, Fratiglioni L, Qiu C
J Psychiatr 55:768–775 (2017) The economic burden of dementia in
11. Wimo A, Jönsson L, Fratiglioni L et al (2016) China, 1990–2030: implications for health
The societal costs of dementia in Sweden policy. Bull World Health Organ 95:18–26
2012 – relevance and methodological chal- 25. Oostdam N, Bosmans J, Wouters MG, Eekhoff
lenges in valuing informal care. Alzheimers Res EM, Mechelen WV, Poppel MNV (2012)
Ther 8:59 Cost-effectiveness of an exercise program dur-
12. Girmens JF, Koerber C, Miadi-Fargier H, ing pregnancy to prevent gestational diabetes:
Wittrup-Jensen KU (2013) Costs of incidence- results of an economic evaluation alongside a
based central retinal vein occlusion in France. randomised controlled trial. BMC Pregnancy
Value Health 16:A504–A504 Childbirth 12:64
13. Andlin-Sobocki P, Jönsson B, Wittchen HU, 26. Gold MR, Russel LB, Siegel JE, Daniels N,
Olesen J (2005) Cost of disorders of the brain Weinstein MC (1996) Cost effectiveness in
in Europe. Eur J Neurol 12(Suppl 1):1–27 health and medicine. JAMA 276:1172–1177
14. Leung GM, Yeung RY, Chi I, Chu LW (2003) 27. Luengo-Fernandez R, Leal J, Gray A, Sullivan
The economics of Alzheimer disease. Dement R (2013) Economic burden of cancer across
Geriatr Cogn Disord 15:34–43 the European Union: a population-based cost
15. Olesen J, Gustavsson A, Svensson M, Wittchen analysis. Lancet Oncol 14:1165–1174
HU, Jönsson B (2012) The economic cost of 28. Xu J, Wang J, Wimo A, Qiu C (2016) The eco-
brain disorders in Europe. Eur J Neurol nomic burden of mental disorders in China,
19:155–162 2005–2013: implications for health policy.
16. Wimo A, Jönsson L, Bond J, Prince M, Winblad BMC Psychiatry 16:137
B, Alzheimer Disease International (2013) The 29. Wimo A, Guerchet M, Ali GC et al (2017) The
worldwide economic impact of dementia 2010. worldwide costs of dementia 2015 and compari-
Alzheimers Dement 9:1–11.e3 sons with 2010. Alzheimers Dement 13:1–7
17. Deb A, Thornton JD, Sambamoorthi U, Innes 30. Wimo A, Winblad B, Jönsson L (2010) The
K (2017) Direct and indirect cost of managing worldwide societal costs of dementia: estimates
Alzheimer’s disease and related dementias in for 2009. Alzheimers Dement 6:98–103
the United States. Expert Rev Pharmacoecon 31. Wang H, Gao T, Wimo A, Yu X (2010)
Outcomes Res 17:189–202 Caregiver time and cost of home care for
18. Schneider J, Hallam A, Islam MK et al (2003) Alzheimer’s disease: a clinic-based observa-
Formal and informal care for people with tional study in Beijing, China. Ageing Int
dementia: variations in costs over time. Ageing 35:153–165
Soc 23:303–326 32. Zhang YY (2012) Study on the economic bur-
19. Prince M, Bryce R, Albanese E et al (2013) den of inpatients with hepatitis B owning basic
The global prevalence of dementia: a systematic medical insurance for urban workers in Tianjin.
review and metaanalysis. Alzheimers Dement Tianjin University, Tianjin
9:63–75.e2 33. Rigaud AS, Fagnani F, Bayle C, Latour F,
20. Jönsson L, Wimo A (2009) The cost of dementia Traykov L, Forette F (2003) Patients with
in Europe: a review of the evidence, and method- Alzheimer’s disease living at home in France:
ological considerations. PharmacoEconomics costs and consequences of the disease. J Geriatr
27:391–403 Psychiatry Neurol 16:140–145
Worldwide Costs of Dementia 13
34. Scuvee-Moreau J, Kurz X, Dresse A (2002) 44. Zencir M, Kuzu N, Beşer NG, Ergin A, Catak
The economic impact of dementia in Belgium: B, Sahiner T (2005) Cost of Alzheimer’s dis-
results of the National Dementia Economic ease in a developing country setting. Int
Study (NADES). Acta Neurol Belg J Geriatr Psychiatry 20:616–622
102:104–113 45. Wang G, Cheng Q, Zhang S et al (2008)
35. Beeri MS, Werner P, Adar Z, Davidson M, Noy Economic impact of dementia in developing
S (2002) Economic cost of Alzheimer disease countries: an evaluation of Alzheimer-type
in Israel. Alzheimers Dis Assoc Disord dementia in shanghai, China. J Alzheimers Dis
16:73–80 15:109–115
36. Jönsson L, Eriksdotter Jö nhagen M, Kilander 46. Hurd MD, Martorell P, Langa KM (2013)
L et al (2006) Determinants of costs of care for Monetary costs of dementia in the United
patients with Alzheimer’s disease. Int J Geriatr States. N Engl J Med 369:489–490
Psychistry 21:449–459 47. Suh GH, Knapp M, Kang CJ (2006) The eco-
37. Francois C, Sintonen H, Sulkava R, Rive DB nomic costs of dementia in Korea, 2002. Int
(2004) Cost effectiveness of memantine in J Geriatr Psychiatry 21:722–728
moderately severe to severe Alzheimer’s dis- 48. Allegri RF, Butman J, Arizaga RL et al (2007)
ease: a Markov model in Finland. Clin Drug Economic impact of dementia in developing
Invest 24:373–384 countries: an evaluation of costs of Alzheimer-
38. Wolstenholme J, Fenn P, Gray A, Keene J, type dementia in Argentina. Int Psychogeriatr
Jacoby R, Hope T (2002) Estimating the rela- 19:705–718
tionship between disease progression and cost 49. Cavallo MC, Fattore G (1997) The economic
of care in dementia. Br J Psychiatry and social burden of Alzheimer disease on fam-
181:36–42 ilies in the Lombardy region of Italy. Alzheimer
39. Livingston G, Katona C, Roch B, Guilhaume Dis Assoc Disord 11:184–190
C, Rive B (2004) A dependency model for 50. Hurd MD, Martorell P, Langa K (2015) Future
patients with Alzheimer’s disease: its validation monetary costs of dementia in the United
and relationship to the costs of care. The States under alternative dementia prevalence
LASER-AD study. Curr Med Res Opin scenarios. J Popul Ageing 8:101–112
20:1007–1016 51. Matthews FE, Arthur A, Barnes LE et al (2013)
40. Lopez-Bastida J, Serrano-Aguilar P, Perestelo- A two-decade comparison of prevalence of
Perez L, Olivamoreno J (2006) Social- dementia in individuals aged 65 years and older
economic costs and quality of life of Alzheimer from three geographical areas of England:
disease in the Canary Islands, Spain. Neurology results of the cognitive function and ageing
67:2186–2191 study I and II. Lancet 382:1405–1412
41. Coduras A, Rabasa I, Frank A et al (2010) 52. Matthews FE, Stephan BC, Robinson L et al
Prospective one-year cost-of-illness study in a (2016) A two decade dementia incidence com-
cohort of patients with dementia of Alzheimer’s parison from the cognitive function and ageing
disease type in Spain: the ECO study. studies I and II. Nat Commun 7:11398
J Alzheimers Dis 19:601–615 53. Satizabal CL, Beiser AS, Chouraki V, Chêne G,
42. Mesterton J, Wimo A, By A, Langworth S, Winblad Dufouil C, Seshadri S (2016) Incidence of
B, Jönsson L (2010) Cross sectional observational dementia over three decades in the Framingham
study on the societal costs of Alzheimer’s disease. heart study. N Engl J Med 374:523–532
Curr Alzheimer Res 7:358–367 54. Langa KM, Larson EB, Crimmins EM et al
43. Kiencke P, Dietmar D, Grimm C, Rychlik R (2017) A comparison of the prevalence of
(2011) Direct costs of Alzheimer disease in dementia in the United States in 2000 and
Germany. Eur J Health Econ 12:533–539 2012. JAMA Intern Med 177:51–58
Chapter 2
Abstract
Alzheimer’s disease (AD) appears to result from a highly dynamical, time-dependent cascade of neuro-
pathological processes, which involve the balance between two antagonist pathways: an accumulation of
lesions in the brain (evolving progressively over decades), balanced by a variety of cellular and tissular
compensatory mechanisms. Preclinical research has indicated that several nutritional factors have the
potential to modulate many components of this complex system; dietary interventions may thus constitute
promising strategies for the prevention of AD. Extensive epidemiological research has been conducted on
nutrition, diet, and AD dementia over the past two decades. This chapter reviews existing epidemiological
literature on nutrition and cognitive aging and AD dementia, with a specific focus on observational, pro-
spective studies, including both clinical outcomes and brain imaging biomarkers. Most studies investigated
candidate foods and nutrients, including “good” fats (e.g., marine long-chain omega-3 polyunsaturated
fatty acids provided by fish intake), antioxidant nutrients (i.e., vitamins C and E, carotenoids, and polyphe-
nols, found in plant products), and B vitamins (e.g., folate provided by green leafy vegetables). Overall,
associations between individual nutrients and both clinical outcomes and brain imaging biomarkers have
been relatively inconsistent. Healthy dietary patterns such as the Mediterranean diet, which combine sev-
eral healthy foods and nutrients, have been more consistently related to AD outcomes. Still, most random-
ized controlled trials on nutritional supplements for the prevention of AD have remained inconclusive so
far. Future research should leverage innovative approaches such as “omics” methods to explore dietary
exposures, novel imaging markers to capture prodromal AD, and longitudinal statistical approaches to
model long-term trajectories of risk factors and AD markers, to better understand how and when diet may
shape the brain and prevent AD.
Key words Alzheimer’s disease, Dementia, Epidemiology, Population-based, Diet, Nutrition, Vitamins,
Fatty acids, Clinical trial
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_2, © Springer Science+Business Media, LLC 2018
15
16 Cécilia Samieri
2.1 Nutrient Several nutrients and foods have neuroprotective effects which have
and Food Candidates been largely established in preclinical research. Overall, these nutri-
for Brain Health ent/food candidates fall into three main categories: (1) “good”
fats, i.e., unsaturated fats, in particular polyunsaturated fats from
the omega-3 family (n-3 PUFAs), mainly provided by vegetable oils
(for precursors, e.g., alpha linolenic acid [ALA]) and by fish (for the
long-chain derivatives, e.g., eicosapentaenoic acid [EPA] and doco-
sahexaenoic acid [DHA] which are directly useful for brain struc-
ture and function); (2) antioxidant nutrients, including vitamins E
and C, carotenoids, and polyphenols, provided by fruits, vegetables,
vegetable oils (e.g., olive oil), and plant-derived beverages (coffee,
tea, wine); (3) other vitamins involved in various neuroprotective
mechanisms, including B vitamins (folate or vitamin B9, particu-
larly abundant in green leafy vegetables, and vitamin B12 provided
by animal products) and vitamin D, found in fatty fish. Each of
these nutrients has been individually related to the risk of AD
clinical outcomes in prospective observational studies.
2.1.1 “Good” Fats: Lipids represent 50–60% of total brain weight, mostly under the
Omega-3 Polyunsaturated form of PUFAs in phospholipids of neuronal membranes or in the
Fats and Fish gain of myelin, and the long-chain (LC) n-3 PUFA DHA repre-
sents up to 60% of esterified fatty acids in neuronal membranes.
DHA is essential to the development of the mammalian brain—in
Diet and Alzheimer’s Disease 19
2.1.3 Vitamins B and D B vitamins, including folate (vitamin B9), vitamin B6, and vitamin
B12, have a fundamental role in brain development and function-
ing and have been extensively examined in relation to age-related
brain diseases [63]. B vitamins regulate homocysteine (Hcy) levels,
and hyperhomocysteinemia is an established risk factor for cardio-
vascular diseases and dementia [64]. Hyperhomocysteinemia may
affect brain function through both vascular mediation and neuro-
toxicity [65]. B vitamins may also have a direct effect on the brain,
independently of Hcy: a lower status in B vitamins decreases
S-adenosylmethionine, a major intermediate of methylation reac-
tions which are involved in synaptic transmission and epigenetic
regulation [66]. Moreover, tetrahydrofolate, the active form of
folate, is involved in DNA replication which is critical for adult hip-
pocampal neurogenesis [67]; direct mechanistic effects of B vita-
mins on the brain independent of Hcy may therefore be particularly
relevant to folate. Prospective epidemiological studies have found
associations between higher intakes of B vitamins and a lower risk
of dementia and AD; four studies have reported associations with
dietary folate (but not with vitamins B6 and B12) [68–71],
although null [72–74] and even harmful associations (when folate
was combined with low vitamin B12 status) [74] were also
reported. Findings were also inconsistent in studies using blood
status in B vitamins (see for review [75]). Importantly, most stud-
ies were conducted in the USA, where fortification of all flour and
uncooked cereal grain product in folic acid became mandatory in
1998, leading to a subsequent dramatic increase in folate status
which may have biased observational findings. Moreover, it is pos-
sible that folate is protective for the brain in lower intake ranges
and become inefficient—and even detrimental for those with low
B12 status—at higher ranges (as those found in North American
population under folic acid fortification policy). In the French 3C
study in France (a country where folic acid fortification is not
Diet and Alzheimer’s Disease 23
2.2 Nutrient Dietary patterns incorporate all dietary habits in a global appraisal
and Dietary Patterns of diet and may be more relevant to brain health and diseases than
nutrients taken in isolation. Indeed, many nutrients exert biologi-
cal effects on the brain (as developed above); yet the combination
of these nutrients in the diet may have a stronger effect than any
nutrient considered individually. Alternatively, individual nutrients
may be not sufficiently powerful to protect the brain from age-
related diseases, and global approaches of diet may be more prom-
ising. Moreover, nutrients may act in synergy (although the concept
of synergy of the food matrix has remained largely hypothetical to
date). For example, antioxidant nutrients (e.g., carotenoids, vita-
min D) may contribute to decrease the peroxidation of PUFAs in
neuronal membranes of the aging brain [83, 84]. Accordingly,
regular intake of fruits and vegetables (the main sources of antioxi-
dant nutrients) appeared necessary to observe a cognitive benefit
of fish consumption (a source of n-3 PUFAs and vitamin D) in the
3C study [28]. As done earlier with cardiovascular diseases, many
cohort studies have thus examined the associations between dietary
patterns and brain aging outcomes.
2.2.1 The Mediterranean Most convincing research on overall diet quality and brain health
Diet: A Landmark Model has certainly been obtained with the Mediterranean diet (MeDi).
of Healthy Diet and Brain The MeDi, now considered as a landmark model for a healthy diet,
Health has been defined as the traditional diet from Mediterranean coun-
tries; it is characterized by high intakes of plant foods (fruits, veg-
etables, cereals, and legumes), low intakes of meats and dairies,
moderate intakes of fish and wine during meals, and olive oil as a
main source of added fats [85]. Primary findings for a protective
role of the MeDi on AD [86] and cognitive aging [87] were pro-
vided by the US cohort WHICAP and the French 3C study. These
initial findings were replicated in several populations (see for review
[85]); a meta-analysis combining five cohort studies found that
subjects with higher MeDi adherence (i.e., in the last tertile of
distribution of a score of adherence to the MeDi) had a 33% lower
risk (95% CI 19–45%) of mild cognitive impairment or AD [88].
However, inconsistent results were also reported, especially with
cognitive decline in the non-Mediterranean North American pop-
ulation (e.g., the MeDi was associated with higher average cogni-
tive status, but not cognitive decline, in two large US cohorts [89,
90]). A Mediterranean diet score adapted to the Australian diet
was associated with less decline in executive function over 3 years,
but only in carriers of the APOEε4 allele [91].
2.2.2 Other Approaches Other indices of diet quality have been investigated in relation to
of Food Quality and Brain dementia outcomes. For example, among 27,860 subjects from 40
Health countries, a modified Alternative Healthy Eating Index, comprising
seven components (including vegetables, fruits, nuts and soybean
proteins, whole grain, deep-fried foods, ratio of fish to meat and
egg, and alcohol), was associated with risk of a ≥3-point decline in
Diet and Alzheimer’s Disease 25
2.2.3 Exploratory Exploratory approaches have also been utilized to identify nutri-
Nutrient/Dietary Patterns ent/dietary patterns related to brain health without formulating
and Brain Health any a priori hypothesis on diet-to-brain relationships. Cluster analy-
sis, principal component analysis, and partial least squares analyses
(and a variant, the reduced rank regression) have been employed to
characterize dietary patterns in various populations (see for review
[97, 98]). Overall, findings from these studies are difficult to com-
pare because the patterns obtained with exploratory approaches are
constitutively specific to the studied population, and limited
research has explored longitudinal associations with dementia out-
comes so far. Studies have often interpreted their patterns as
“healthy” or “prudent” versus “unhealthy” or “western” diets, yet
definitions have varied largely across populations and associations
with cognitive decline/dementia were relatively inconsistent. For
example, in the Whitehall II study, the “prudent” diet, character-
ized by higher intakes of vegetables, fruits, dried legume, and fish,
was related to lower odds of cognitive impairment, while the “west-
ern” pattern (sweetened desserts, chocolates, fried food, processed
meat, pies, refined grains, high-fat dairy products, margarine) was
related to higher odds of cognitive deficit; however, adjustment for
education attenuated these cross-sectional associations [99]. In the
Australian Imaging, Biomarkers and Lifestyle (AIBL) study, the
“prudent” pattern obtained by factor analysis (characterized by high
intakes of vegetables, fruits, nuts, whole grains) was not associated
26 Cécilia Samieri
3.1 Nutrition, Diet, Among individual dietary candidates for brain health, fish intake/
and Topographical LC n-3 PUFA exposures have certainly been among the most
Imaging Biomarkers studied in relation to brain volumes in previous research (Table 1).
(Reflecting Macro-/ We have reviewed six large epidemiological studies, mostly cross-
Microstructural and/or sectional, on the relationships between fish intake or blood status
Metabolic Changes in EPA and/or DHA and volumes of the whole brain, the hippo-
Associated campus, and/or the medial temporal lobe (which includes hippo-
with Neuronal Loss campus and other structures of the limbic system, in particular the
and/or Brain Vascular amygdala). Overall, studies have been inconsistent though. Two
Injury) cross-sectional analyses from the Cardiovascular Health Study
reported null associations [107, 108] (Table 1 lines 1–2); a longi-
3.1.1 Brain Atrophy tudinal retrospective analysis from the ADNI found relationships
limited to APOE4 carriers (Table 1 line 3); whereas three other
cohorts [109, 110] (lines 5–6) (including a single longitudinal
28 Cécilia Samieri
Table 1
Epidemiological studies on the relationship between nutrition, diet, and brain imaging biomarkers of AD dementia
(longitudinal studies are underlined in gray)
Line Type of imaging biomarker Study Study (1st and last Design (cross-sectional Nutritional Main findings (based on
author, year, country, [CS] versus longitudinal Exposure multivariate models)
cohort name) [L]; N; age at baseline
[minimum, mean or
range, whatever
available])
Topographical imaging biomarkers
(reflecting macro/microstructural and/or
metabolic changes associated with
neuronal loss and/or brain vascular injury)
Brain atrophy
1 Atrophy of the wholebrain/hippocampus/ Virtanen and CS; N=4,128 Fish intake Fish intake not associated with
enthorinal cortex/medial temporal lobe Mozaffarian; 2008; participants sulcal or ventricular grades
USA; Cardiovascular without known (visual grading), markers of
Health Study [107] cerebrovascular brain atrophy
disease; ≥65 years
2 Virtanen and CS; N=2,293 Plasma Higher plasma phospholipid
Mozaffarian; 2013; participants Phospholipid n-3 long-chain n-3 PUFA
USA; Cardiovascular without known PUFA (EPA+DPA+DHA) not associated
Health Study cerebrovascular with sulcal or ventricular grades
[107,108] disease; ≥65 years (visual grading), markers of
brain atrophy
3 Daiello and Ott for L (retrospective cohort; Fish oil use (mostly Fish oil use associated with
the Alzheimer’s exposure an outcomes of moderate to lower of both the whole brain
Disease modeled concomitantly long duration, ie, (for normal controls and MCI
Neuroimaging over 4 years); N=819; 70% had used fish participants) and the
Initative (ADNI); 55-90 years oil during > 1 year hippocampus (for normal
2015; USA; ADNI [30] before baseline) controls and AD patients), and
less cognitive decline but only
among APOE4 non-carriers
4 Samieri and L (volume change over Plasma long-chain Plasma EPA associated with
Barberger-Gateau; 4 years); N=281; 72.3 n-3 PUFA at lower atrophy in part of the
2012; France; Three- years baseline right medial-temporal lobe;
City Study [111] anatomical changes linked with
slower cognitive decline
5 Tan and Seshadri; CS; N=1,575 without EPA and DHA in red Lower DHA level (Q1 versus Q2-
2012; USA; the dementia; 67 years blood cells Q4) associated with lower total
Framingham brain volume
Offspring Study [109]
6 Pottala and Harris; CS (with brain imaging EPA and DHA in red Higher EPA+DHA in red blood
2014; USA; Women’s 8 years after blood blood cells cells (ie, omega-3 index)
Health Initiative draw); N=1,111 associated with larger brain
Memory Study women; 65 to 80 years volume and larger hippocampal
(WHIMS) [110] volume
7 Bowman and Quinn; CS; N=42 (with brain Nutrient biomarker A nutrient biomarker pattern
2012; USA; the imaging) with CDR<0.5; patterns (based on “BCDE” characterized by higher
Oregon Brain Aging 87 years a set of 30 plasma vitamins B (B1, B2, B6,
Study cohort [125] biomarkers) at folate, B12), C, D and E
baseline associated with higher total
brain volume
8 Titova and Benedict; CS (with brain imaging Mediterranean diet Higher Mediterranean diet
2013; Sweden; 5 years after diet score at baseline score not associated total brain
Prospective assessment); N=194; volume 5 years later (higher
Investigation of the 70.1 years meat intake significantly
Vasculature in associated with worse volume
Uppsala Seniors [114] and worse cognition).
9 Gu and Scarmeas; CS; N=674; 80.1 years Mediterranean diet Higher Mediterranean diet
2015; USA; score score associated with larger
Washington total brain volumes (especially
Heights/Hamilton high fish and low meat);
Heights Inwood associations were equivalent to
Columbia Aging 5 years of aging.
Project (WHICAP)
[113]
10 Pelletier and Samieri, CS (with brain imaging Mediterranean diet Higher Mediterranean diet
2015, Three-City 9 years after diet score at baseline score not associated with grey
Study [115] assessment); N=146; matter or white matter brain
73 years volumes in any region
11 Luciano and Deary; L (volume and corticial Mediterranean diet Higher Mediterranean diet
2017; Scotland; thickness change over score score not associated with total
Lothian Birth Cohort 3 years, with baseline brain volumes 3 years and 6
of 1936 [116] brain imaging 3 years years after diet assessment, but
after diet assessment); significantly associated with
N=401 (346 for cortical lower atrophy over 3 years (no
thickness) specific association with fish or
meat found)
(continued)
Diet and Alzheimer’s Disease 29
12 Cortical thickness Walhovd and Fjell; L (change over 3.6 Several exposures Higher DHA associated with
2014; Norway; years, with blood including plasma less cortical thinning (preceding
Cognition and biomarkers assessed at long-chain n-3 biomarker assessment) in the
Plasticity through the the second MRI visit); PUFA and vitamin left middle and superior
Lifespan project [112] N=92; 53.8 years D temporal cortex; higher vitamin
(range 23.5 to 87.8 D associated with less cortical
years) thinning in right lateral
prefrontal cortex
13 Mosconi and de Leon; CS; N=52; 54 years Mediterranean diet Higher Mediterranean diet
2014; USA; study at score score associated with larger
New York University cortical thickness in the
school of Medicine entorhinal, orbitofrontal and
[117] posterior cingulate regions
14 Gu and Scarmeas; CS; N=674; 80.1 years Mediterranean diet Higher Mediterranean diet
2015; USA; score score not associated with
Washington cortical thickness (association
Heights/Hamilton of fish to greater cortical
Heights Inwood thickness only).
Columbia Aging
Project (WHICAP)
[113]
15 Staubo and Roberts, CS; N=672; 79.8 years Mediterranean diet Higher Mediterranean diet
2017, USA, Mayo score score associated with larger
Clinic Study of Aging cortical thickness in many
[118] regions (especially high legume
and fish and low carbohydrate
and sugar), but not significantly
with an AD signature cortical
thickness
16 Luciano and Deary; L (change over 3 years, Mediterranean diet Higher Mediterranean diet
2017; Scotland; with baseline brain score score neither associated with
Lothian Birth Cohort imaging 3 years after cortical thickness 3 years and 6
of 1936 [116] diet assessment); years after diet assessment nor
N=401 (346 for cortical with cortical thickness change
thickness) over 3 years
Brain microstructure
17 White matter microstructure (DTI) Gu and Brickman, CS (with brain imaging Nutrient intake A pattern characterized by
2016, USA, WHICAP 5.1 years after diet patterns (based on higher intakes of PUFA (n-3 and
[119] assessment); N=239; a set of 24 n-6) and vitamin E associated
84.1 years nutrients) with higher average fractional
anisotropy; this association
with white matter
microstructure mediated the
relations of the pattern to
memory and cognition
18 Pelletier and Samieri, CS (with brain imaging Mediterranean diet Higher Mediterranean diet
2015, Three-City 9 years after diet score at baseline score associated with higher
Study [115] assessment); N=146 fractional anisotropy and lower
not demented at mean diffusivity in many white
baseline; 73 years matter regions; no significant
association with brain volumes
Brain hypometabolism
19 Mosconi and de Leon; CS; N=49 cognitively Intakes of 10 Higher intake of β carotene and
2014; USA; study at normal individuals candidate nutrients folate from food sources
New York University (70% with family associated with higher brain
school of Medicine history of late-onset AD metabolism in AD regions of
[121] and 39% carriers of the interest at 18F-
APOE4 allele); 54 years Fluorodeoxyglucose (FDG)-PET
(range 25 to 72 years)
20 Berti and Mosconi; CS; N=52 cognitively Nutrient intake Several nutrient intake patterns
2015; USA; study at normal individuals patterns (based on associated with higher brain
New York University (69% with family 35 nutrients) metabolism in AD regions:
school of Medicine history of late-onset AD - A pattern with higher intakes
[122] and 39% carriers of the of vitamin B12, vitamin D and
APOE4 allele); 54 years Zinc (also associated with
(range 25 to 72 years) lower amyloid load and higher
grey matter volume)
- A pattern with lower intakes
of saturated and trans fats,
cholesterol and sodium (also
(continued)
30 Cécilia Samieri
22 Subclinical infarcts Virtanen and CS and L (2 MRI 5 years Fish intake Consuming tuna/other fish ≥3
Mozaffarian; 2008; apart); N=4,128 for CS times/week associated with
USA; Cardiovascular and N=1,124 for L lower prevalence of subclinical
Health Study [107] analyses, without known infarcts (ischemic lesions
cerebrovascular ≥3mm) compared to
disease; ≥65 years <once/month; non-significant
tend for association with
incident subclinical infarcts
23 Virtanen and CS and L (2 MRI 5 years Plasma Higher plasma phospholipid
Mozaffarian; 2013; apart); N=2,293 for CS Phospholipid n-3 long-chain n-3 PUFA
USA; Cardiovascular and N=1,056 for L PUFA (EPA+DPA+DHA) associated
Health Study analyses, without known with lower prevalence of
[107,108] cerebrovascular subclinical infarcts; no
disease; ≥65 years association with incident
subclinical infarcts
24 White matter hyperintensities Tan and Seshadri; CS; N=1,575 without EPA and DHA in red Higher DHA level associated
2012; USA; the dementia; 67 years blood cells with lower white matter hyper-
Framingham intensities volume but not after
Offspring Study [109] adjustment for the full set of
potential confounders; no
association with silent cerebral
brain infarcts
25 Pottala and Harris; CS (with brain imaging EPA and DHA in red EPA+DHA in red blood cells (ie,
2014; USA; Women’s 8 years after blood blood cells omega-3 index) not associated
Health Initiative draw); N=1,111 with ischemic lesion volume
Memory Study women; 65 to 80 years (represented by both diffuse
(WHIMS) [110] small-vessel disease and white
matter hyperintensities)
26 Bowman and Quinn; CS; N=42 (with brain Nutrient biomarker A nutrient biomarker pattern
2012; USA; the imaging) with CDR<0.5; patterns (based on characterized by higher plasma
Oregon Brain Aging 87 years a set of 30 long-chain n-3 PUFA associated
Study cohort [125] biomarkers) at with lower white matter hyper-
baseline intensities volume, but only
among participants without
depression
report from the Three-City (3C) study [111] (line 4)) found
significant associations of higher blood LC n-3 PUFA status to
higher brain volumes (/lower brain atrophy in 3C). In comple-
ment to these studies on brain volumes, two studies reported inter-
esting associations between fish/DHA and cortical thickness. The
Cognition and Plasticity through the Lifespan project found a lon-
gitudinal association between higher blood DHA and less cortical
thinning in part of the left temporal cortex [112] (line 12).
However, the study was strongly limited by a biomarker assess-
ment performed after ascertainment of cortical thinning.
Furthermore, in a cross-sectional analysis from the large MRI sam-
ple of the WHICAP, higher fish intake was the single component
of the MeDi score associated with greater cortical thickness [113]
(line 14). Thus, the relation of fish/LC n-3 PUFAs to cortical
thickness (a subtle marker of neuronal loss) certainly deserves fur-
ther research.
Aside from studies on fish/n-3 PUFAs, a number of cohorts
investigated overall diet through adherence to the MeDi; however,
as with fish, inconsistent results were found in relation to brain
atrophy. For example, both the Swedish Prospective Investigation
of the Vasculature in Uppsala Seniors and the 3C study found no
association of the MeDi to brain volumes [114, 115] (lines 8 and
10). Likewise, in the Lothian Birth Cohort, there was no associa-
tion between the MeDi and brain volumes assessed 3 years apart;
yet adherence to the MeDi was related to lower brain atrophy
between the two time points [116] (line 10). Only the WHICAP
reported a significant association between the MeDi and total brain
volumes [113] (line 9). More recent studies on cortical thickness
have not been more consistent than those which targeted volumes.
Two US cohorts, including the large sample (N > 500) from the
Mayo Clinic Study of Aging, reported a cross-sectional relation
between the MeDi and larger cortical thickness [117, 118] (lines
13 and 15). In contrast, none of the two large WHICAP and
Lothian Birth Cohort studies found any significant association of
the MeDi to cortical thickness [113, 116] (lines 14 and 16).
3.1.2 Alterations of Brain Studies on diet and brain microstructure, which have been
Microstructure extremely limited to date, yielded promising findings. Both the
WHICAP and the 3C study found associations of (1) a nutrient
pattern with higher PUFAs and vitamin E and (2) the MeDi,
respectively, to preserved white matter structure [115, 119]
(Table 1 lines 17 and 18). Interestingly, in both studies, the
reported associations were accompanied by strong relations with
cognitive function, suggesting that alterations in brain connectivity
may serve as a mediator in the relationship between low diet qual-
ity and cognitive aging. Moreover, in a small clinical trial (N = 65),
26 weeks of supplementation with EPA + DHA (2.2 g/day) in
older persons led to an improvement of white matter microstruc-
32 Cécilia Samieri
3.2 Nutrition, Diet, The term cerebral small vessel disease refers to a group of patho-
and Pathophysiological logical processes with various etiologies that affect the small arter-
Imaging Biomarkers ies, arterioles, venules, and capillaries of the brain. The causes are
3.2.1 Markers multiple and include cerebral amyloid angiopathy, hypertension,
of Cerebral Small and vascular risk factors in general. As a consequence of small ves-
Vessel Disease sel disease, various subcortical lesions develop in brain parenchyma,
including lacunar infarcts, white matter lesions, large hemorrhages,
and microbleeds [123]. These lesions reflecting small vessel disease
can be detected at MRI; as their prevalence dramatically increases
with age, they are commonly considered part of the spectrum of
vascular-related brain injury associated with brain aging. For exam-
ple, white matter hyperintensities (WMH), defined as white matter
lesions appearing as hyperintensities on T2-weighted images at
MRI, range from 11–21% around 64 years old to 94% at age 82
[124].
Assuming that diet, which has a primary influence on vascular
risk factors, may exert a beneficial role on the brain vasculature,
several of the largest cohorts on diet and dementia have examined
the relations between nutritional factors and MRI markers of cere-
bral small vessel disease. Primary studies were published a decade
ago; still, existing literature has remained scarce (mostly limited to
fish/n-3 PUFA) and, as with brain atrophy, generally inconsistent
(Table 1). The Cardiovascular Health Study was among the first to
examine the relation of fish/LC n-3 PUFA status to subclinical
infarcts; despite significant cross-sectional associations and a very
large sample size (N > 1000), findings were null for the prospective
relationships with incident lesions [107, 108] (Table 1 lines 22 and
23). In addition, two large US cohorts (N= > 1000) failed to evi-
dence any relation of blood LC n-3 PUFA to WMH volume [109,
110] (lines 24 and 25). A relation between a nutrient biomarker
pattern reflecting higher plasma LC n-3 PUFA and lower WMH
volume was reported in a small sample from the Oregon Brain
Diet and Alzheimer’s Disease 33
3.2.2 Markers of Brain PET amyloid reflects the neuropathological hallmark of AD and is
Amyloid thus considered the “king” biomarker in AD research. However,
given its elevated cost, virtually no research has been made on diet
and PET amyloid so far. Recently, the Atherosclerosis Risk in
Communities (ARIC) study found a relationship between midlife
obesity and brain amyloid deposition 20 years later, suggesting a
strong, lifelong influence of diet-related risk factors on AD neuro-
pathology [127]. However, the dietary factors likely to influence
amyloid load have remained understudied to date. One of the sin-
gle studies has been conducted by the NY University School of
Medicine, which recently published two pilot studies on nutrients
and brain amyloid [121, 122] (Table 1, lines 27 and 28). In one of
these studies, which included 49 cognitively normal individuals
with high familial and genetic risk for AD, higher intakes of vita-
min B12, vitamin D, and n-3 PUFA were related to lower amyloid
load in regions with typical amyloid accumulation in AD. Moreover,
a small cross-sectional study from the Aging Brain Study reported
a correlation between higher blood DHA and lower amyloid load
among cognitively normal older adults (line 29). Added to the
important findings on midlife obesity and brain amyloid load, these
promising preliminary findings with nutrients call for additional
large-scale studies on diet and brain amyloid in future research.
References
1. World Alzheimer report 2016 (2016) 12. Su HM (2010) Mechanisms of n-3 fatty
https://www.alz.co.uk/research/world- acid-mediated development and maintenance
report-2016. Accessed 16 May 2017 of learning memory performance. J Nutr
2. Lambert JC, Ibrahim-Verbaas CA, Harold Biochem 21(5):364–373. https://doi.
D et al (2013) Meta-analysis of 74,046 indi- org/10.1016/j.jnutbio.2009.11.003
viduals identifies 11 new susceptibility loci for 13. Lim GP, Calon F, Morihara T et al (2005) A
Alzheimer’s disease. Nat Genet 45(12):1452– diet enriched with the omega-3 fatty acid doc-
1458. https://doi.org/10.1038/ng.2802 osahexaenoic acid reduces amyloid burden in
3. Scheltens P, Blennow K, Breteler MM et al an aged Alzheimer mouse model. J Neurosci
(2016) Alzheimer’s disease. Lancet. https:// 25(12):3032–3040
doi.org/10.1016/S0140-6736(15)01124-1 14. Barberger-Gateau P, Letenneur L, Deschamps
4. Grasset L, Brayne C, Joly P et al (2016) V et al (2002) Fish, meat, and risk of demen-
Trends in dementia incidence: evolution tia: cohort study. BMJ 325(7370):932–933
over a 10-year period in France. Alzheimers 15. Morris MC, Evans DA, Tangney CC et al
Dement 12(3):272–280. https://doi. (2005) Fish consumption and cognitive
org/10.1016/j.jalz.2015.11.001 decline with age in a large community study.
5. Satizabal CL, Beiser AS, Chouraki V et al Arch Neurol 62(12):1849–1853
(2016) Incidence of dementia over three 16. Schaefer EJ, Bongard V, Beiser AS et al (2006)
decades in the Framingham Heart Study. N Plasma phosphatidylcholine docosahexae-
Engl J Med 374(6):523–532. https://doi. noic acid content and risk of dementia and
org/10.1056/NEJMoa1504327 Alzheimer disease: the Framingham Heart
6. Small SA, Duff K (2008) Linking abeta and Study. Arch Neurol 63(11):1545–1550
tau in late-onset Alzheimer’s disease: a dual 17. Samieri C, Feart C, Letenneur L et al (2008)
pathway hypothesis. Neuron 60(4):534– Low plasma eicosapentaenoic acid and
542. https://doi.org/10.1016/j. depressive symptomatology are independent
neuron.2008.11.007 predictors of dementia risk. Am J Clin Nutr
7. Kumar DK, Choi SH, Washicosky KJ et al 88:714–721
(2016) Amyloid-beta peptide protects 18. Lopez LB, Kritz-Silverstein D, Barrett Connor
against microbial infection in mouse and E (2011) High dietary and plasma levels of
worm models of Alzheimer’s disease. Sci the omega-3 fatty acid docosahexaenoic acid
Transl Med 8(340):340ra372. https://doi. are associated with decreased dementia risk:
org/10.1126/scitranslmed.aaf1059 the Rancho Bernardo study. J Nutr Health
8. Zhao Z, Nelson AR, Betsholtz C et al Aging 15(1):25–31
(2015) Establishment and dysfunction of 19. Huang TL (2010) Omega-3 fatty acids,
the blood-brain barrier. Cell 163(5):1064– cognitive decline, and Alzheimer’s disease: a
1078. https://doi.org/10.1016/j. critical review and evaluation of the literature.
cell.2015.10.067 J Alzheimers Dis 21(3):673–690. https://
9. Stern Y (2009) Cognitive reserve. doi.org/10.3233/JAD-2010-090934
Neuropsychologia 47(10):2015–2028 20. Zhang Y, Chen J, Qiu J et al (2016) Intakes of
10. Shah H, Albanese E, Duggan C et al fish and polyunsaturated fatty acids and mild-
(2016) Research priorities to reduce the to-severe cognitive impairment risks: a dose-
global burden of dementia by 2025. Lancet response meta-analysis of 21 cohort studies.
Neurol 15(12):1285–1294. https://doi. Am J Clin Nutr 103(2):330–340. https://
org/10.1016/s1474-4422(16)30235-6 doi.org/10.3945/ajcn.115.124081
11. Cunnane SC, Plourde M, Pifferi F et al (2009) 21. Burdge GC, Calder PC (2005) Conversion of
Fish, docosahexaenoic acid and Alzheimer’s alpha-linolenic acid to longer-chain polyun-
disease. Prog Lipid Res 48(5):239–256. saturated fatty acids in human adults. Reprod
doi:S0163-7827(09)00018-6 [pii]10.1016/j. Nutr Dev 45(5):581–597. https://doi.
plipres.2009.04.001 org/10.1051/rnd:2005047
Diet and Alzheimer’s Disease 37
22. Morris MC, Brockman J, Schneider JA et al 34. Geleijnse JM, Hollman P (2008) Flavonoids
(2016) Association of seafood consumption, and cardiovascular health: which com-
brain mercury level, and APOE epsilon4 pounds, what mechanisms? Am J Clin Nutr
status with brain neuropathology in older 88(1):12–13
adults. JAMA 315(5):489–497. https://doi. 35. Corder R, Mullen W, Khan NQ et al (2006)
org/10.1001/jama.2015.19451 Oenology: red wine procyanidins and vascular
23. van de Rest O, Wang Y, Barnes LL et al health. Nature 444(7119):566. https://doi.
(2016) APOE epsilon4 and the associa- org/10.1038/444566a
tions of seafood and long-chain omega-3 36. Brickman AM, Khan UA, Provenzano FA
fatty acids with cognitive decline. Neurology et al (2014) Enhancing dentate gyrus func-
86(22):2063–2070. https://doi. tion with dietary flavanols improves cognition
org/10.1212/WNL.0000000000002719 in older adults. Nat Neurosci 17(12):1798–
24. Yassine HN, Braskie MN, Mack WJ et al 1803. https://doi.org/10.1038/nn.3850
(2017) Association of docosahexaenoic acid 37. Andres-Lacueva C, Shukitt-Hale B,
supplementation with Alzheimer disease stage Galli RL et al (2005) Anthocyanins in
in apolipoprotein E epsilon4 carriers: a review. aged blueberry-fed rats are found cen-
JAMA Neurol. https://doi.org/10.1001/ trally and may enhance memory. Nutr
jamaneurol.2016.4899 Neurosci 8(2):111–120. https://doi.
25. Laitinen MH, Ngandu T, Rovio S et al (2006) org/10.1080/10284150500078117
Fat intake at midlife and risk of dementia and 38. Marambaud P, Zhao H, Davies P (2005)
Alzheimer’s disease: a population-based study. Resveratrol promotes clearance of Alzheimer’s
Dement Geriatr Cogn Disord 22(1):99–107 disease amyloid-beta peptides. J Biol Chem
26. Samieri C, Feart C, Proust-Lima C et al (2011) 280(45):37377–37382. https://doi.
Omega-3 fatty acids and cognitive decline: org/10.1074/jbc.M508246200
modulation by ApoEepsilon4 allele and 39. Ho L, Ferruzzi MG, Janle EM et al (2013)
depression. Neurobiol Aging 32(12):2313– Identification of brain-targeted bioactive
2322. https://doi.org/10.1016/j. dietary quercetin-3-O-glucuronide as a novel
neurobiolaging.2010.03.020 intervention for Alzheimer’s disease. FASEB
27. Huang TL, Zandi PP, Tucker KL et al (2005) J 27(2):769–781. https://doi.org/10.1096/
Benefits of fatty fish on dementia risk are fj.12-212118
stronger for those without APOE epsilon4. 40. Wang J, Ho L, Zhao Z et al (2006)
Neurology 65(9):1409–1414 Moderate consumption of Cabernet
28. Barberger-Gateau P, Raffaitin C, Letenneur Sauvignon attenuates Abeta neuropathology
L et al (2007) Dietary patterns and risk in a mouse model of Alzheimer’s disease.
of dementia: the Three-City cohort study. FASEB J 20(13):2313–2320. https://doi.
Neurology 69(20):1921–1930 org/10.1096/fj.06-6281com
29. Whalley LJ, Deary IJ, Starr JM et al (2008) 41. Boeing H, Bechthold A, Bub A et al (2012)
n-3 Fatty acid erythrocyte membrane con- Critical review: vegetables and fruit in the
tent, APOE {varepsilon}4, and cognitive vari- prevention of chronic diseases. Eur J Nutr
ation: an observational follow-up study in late 51(6):637–663. https://doi.org/10.1007/
adulthood. Am J Clin Nutr 87(2):449–454 s00394-012-0380-y
30. Daiello LA, Gongvatana A, Dunsiger S 42. Xu W, Wang H, Wan Y et al (2017) Alcohol
et al (2015) Association of fish oil supple- consumption and dementia risk: a dose-
ment use with preservation of brain volume response meta-analysis of prospective studies.
and cognitive function. Alzheimers Dement Eur J Epidemiol. https://doi.org/10.1007/
11(2):226–235. https://doi.org/10.1016/j. s10654-017-0225-3
jalz.2014.02.005 43. Samieri C, Feart C, Proust-Lima C et al
31. Yassine HN, Feng Q, Azizkhanian I et al (2011) Olive oil consumption, plasma oleic
(2016) Association of serum docosa- acid, and stroke incidence: the Three-City
hexaenoic acid with cerebral amyloidosis. Study. Neurology 77(5):418–425. https://
JAMA Neurol. https://doi.org/10.1001/ doi.org/10.1212/WNL.0b013e318220abeb
jamaneurol.2016.1924 44. Berr C, Portet F, Carriere I et al (2009)
32. Grimm MO, Mett J, Hartmann T (2016) Olive oil and cognition: results from the
The impact of vitamin E and other fat-soluble three-city study. Dement Geriatr Cogn
vitamins on Alzheimer’s disease. Int J Mol Disord 28(4):357–364. https://doi.
Sci 17(11). https://doi.org/10.3390/ org/10.1159/000253483
ijms17111785 45. Kim YS, Kwak SM, Myung SK (2015)
33. Ransom J, Morgan PJ, McCaffery PJ et al Caffeine intake from coffee or tea and cogni-
(2014) The rhythm of retinoids in the brain. tive disorders: a meta-analysis of observational
J Neurochem 129(3):366–376. https://doi. studies. Neuroepidemiology 44(1):51–63.
org/10.1111/jnc.12620 https://doi.org/10.1159/000371710
38 Cécilia Samieri
Diagnostic Concepts
Chapter 3
Abstract
The self-experienced persistent decline in one’s cognitive abilities in comparison with a previously normal
status is referred to as subjective cognitive decline (SCD). During the last decades, evidence has emerged
about the close relationship between SCD and incident cognitive impairment to such an extent that SCD
is currently considered as a very early marker of future dementia. Here, we first discuss the strengths and
the weaknesses of this concept, and then we describe a procedure to measure SCD accurately. Our goal is
to provide the reader with specific guidelines on how to assess and classify individuals in terms of SCD in
order to identify individuals at high risk of developing cognitive impairment.
Key words Aging, Alzheimer’s disease, Cognitive symptoms, Dementia, Mild cognitive impairment,
Subjective cognitive decline
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_3, © Springer Science+Business Media, LLC 2018
45
46 Miguel A. Fernández-Blázquez et al.
uring the last decades there has been an important increase in the
d
study of SCD as a very early sign of cognitive deterioration.
There are overwhelming epidemiological data in favor of the
relationship between SCD and incident cognitive impairment.
Thus, a recent meta-analysis focused on the longitudinal value of
SCD for detecting later MCI and dementia has shown that, inde-
pendently of the objective memory performance, 6.6% of older
adults with SCD develop MCI per year [15]. In addition, the rate
of progression to dementia among those individuals who report
complaints about their own cognitive performance is also twofold
during a 5-year follow-up period.
Despite increasing evidence indicating that SCD may represent
a very early manifestation of AD [16], little is known about the
clinical role of specific complaints on the transition from normal
aging to cognitive impairment. Three types of cognitive concerns
have demonstrated their usefulness to discriminate between cogni-
tively healthy older adults and MCI [17]; particularly higher scores
in specific complaints on recalling immediate events, executive
functioning, and prospective memory are related to prodromal
stages of dementia. The fact that other types of complaints such as
forgetfulness of objects or spatial orientation did not show differ-
ences between controls and MCI could be explained because the
first of them refers to a high prevalent oversight in the elderly pop-
ulation (“forgetting where you have put something” and “forget-
ting where things are normally kept or looking for them in the
wrong place”) and the second one is an idiosyncratic sign of very
mild dementia (“getting lost or turning in the wrong direction on
a journey, on a walk, or in a building that one really knows because
he or she has been there before”). These findings emphasize that
not all cognitive complaints have the same clinical significance for
predicting cognitive impairment.
However, despite its emerging role as a marker of preclinical
AD, the concept of SCD is not free from some limitations which
are necessary to address. For instance, many terms such as subjec-
tive memory complaints, subjective cognitive decline, or subjective
memory impairment have been used interchangeably to refer to the
same concept. This lack of consensus on a single definition of SCD
affects to the comparison of findings from different investigations
and epidemiological studies. Moreover, there is no accepted
approach among researchers about the assessment of SCD, includ-
ing the mode of administration (structured interview versus ques-
tionnaires), the cognitive domains to be examined (memory versus
non-memory domains), the number of items to be used (one or
two questions versus scales with a large number of items), and the
optimal way to respond the items (opened questions versus multi-
ple choice). Finally, it becomes difficult to determine which com-
plaints underlie AD because there is a close relationship between
48 Miguel A. Fernández-Blázquez et al.
2 Materials
Table 1
Structured SCD questionnaires frequently used in research
3 Methods
3.4 Classification Besides analyzing the implication of specific SCD features as early
of Individuals in SCD signs of AD, information about complaints should be also exam-
Groups ined according to the guidelines proposed by the SCD-I [23].
Following these guidelines, individuals might be grouped in three
different categories pursuant to the extent of SCD reported: (1) no
complaints (NCg); (2) subjective cognitive decline group (SCDg),
when subjects report some kind of cognitive complaint; and (3)
subjective cognitive decline plus (SCD-Pg), when individuals show
complaints in memory plus another cognitive domain and addi-
tionally they fulfill the rest of the criteria for SCD plus.
The classification in any of the three groups of SCD may be
carried out in two steps (a full description of this procedure is
shown in Fig. 1). Initially, based on the overall information gath-
ered both in clinical interviews and in self-administered EMQ,
SCD may be operationally defined as the self-rated presence of
cognitive deterioration using two criteria: (1) at least a positive
response to any yes/no-type question regarding complaints in any
cognitive domain from the neurological interview and (2) scores
above 1 on the SCD scale administered in the neuropsychological
assessment and above 8 on the self-administered EMQ. To be clas-
sified as SCDg, individuals have to mandatorily accomplish both
conditions. Thus, subjects who only fulfill one criterion or neither
of them will be considered as no complaints.
The second step of classification is only applied for those cases
categorized as SCDg. For these individuals some specific features
must be considered such as age at onset of SCD beyond 60 years,
turning up of complaints within the last 5 years, worry associated
with SCD, and feeling of worse performance than others of the
same age group. When all these conditions accompany the self-
experience of decline, then an individual is classified as SCD-Pg.
Fig. 1 Flow diagram for SCD classification. NCg no complaints group, SCDg subjective cognitive decline group,
and SCD-Pg subjective cognitive decline plus group
Table 2
Example of a comprehensive neuropsychological battery and cognitive domains covered
Vallecas Project is shown. All these tests cover the whole spectrum
of cognition.
In addition, scales for measuring functional activities and
neuropsychiatric variables are very important. For instance, to this
end, the functional activities questionnaire (FAQ) [50] can be
administered to collect data with regard to instrumental activities
of daily living; the Geriatric Depression Scale (GDS) [51] and the
State-Trait Anxiety Inventory (STAI) [52] might be also adminis-
tered as part of the neuropsychological battery to quickly estimate
mood and anxiety symptoms.
The Dimensional Structure of Subjective Cognitive Decline 57
4 Conclusions
Fig. 2 Distribution of SCD groups and association with conversion rate to MCI. NCg no complaints group, SCDg
subjective cognitive decline group, and SCD-Pg subjective cognitive decline plus group. This graph is based on
the results published in a previous paper entitled “Specific Features of Subjective Cognitive Decline Predict
Faster Conversion to Mild Cognitive Impairment” (Fernández-Blázquez et al. [55]). No metric multidimensional
scaling was applied in order to analyze the relative position of SCD groups in terms of euclidean distance. The
analysis was based on all significant variables found in nonparametric ANOVAs. Sizes of bubbles indicate
the risk of annual conversion to MCI (the larger is the area, the greater is the risk). As is apparent, SCD-Pg is
the group with the highest rate of conversion (18.9%) followed by SCDg (5.6%) and NCg (4.9%). In addition,
the separation in perceptual map among the three groups indicates that there are three latent profiles underlying
psychiatric and cognitive variables
References
1. Sperling R, Aisen PS, Beckett L et al (2011) Alzheimers Dement 7:263–269. https://doi.
Toward defining the preclinical stages of org/10.1016/j.jalz.2011.03.005
Alzheimer’s disease: recommendations 4. Selkoe DJ (2011) Resolving controversies on
from the National Institute on Aging and the path to Alzheimer’s therapeutics. Nat Med
the Alzheimer’s Association workgroup. 17:1060–1065. https://doi.org/10.1038/
Alzheimers Dement 7:280–292. https://doi. nm.2460
org/10.1016/j.jalz.2011.03.003 5. Hampel H, Lista S, Teipel SJ et al (2014)
2. Albert MS, DeKosky ST, Dickson D et al Perspective on future role of biological mark-
(2011) The diagnosis of mild cognitive impair- ers in clinical therapy trials of Alzheimer’s dis-
ment due to Alzheimer’s disease: recommen- ease: a long-range point of view beyond 2020.
dations from the National Institute on Aging Biochem Pharmacol 88:426–449. https://
and Alzheimer’s Association workgroup. doi.org/10.1016/j.bcp.2013.11.009
Alzheimers Dement 7:270–279. https://doi. 6. Reisberg B, Prichep L, Mosconi L et al (2008)
org/10.1016/j.jalz.2011.03.008 The pre-mild cognitive impairment, subjec-
3. McKhann GM, Knopman DS, Chertkow tive cognitive impairment stage of Alzheimer’s
H et al (2011) The diagnosis of dementia disease. Alzheimers Dement 4:S98–S108.
due to Alzheimer’s disease: recommendations https://doi.org/10.1016/j.jalz.2007.11.017
from the National Institute on Aging- 7. Vannini P, Hanseeuw B, Munro CE et al (2017)
Alzheimer’s Association workgroups on diag- Hippocampal hypometabolism in older adults
nostic guidelines for Alzheimer’s disease. with memory complaints and increased amyloid
60 Miguel A. Fernández-Blázquez et al.
28. Farias ST, Mungas D, Reed BR et al (2008) The clinician. J Psychiatr Res 12:189–198. https://
measurement of everyday cognition (ECog): doi.org/10.1016/0022-3956(75)90026-6
scale development and psychometric proper- 40. Rey A (1941) L’examen psychologique dans
ties. Neuropsychology 22:531–544. https:// les cas d’encéphalopathie traumatique. Arch
doi.org/10.1037/0894-4105.22.4.531 Psychol (Geneve) 28:215–285. http://psyc-
29. Sunderland A, Harris JE, Gleave net.apa.org/psycinfo/1943-03814-001
J (1984) Memory failures in everyday 41. Peña-Casanova J, Gramunt-Fombuena N,
life following severe head injury. J Clin Quiñones-Úbeda S et al (2009) Spanish mul-
Neuropsychol 6:127–142. https://doi. ticenter normative studies (NEURONORMA
org/10.1080/01688638408401204 project): norms for the rey-osterrieth com-
30. Jorm AF, Jacomb PA (1989) The Informant plex figure (copy and memory), and free
Questionnaire on Cognitive Decline in the and cued selective reminding test. Arch Clin
Elderly (IQCODE): socio-demographic cor- Neuropsychol 24:371–393
relates, reliability, validity and some norms. 42. Buschke H (1984) Cued recall in amnesia.
Psychol Med 19:1015–1022 J Clin Neuropsychol 6:433–440. https://doi.
31. Crook TH, Feher EP, Larrabee GJ (1992) org/10.1080/01688638408401233
Assessment of memory complaint in age- 43. Peña-Casanova J, Quiñones-Ubeda S,
associated memory impairment: the MAC- Gramunt-Fombuena N et al (2009)
Q. Int Psychogeriatr 4:165–176. https://doi. Spanish Multicenter Normative Studies
org/10.1017/S1041610292000991 (NEURONORMA Project): norms for verbal
32. Gilewski MJ, Zelinski EM, Schaie KW (1990) fluency tests. Arch Clin Neuropsychol 24:395–
The Memory Functioning Questionnaire for 411. https://doi.org/10.1093/arclin/
assessment of memory complaints in adult- acp042
hood and old age. Psychol Aging 5:482–490 44. Wechsler D (1997) Wechsler Adult Intelligence
33. Rami L, Mollica MA, García-Sanchez C et al Scale-III. The Psychological Corporation, San
(2014) The Subjective Cognitive Decline Antonio, TX
Questionnaire (SCD-Q): a validation study. 45. Lee GP, Loring DW, Newell J, McCloskey L
J Alzheimers Dis 41:453–466. https://doi. (1994) Figural fluency on the Five-Point Test:
org/10.3233/JAD-132027 preliminary normative and validity data. Int
34. Youn JC, Kim KW, Lee DY et al (2009) Neuropsychol Soc Progr Abstr, p 51
Development of the Subjective Memory 46. Wilson BA, Alderman N, Burgess PW et al
Complaints Questionnaire. Dement Geriatr (1996) The behavioural assessment of the dys-
Cogn Disord 27:310–317. https://doi. executive syndrome. Thames Valley Company,
org/10.1159/000205512 Bury St Edmunds
35. Squire LR, Wetzel CD, Slater PC (1979) 47. Fernández-Blázquez MA, Ruiz-Sánchez de
Memory complaint after electroconvulsive León JM, López-Pina JA et al (2012) A new
therapy: assessment with a new self-rating shortened version of the Boston Naming
instrument. Biol Psychiatry 14:791–801 Test for those aged over 65: an approach
36. Olazarán J, Valentí M, Frades B et al (2015) from item response theory. Rev Neurol
The Vallecas Project: a cohort to identify early 55:399–407
markers and mechanisms of Alzheimer’s dis- 48. Peña-Casanova J (1990) Programa inte-
ease. Front Aging Neurosci 7:181. https:// grado de exploración neuropsicológica-Test
doi.org/10.3389/fnagi.2015.00181 Barcelona. Masson, Barcelona
37. Montejo Carrasco P, Montenegro Peña M, 49. Warrington EK, James M (1991) The visual
Sueiro MJ (2012) The Memory Failures object and space perception battery. Thames
of Everyday (MFE) test: normative data Valley Test Company, Bury St Edmunds
in adults. Span J Psychol 15:1424–1431. 50. Pfeffer RI, Kurosaki TT, Harrah CH et al
https://doi.org/10.5209/rev_SJOP.2012. (1982) Measurement of functional activities
v15.n3.39426 in older adults in the community. J Gerontol
38. Hughes CP, Berg L, Danziger WL (1982) A 37:323–329. https://doi.org/10.1093/
new clinical scale for the staging of dementia. geronj/37.3.323
Br J Psychiatry 140:566–572. https://doi. 51. Yesavage JA, Brink TL, Rose TL et al (1982)
org/10.1192/bjp.140.6.566 Development and validation of a geriat-
39. Folstein MF, Folstein SE, McHugh PR (1975) ric depression screening scale: a preliminary
“Mini-mental state”: a practical method for report. J Psychiatr Res 17:37–49. https://doi.
grading the cognitive state of patients for the org/10.1016/0022-3956(82)90033-4
62 Miguel A. Fernández-Blázquez et al.
52. Spielberger C, Gorsuch R, Leshene R (1970) involved in the relationship between mood
Manual for the State-Trait Anxiety Inventory. and mild cognitive impairment. J Alzheimers
Consulting Psychologists Press, Palo Alto, CA Dis 48(Suppl 1):S115–S123. https://doi.
53. Molinuevo JL, Rabin LA, Amariglio R et al org/10.3233/JAD-150371
(2017) Implementation of subjective cog- 55. Fernández-Blázquez MA, Ávila-Villanueva M,
nitive decline criteria in research studies. Maestú F, Medina M (2016) Specific features
Alzheimers Dement 13:296–311. https://doi. of subjective cognitive decline predict faster
org/10.1016/j.jalz.2016.09.012 conversion to mild cognitive impairment.
54. Yates JA, Clare L, Woods RT, Matthews FE J Alzheimers Dis 52:271–281. https://doi.
(2015) Subjective memory complaints are org/10.3233/JAD-150956
Chapter 4
Abstract
Empirical support has established cognitive reserve (CR) well as a concept since its inception three decades
ago, and most brain researchers subscribe to some version of CR as a collection of subject factors that influence
cognitive performance beyond simple brain structural health. We give a simple but precise analytic recipe to
test requirements for any plausible cognitive reserve candidate based on brain imaging. Gradations of partial
fulfillment of some but not all of these requirements are possible.
Key words Cognitive reserve, Brain aging, Resting-state fMRI, Cognitive aging
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_4, © Springer Science+Business Media, LLC 2018
63
64 Yaakov Stern and Christian Habeck
1.1 Cognitive 1. Cognitive reserve has well-established proxies like education, intel-
Reserve Axioms ligence, occupational attainment, leisure activities, and other sub-
ject variables that express cognitively stimulating engagement.
2. Cognitive reserve is either an independent factor that influences
cognitive performance in addition to brain structure or a moder-
ating factor that influences the relationship between brain struc-
ture and cognitive performance; these two manifestations are not
exclusive and can conveniently be incorporated in linear regres-
sion models that contain (1) brain structure and (2) the putative
CR measure as direct effects and (3) an interaction term.
3. Although some of the cognitive reserve proxies might corre-
late with brain structural measures, cognitive reserve itself is
not based on brain structure; this entails in particular that:
●● It is not a mediating influence of brain structure on cogni-
tive performance.
●● It should be uncorrelated with brain structural measures.
These three axioms can be used to constrain the derivation of plau-
sible cognitive reserve candidates. Specifically, (1) cognitive reserve
should display correlations with some, or all, of the preexisting proxies
and (2) should show direct effects on cognitive performance in addi-
tion to brain structure or moderate the effects of brain structure on
performance while (3) being uncorrelated to brain structure. This last
point is contentious with some researchers who might refer to obvious
correlations between brain structure and CR proxies like intelligence
or education. Recently, we have laid out how such correlations can be
conceptualized as brain maintenance [18] which should be considered
as separate from, and in correlational terms orthogonal to, cognitive
reserve. CR proxies thus might correlate with cognitive reserve and
brain structure, but the latter correlation strictly separates them from
being a viable candidate for cognitive reserve directly. In other words,
while CR proxies might indicate the state of somebody’s brain struc-
tural health and how well the brain has been preserved in the course of
aging, cognitive reserve strictly speaks to how well somebody utilizes their
brain regardless of how well it has been preserved. CR proxies thus consist
Deriving and Testing Cognitive Reserve 65
of two parts: (1) a part that correlates with brain structure and points
to possible formative influences to build and maintain a healthy brain
and (2) cognitive reserve, which in our rigorous definition is indepen-
dent of brain structure. The Venn diagram in Fig. 1 gives a simple
schematic explanation.
2 Materials
3 Methods
have accuracy comparable to manual labeling [19, 20]. From this label-
ing, a set of volumetric ROIs was defined [21]. Although the estima-
tion procedure is automated, we manually check the accuracy of the
spatial registration and the white matter and gray matter segmentations
following the analytic procedures outlined in Fjell et al. [22] . The
calculated volume within each region was adjusted for variations in
individual’s intracranial brain volume which is measured using
BrainWash [23]. Using FreeSurfer software, cortical thickness was
measured by first reconstructing the gray/white matter boundary [24]
and the cortical surface and then calculating the distances between
these surfaces at each point across the cortical mantle. The maps pro-
duced are capable of detecting submillimeter differences between
groups [25]. Although the thickness estimation procedure is auto-
mated, we manually check the accuracy of the spatial registration and
the white matter and gray matter segmentations following the analytic
procedures outlined in Fjell et al. [22]. Using a validated automated
labeling system [20], FreeSurfer divided the cortex into 33 different
gyral-based areas in each hemisphere and calculates the mean thickness
in each area.
Furthermore, resting-state fMRI was collected for either 5 or
9.5 min, while the participant is instructed to lie still with their eyes
closed, to not think of any one thing in particular, and to not fall asleep.
MCFLIRT is used to register all the volumes to a reference image [26]
followed by scrubbing and motion correction as described in Power
et al. [27] and incorporating the recommendation of replacing deleted
volumes with zero volumes prior to filtering [28]. Using the FSL com-
mand “flsmaths –bptf t,” the motion-corrected signals are passed
through a band-pass filter with the cutoff frequencies of 0.01 and
0.08 Hz. After filtering, the first three volumes are discarded due to the
lag of the digital filter. Finally, we residualized the motion-corrected,
scrubbed, and temporally filtered volumes by regressing out the FD,
RMSD, left- and right-hemisphere white matter, and lateral-ventricular
signals [29]. We followed the taxonomy of functional units presented
by Power et al. [30] and produced residualized and motion-corrected
resting BOLD time series at 264 ROI locations. This data reduction
has the advantage that it enables easy computation of global functional
connectivity between all 264 × 263/2 = 34,716 node pairs.
The demonstrations that follow used omnibus measures that col-
lapse across all ROIs and all ROI pairs. This was done for the sake of
simplicity; an ecologically valid research program most likely would con-
sider structural variables and CR candidates with a topographic specific-
ity. In particular functional connectivity did not respect the intricate
taxonomy of different resting-state networks [30] and did not break
down connectivity into intra- and inter-network connectivity, which
would be advisable for real basic and diagnostic research questions.
3.1 Consideration Deriving a candidate measure for cognitive reserve should ideally fulfill
of a Connectivity- our three above postulates. Further, ideally it should be motivated with
Based Measure a convincing mechanistic insight or hypothesis, which suggests
Deriving and Testing Cognitive Reserve 67
3.2 Test Let us now turn to our three postulates, which represent the linchpin
of Postulates of this report. The variability measure is denoted as stdZ and is a scaler
for Cognitive Reserve value, i.e., it gives one number for each of the 451 participants.
1. The first postulate concerns a relationship with some well-
known CR proxies. We checked the correlation with NART
and years of education. The correlation with education years
was nonsignificant (R = −0.0376, p = 0.4264), while it was
modestly significant with NART (R = −0.1076, p = 0.0267).
Thus, people with higher verbal intelligence had slightly less
regional variability of functional connectivity in their brains.
2. The test of the second postulate is computationally somewhat
more demanding and involves many more, potentially arbi-
trary, decisions on account of the analyst. The second postulate
embodies a key requirement: Our CR candidate should
account for a particular cognitive performance above and
beyond brain structural independent variables and possibly
moderate the structure-cognition relationship. Thus CR could
be an independent factor that determines cognitive perfor-
mance in addition to brain structure, or a moderator, or both.
We can write down the regression equation
Dep = sIVβ + CRγ + sIV × CRδ + ε .
where Dep denotes our cognitive outcome measure, vocabu-
lary, sIV denotes a structural independent variable, CR stands for
the cognitive reserve candidate, and sIV × CR denotes the interac-
tion term. Greek letters βγδ denote regression weights, and ɛ stands
for the unexplained residual of Dep. It is easily conceivable that
there are more than just one structural independent variable and
correspondingly more than one interaction term. Concretely, we
will use mean cortical volume and mean cortical thickness, which
results in direct effects of two structural independent variables and
CR, and two interaction terms. More fundamental is the depen-
dency of the postulate check on the choice of dependent and inde-
68 Yaakov Stern and Christian Habeck
Table 1
Linear regression to explain vocabulary performance. Listed are T-
and p-values for each independent variable
Independent variable T P
Mean cortical volume 3.0931 0.0021
Mean cortical thickness −1.7053 0.0889
stdZ −2.6077 0.0094
stdZ × volume 0.2702 0.7871
stdZ × thickness 0.6915 0.4896
Deriving and Testing Cognitive Reserve 69
4 Conclusions
References
1. Verghese J, Lipton RB, Katz MJ et al (2003) rate of hippocampal atrophy. PLoS One
Leisure activities and the risk of dementia in 3(7):e2598. https://doi.org/10.1371/jour-
the elderly. N Engl J Med 348(25):2508–2516 nal.pone.0002598
2. Hu G, Sarti C, Jousilahti P et al (2005) 8. Gates N, Valenzuela M (2010) Cognitive exercise
Leisure time, occupational, and commuting and its role in cognitive function in older adults.
physical activity and the risk of stroke. Stroke Curr Psychiatry Rep 12(1):20–27. https://doi.
36(9):1994–1999 org/10.1007/s11920-009-0085-y
3. Rovio S, Kareholt I, Helkala EL et al (2005) 9. Bonaiuto S, Rocca W, Lippi A (1990) Impact
Leisure-time physical activity at midlife and of education and occupation on prevalence
the risk of dementia and Alzheimer's disease. of Alzheimer’s disease (AD) and multi-infarct
Lancet Neurol 4(11):705–711. https://doi. dementia (MID) in Appignano, Macerata
org/10.1016/S1474-4422(05)70198-8 Province, Italy. Neurology 40(suppl 1):346
4. Helzner EP, Scarmeas N, Cosentino S et al 10. Cohen CI (1994) Education, occupation, and
(2007) Leisure activity and cognitive decline Alzheimer’s disease. JAMA 272(18):1405.
in incident Alzheimer disease. Arch Neurol Author reply 1406
64(12):1749–1754 11. Gun RT, Korten AE, Jorm AF et al (1997)
5. Akbaraly TN, Portet F, Fustinoni S et al (2009) Occupational risk factors for Alzheimer disease:
Leisure activities and the risk of dementia in a case-control study. Alzheimers Dis Assoc
the elderly: results from the Three-City Study. Disord 11(1):21–27
Neurology 73(11):854–861. https://doi. 12. Helmer C, Letenneur L, Rouch I et al (2001)
org/10.1212/WNL.0b013e3181b7849b Occupation during life and risk of dementia in
6. Scarmeas N, Levy G, Tang MX et al (2001) French elderly community residents. J Neurol
Influence of leisure activity on the inci- Neurosurg Psychiatry 71(3):303–309
dence of Alzheimer’s disease. Neurology 13. Anttila T, Helkala EL, Kivipelto M et al (2002)
57(12):2236–2242 Midlife income, occupation, APOE status, and
7. Valenzuela MJ, Sachdev P, Wen W et al (2008) dementia: a population-based study. Neurology
Lifespan mental activity predicts diminished 59(6):887–893
70 Yaakov Stern and Christian Habeck
Abstract
The hallmark protein aggregates required for a neuropathological diagnosis of Alzheimer’s disease are
intracellular neurofibrillary tangles and neuropil threads consisting of hyperphosphorylated tau, extracellular
parenchymal amyloid β plaques, and neuritic plaques. In this chapter, we describe the neuropathological
criteria for the neuropathological diagnosis of Alzheimer’s disease, as well as the fixation, processing, and
histology protocols required for the assessment of post-mortem human brains.
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_5, © Springer Science+Business Media, LLC 2018
71
72 Johannes Attems et al.
1.1.2 Neurofibrillary Neurofibrillary tangles (NFTs) and neuropil threads (NTs) are
Tangles and Neuropil aggregates of hyperphosphorylated microtubule-associated protein
Threads (MAP) tau (HP-T): a microtubule-stabilizing protein that is abun-
dant in axons and dendrites. Six isoforms of MAP tau exist in the
human brain, and aggregates of 3-repeat (3R) and 4-repeat (4R)
isoforms are seen in AD. Under pathological conditions, MAP tau
is hyperphosphorylated resulting in the disintegration of microtu-
bules and aggregation of insoluble filaments/fibrils composed of
HP-T [2]. NFTs/NTs are composed of aggregates of HP-T, while
so called pre-tangles represent non-aggregated HP-T and are con-
sidered to be precursors of NFT/NT. Using appropriate immuno-
histochemical antibodies, i.e., AT8 antibody, both NFT and NT
can be visualized for histological assessment. NFTs are located in
neuronal cell bodies and NTs in axons and dendrites, the latter
appearing in immunohistochemically stained sections as immu-
nopositive threads in the neuropil (Fig. 2). In AD, NFT and NT
primarily manifest in trans-entorhinal and entorhinal cortices and
then gradually spread toward the hippocampus and isocortex [3],
and this pattern is the basis for diagnostic Braak NFT stages [4]
(see Sect. 3.6.1). Autopsy studies have revealed pre-tangle HP-T
was found in the locus coeruleus in the majority of individuals
under 30 years of age who were devoid of cortical HP-T pathology,
suggesting that HP-T pathology begins in the locus coeruleus
rather than the trans-/entorhinal cortex [5, 6], but this is
controversial [7]. Importantly, the extent of neocortical NFT/NT
has been shown to correlate with cognitive deficits [8].
Pathological Classification of Alzheimer’s Disease 73
Fig. 1 Comparison between formalin-fixed tissue slices taken at the level of the
anterior hippocampus of an individual without dementia (A) and a patient with
severe AD. ((B) and (C)) There are marked enlargement of the lateral ventricle
(blue asterisk), considerable atrophy of the medial temporal lobe (red asterisk),
as well as thinning of the gyri and deepening of the sulci (black arrowheads).
However, the substantial nigra of the midbrain remains fairly well pigmented (c).
Photographs courtesy of Dr. Christopher Morris and the Newcastle Brain Tissue
Resource. Both scale bars represent 1 cm
1.1.3 Amyloid β Plaques Amyloid β (Aβ) peptides (4kDA) are generated by β- and γ-secretase
cleavage of the transmembraneous amyloid precursor protein
(APP) and comprise peptides terminating at carboxyl terminus 40
(Aβ40) and 42 (Aβ42) [9]. While Aβ40 constitutes the majority of
cerebral Aβ (over 95%), Aβ42 aggregates more readily and, there-
fore, is believed to initiate the formation of oligomers, fibrils, and
plaques [10, 11]. Aβ aggregates extracellularly, and using appro-
priate antibodies, i.e., 4G8 antibody, various morphological forms
of Aβ aggregates can be detected. The main parenchymal Aβ
deposits are diffuse or focal: diffuse Aβ deposits are usually large
(up to several 100 μm) and show ill-defined borders and are often
referred to as “fleecy” and “subpial band-like,” while focal Aβ
plaques incorporate a dense, spherical-like accumulation of Aβ
peptides (Fig. 3). The topographical locations of Aβ deposits
follow a distinct sequence that can be staged diagnostically using
Thal Aβ phases [12]. Cerebral Aβ depositions are a hallmark of
AD, but in the majority of clinicopathological correlative studies,
74 Johannes Attems et al.
Fig. 2 Photomicrographs of neurofibrillary tangles and neuropil threads. (A) Distribution of HP-T immunopositiv-
ity in the hippocampus, subiculum, transentorhinal, and occipital-temporal cortex of an Alzheimer’s disease
case. (Ai) Magnified image of neurofibrillary tangles and neuropil threads in the CA1 region of the hippocam-
pus. (Aii) Neurofibrillary tangles encompass immunopositivity within the neuronal soma (black arrow); neuropil
thread comprises of immunopositivity in the neuronal process (red arrow head). CA1 and CA4, hippocampal
cornu ammonis (Ammon’s horn) sectors 1 and 4; GR, granule cell layer of the dentate gyrus; Sub, subiculum;
Trans Ent, transentorhinal cortex; OTC, occipital-temporal cortex. Immunohistochemistry with antibody against
hyperphosphorylated tau, AT8. Scale bars: A1, 50 μm; Aii, 10 μm
1.1.4 Neuritic Plaques Neuritic plaques (NP) consist of a focal deposit of Aβ42 [13], with
an external halo (the corona) of HP-T-positive processes forming
dystrophic neurites (Fig. 4). NPs are strongly associated with AD,
and assessment of the presence and density of NP is performed
using the age-adjusted criteria of the Consortium to Establish a
Registry for Alzheimer’s Disease (CERAD) [14] (see Sect. 3.6.3).
1.1.5 Cerebral Amyloid Cerebral amyloid angiopathy (CAA) is defined as the deposition of
Angiopathy a congophilic material (i.e., positive staining with a Congo red dye)
in cerebral leptomeningeal and intracortical arteries, arterioles,
capillaries, and rarely veins. CAA is also referred to as congophilic
amyloid angiopathy [15, 16] and is strongly associated with
AD-related pathology, present in 80–100% of AD cases [17, 18].
CAA is characterized by the deposition of predominantly Aβ40 [9]
in the tunica media of vessel walls that eventually spreads into all
vessel wall layers causing vessel wall thickening, smooth muscle cell
migration, and overall disruption of vessel architecture, although
Fig. 4 Photomicrographs of neuritic plaques. (A) Distribution of neuritic plaques in the hippocampus, subiculum,
transentorhinal, and occipital-temporal cortex of an Alzheimer’s disease case. (Ai) Magnified image of neurotic
plaques in the CA1 region of the hippocampus. (Aii) Neuritic plaque consisting of a Aβ core (red arrow head)
and dystrophic neurites consisting of HPτ (blue arrow). CA1 and CA4, hippocampal cornu ammonis (Ammon’s
horn) sectors 1 and 4; GR, granule cell layer of the dentate gyrus; Sub, subiculum; Trans Ent, transentorhinal
cortex; OTC, occipital-temporal cortex; Aβ, amyloid beta; HPτ, hyperphosphorylated tau. Histological staining
with Gallyas silver stain. Scale bars: Ai, 50 μm; Aii, 10 μm
76 Johannes Attems et al.
2 Materials
3 Methods
3.1 Fresh Brain The cerebrum, and spinal cord if required, is removed from the
Dissection deceased patient following standard protocols. All tissue, blood,
and Fixation and CSF must be treated as a potentially infectious case. The
pathologist and assisting personnel must wear a gown, nitrile
gloves, and shoe covers, and dissection of neurologic tissues is
carried out in a fume hood. The cerebrum, cerebellum, and brain-
stem are inspected and weighed, and the cerebrum length is mea-
sured from the frontal pole to the occipital pole. The arteries of the
Pathological Classification of Alzheimer’s Disease 79
3.2 Fixed Dissection Tissue is fixed for 4 weeks at room temperature. The right cere-
and Embedding brum is weighed, and length is measured from the frontal pole to
the occipital pole. The arteries of the circle of Willis are examined
for the presence of atherosclerotic changes. The midbrain is
removed from the cerebrum cutting in between the mammillary
body and superior colliculus. The cerebral hemisphere, cerebellum,
and brainstem are then serially dissected at 7 mm intervals (Fig. 6).
Regions (including specific Brodmann areas (BA)) required for
Fig. 6 Following fixation, the right hemisphere (A) is dissected at 7 mm intervals (B) as are the fixed semi-
brainstem and right cerebellum (C). Brain regions important of the neuropathological diagnosis of AD are then
sub-dissected and processed into paraffin wax. Photographs courtesy of Dr. Christopher Morris and the
Newcastle Brain Tissue Resource. Scale bar in A and B represents 2 cm and 1 cm in C
80 Johannes Attems et al.
Table 1
Brain regions (including specific Brodmann areas, BA) required for
neuropathological diagnosis
Table 2
Three-day CNS processing schedule
Step Time
4% formalin (no marble chips) 34 min
50% ethanol 2 h
70% ethanol 3 h 50 min
95% ethanol 5 h
100% ethanol 6 h
100% ethanol 6 h
100% ethanol 8 h
Chloroform 3 h
Chloroform 6 h
Chloroform 7 h
Histology wax 5 h 60 °C (pressure/
vacuum)
Histology wax 5 h 60 °C (pressure/
vacuum)
Histology wax 5 h 60 °C (pressure/
vacuum)
3.4 Gallyas Staining Silver staining such as Gallyas is used to visualize dystrophic neu-
rites associated with neuritic plaques; however, amyloid deposits
are not detected using this method. The protocol is as follows:
1. Sections placed in 60 °C oven to aid dewaxing.
2. Deparaffinize sections in two changes of xylene (5 min each).
3. Rehydrate through decreasing concentrations of alcohol
(2 × 100%, 1 × 75%, 1 × 50%, 2 min each) to water.
4. Postfix in 4% formalin (10 min).
5. Wash in dH2O.
6. Immerse in 1% potassium permanganate (5 min).
7. Wash in dH2O.
82 Johannes Attems et al.
Table 3
Optimum antibody dilutions and antigen retrieval protocol
Working
dilution
Antibody Target Host Manufacturer (in TBS) Antigen retrieval
AT8 Hyperphosphorylated Mouse Innogenetics, 1:4000 Immersion in 0.01 m.L-1
tau Ser202/205 Ghent, citrate buffer (pH 6) and
(HP-τ) Belgium microwaving for 10 min
4G8 β amyloid 17-24 Mouse Covance, 1:15,000 Immersion in concentrated
Alnwick, UK formic acid for 1 h
Pathological Classification of Alzheimer’s Disease 83
3.6.1 Assessment Braak stages describe the pattern of neurofibrillary pathology (NTs
of Hyperphosphorylated and NFTs) as outlined by Braak and colleagues [4]. Subsequently,
Tau (NTs and NFTs) the BrainNet Europe Consortium identified four distinct anatomi-
cal regions required to accurately assign Braak stage, the anterior
hippocampus at the level of the uncus, posterior hippocampus at
84 Johannes Attems et al.
Fig. 7 Schematic illustrating semiquantitative scoring utilized in Braak staging for assessment of neurofibril-
lary tangles (NFTs) and neuropil threads (NTs) (A). Regions are classed as having mild pathology when immu-
noreactive structures positive for AT8 antibody are barely noted at X100 magnification. Moderate pathology is
assigned when immunoreactive structures are easily visible at ×100 magnification, while the severe score is
assigned when NFTs and NTs are visible even without a microscope. Specific brain regions are required to
assign Braak stages (B) including the anterior hippocampus at the level of the uncus (i), the posterior hippo-
campus at the level of the lateral geniculate (ii), and the visual cortex including the calcarine fissure (iii). The
semiquantitative stage is important when assessing Braak stages as Braak stage I can be assigned with mild
pathology in the transentorhinal cortex (green square, Bi); however, the remaining stages require pathology of
at least moderate density to qualify as the next Braak stage (orange or red squares, Bi, Bii, and Biii). For a full
description of regions affected by each stage, see Sect. 3.6.1. Scale bar represents 100 μm and is valid for all
photomicrographs
Fig. 8 Diagram illustrating progression of amyloid β (Aβ) through the brain. Aβ plaques are first seen in phase
1 in the neocortex, in particular the inferior parietal and superior temporal gyrus (A). In phase 2, Aβ extends to
the entorhinal region, cornu ammonis (CA) sector 2 of the hippocampus and the cingulate cortex (B). Subcortical
structures are affected in phase 3 including striatum, thalamus, hypothalamus, and claustrum (C). Phase 4 is
characterized by Aβ deposits in the brainstem including the substantia nigra and medulla oblongata (D), and
finally Aβ extends to the molecular layer of the cerebellum in phase 5 (E)
Fig. 9 Photomicrographs stained with Gallyas silver stain, demonstrating semiquantitative scoring of neuritic
plaque pathology. Density of plaques per square millimeter is assessed at X100 magnification and described
as mild (scored A) (A), moderate (scored B) (B), or severe (scored C) (C). Arrows point to individual neuritic
plaques. Scale bar represents 100 μm and is also representative of B and C
3.6.4 Determining Level Individual “scores” taken from Braak stages, Thal phases, and
of AD Neuropathologic CERAD assessment are combined to determine the level of AD
Change neuropathologic change using guidelines set out by the National
Pathological Classification of Alzheimer’s Disease 87
Fig. 10 The level of AD neuropathologic change is determined by considering all three pathologies. Amyloid β
Thal phase (A score), Braak stage (B score), and CERAD (C score) are combined to determine the level of AD
neuropathologic change as no, low, intermediate, or high. CERAD, Consortium to Establish a Registry for
Alzheimer’s disease
Table 4
Protocol to assess CAA severity
Type of CAA
assessed 0 1 2 3
Parenchymal CAA Absent Scant Aβ deposition Some circumferential Widespread circumferential
CAA CAA
Meningeal CAA Absent Scant Aβ deposition Some circumferential Widespread circumferential
CAA CAA
Capillary CAA Absent Present NA NA
Vasculopathy Absent Occasional vessel Many vessels affected NA
affected
3.7 Cerebral Amyloid Although not required for the neuropathological diagnosis of AD,
Angiopathy it is important to note the level of CAA when considering Aβ
depositions in post-mortem tissue. A recent consensus protocol has
provided a semiquantitative scoring system to assess parenchymal
CAA, meningeal CAA, capillary CAA, and vasculopathy [24].
Using immunohistochemically sections for Aβ from frontal, tem-
poral, parietal, and occipital cortices (for protocol, see Sect. 3.5),
the consensus-based protocol for assigning CAA severity is out-
lined in Table 4.
88 Johannes Attems et al.
4 Conclusions
5 Notes
Acknowledgments
We thank Lynne Ramsay and Ros Hall for their technical expertise
and assistance in writing this chapter and Dr. Christopher Morris
and the Newcastle Brain Tissue Resource for providing the photo-
graphic images.
References
1. Halliday GM, Double KL, Macdonald V et al 3. Braak H, Braak E (1991) Neuropathological
(2003) Identifying severely atrophic cortical stageing of Alzheimer-related changes. Acta
subregions in Alzheimer's disease. Neurobiol Neuropathol 82(4):239–259
Aging 24(6):797–806 4. Braak H, Alafuzoff I, Arzberger T et al (2006)
2. Ballatore C, Lee VM, Trojanowski JQ (2007) Staging of Alzheimer disease-associated neuro-
Tau-mediated neurodegeneration in fibrillary pathology using paraffin sections and
Alzheimer's disease and related disorders. Nat immunocytochemistry. Acta Neuropathol
Rev Neurosci 8(9):663–672. https://doi. 112(4):389–404. https://doi.org/10.1007/
org/10.1038/nrn2194 s00401-006-0127-z
Pathological Classification of Alzheimer’s Disease 89
5. Braak H, Del Tredici K (2011) The pathologi- 17. Attems J (2005) Sporadic cerebral amyloid
cal process underlying Alzheimer's disease in angiopathy: pathology, clinical implications,
individuals under thirty. Acta Neuropathol and possible pathomechanisms. Acta
121(2):171–181. https://doi.org/10.1007/ Neuropathol 110(4):345–359. https://doi.
s00401-010-0789-4 org/10.1007/s00401-005-1074-9
6. Braak H, Thal DR, Ghebremedhin E et al 18. Attems J, Jellinger KA (2004) Only cerebral
(2011) Stages of the pathologic process in capillary amyloid angiopathy correlates with
Alzheimer disease: age categories from 1 to Alzheimer pathology--a pilot study. Acta
100 years. J Neuropathol Exp Neurol Neuropathol 107(2):83–90. https://doi.
70(11):960–969. https://doi.org/10.1097/ org/10.1007/s00401-003-0796-9
NEN.0b013e318232a379 19. Thal DR, Ghebremedhin E, Orantes M et al
7. Attems J, Thomas A, Jellinger K (2012) (2003) Vascular pathology in Alzheimer dis-
Correlations between cortical and subcortical ease: correlation of cerebral amyloid angiopa-
tau pathology. Neuropathol Appl Neurobiol thy and arteriosclerosis/lipohyalinosis with
38(6):582–590. https://doi. cognitive decline. J Neuropathol Exp Neurol
org/10.1111/j.1365-2990.2011.01244.x 62(12):1287–1301
8. Giannakopoulos P, Herrmann FR, Bussiere T 20. Pfeifer LA, White LR, Ross GW et al (2002)
et al (2003) Tangle and neuron numbers, but Cerebral amyloid angiopathy and cognitive
not amyloid load, predict cognitive status in function: the HAAS autopsy study. Neurology
Alzheimer's disease. Neurology 58(11):1629–1634
60(9):1495–1500 21. Vinters HV, Gilbert JJ (1983) Cerebral amy-
9. Glenner GG, Wong CW (1984) Alzheimer's loid angiopathy: incidence and complications
disease: initial report of the purification and in the aging brain. II. The distribution of amy-
characterization of a novel cerebrovascular loid vascular changes. Stroke 14(6):924–928
amyloid protein. Biochem Biophys Res 22. Tomonaga M (1981) Cerebral amyloid angi-
Commun 120(3):885–890 opathy in the elderly. J Am Geriatr Soc
10. Naslund J, Haroutunian V, Mohs R et al 29(4):151–157
(2000) Correlation between elevated levels of 23. Thal DR, Ghebremedhin E, Rub U et al
amyloid beta-peptide in the brain and cognitive (2002) Two types of sporadic cerebral amyloid
decline. JAMA 283(12):1571–1577 angiopathy. J Neuropathol Exp Neurol
11. Younkin SG (1995) Evidence that a beta 42 is 61(3):282–293
the real culprit in Alzheimer's disease. Ann 24. Love S, Chalmers K, Ince P et al (2014)
Neurol 37(3):287–288. https://doi. Development, appraisal, validation and imple-
org/10.1002/ana.410370303 mentation of a consensus protocol for the
12. Thal DR, Rub U, Orantes M et al (2002) assessment of cerebral amyloid angiopathy in
Phases of a beta-deposition in the human brain post-mortem brain tissue. Am J Neurodegener
and its relevance for the development of Dis 3(1):19–32
AD. Neurology 58(12):1791–1800 25. Montine T, Phelps C, Beach T et al (2012)
13. Guntert A, Dobeli H, Bohrmann B (2006) National Institute on Aging–Alzheimer’s
High sensitivity analysis of amyloid-beta pep- Association guidelines for the neuropatho-
tide composition in amyloid deposits from logic assessment of Alzheimer’s disease: a
human and PS2APP mouse brain. Neuroscience practical approach. Acta Neuropathologica
143(2):461–475. https://doi.org/10.1016/j. 123:1–11
neuroscience.2006.08.027 26. Hyman BT, Phelps CH, Beach TG et al (2012)
14. Mirra SS, Heyman A, McKeel D et al (1991) National Institute on Aging-Alzheimer's
The consortium to establish a registry for Association guidelines for the neuropathologic
Alzheimer's disease (CERAD). Part assessment of Alzheimer's disease. Alzheimer's
II. Standardization of the neuropathologic & dementia : the journal of the Alzheimer's
assessment of Alzheimer's disease. Neurology Association 8(1):1–13. https://doi.
41(4):479–486 org/10.1016/j.jalz.2011.10.007
15. Attems J, Jellinger K, Thal DR et al (2011) Review: 27. Alafuzoff I, Arzberger T, Al-Sarraj S et al
sporadic cerebral amyloid angiopathy. Neuropathol (2008) Staging of neurofibrillary pathology in
Appl Neurobiol 37(1):75–93. https://doi. Alzheimer's disease: a study of the BrainNet
org/10.1111/j.1365-2990.2010.01137.x Europe consortium. Brain pathol (Zurich,
16. Vinters HV (1987) Cerebral amyloid angiopa- Switzerland) 18(4):484–496. https://doi.
thy. A critical review. Stroke 18(2):311–324 org/10.1111/j.1750-3639.2008.00147.x
Part III
Abstract
The recent paradigm shift toward a more biologically oriented definition of Alzheimer’s disease (AD) in
clinical settings increases the importance of biomarkers of AD. The established cerebrospinal fluid (CSF)
biomarkers of AD, reflecting both amyloidopathy and tauopathy, are being increasingly incorporated into
both diagnostic guidelines, enabling the diagnosis of AD independently of clinical symptoms, and inclu-
sionary criteria for clinical trials. The present chapter provides an overview of the clinical utility of the three
established CSF AD biomarkers and covers both clinical and methodological issues. A brief summary is
given on relevant laboratory techniques to determine levels of AD CSF biomarkers; methodological and
clinical challenges in the field are also discussed.
Key words Cerebrospinal fluid amyloid β 1–42, Amyloid β 1–40, Total tau, Hyperphosphorylated tau,
Preclinical Alzheimer’s disease, Prognosis, Diagnosis, ELISA, Luminex xMAP, MSD multi-array
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_6, © Springer Science+Business Media, LLC 2018
93
94 Panagiotis Alexopoulos and Chaido Sirinian
2 Materials
3 Methods
3.1 Enzyme-Linked Levels of Aβ40 and Αβ42 can be determined using high sensitivity
Immunosorbent Assay ELISA kits (RE59651 and RE59661) from IBL International
(ELISA) (Hamburg, Germany) [29]. Both assays are based on microtiter
plates, pre-coated with a monoclonal antibody (capture antibody)
3.1.1 Quantitative
raised against the C terminus of either the Aβ40 or Aβ42 peptides.
Determination of Aβ40
The N terminus of the Aβ40 and Αβ42 peptides is recognized by a
and Aβ42
second monoclonal antibody (detection antibody), which is conju-
gated to a horseradish peroxidase enzyme that is sequentially
detected by the addition of the chromogenic substrate tetrameth-
ylbenzidine (TMB). The concentration of both Aβ40 and Αβ42 is
proportional to the obtained optical density.
1. Add 100 μL of each Standard, Control and diluted patient
sample into the respective wells of microtiter plate in duplicate.
Cover plate with lid, and incubate for 120 min at room tem-
perature (RT) (18–25 °C) on an orbital shaker (500 rpm).
2. Aspirate each well and wash all unbound antigen five times.
3. Add 100 μL of diluted enzyme conjugate in each well. Cover
plate with lid, and incubate for 60 min at RT (18–25 °C) on an
orbital shaker (500 rpm).
4. Aspirate each well and wash five times.
5. Pipette 100 μL of TMB substrate solution into each well.
Briefly mix contents by gently shaking the plate.
6. Incubate microtiter plate for 30 min at RT (18–25 °C), and
stop the substrate reaction by adding 100 μL of TMB stop
solution into each well.
7. Measure optical density with a photometer at 450 nm.
Established CSF Markers of Alzheimer’s Disease 97
3.2.1 Luminex Multi- The Luminex xMAP technology allows the measurement of mul-
Analyte Profiling (xMAP) tiple proteins simultaneously in a single sample. This technology
uses 5.6-μm microsphere beads, either polystyrene or paramag-
netic. The microsphere beads are internally dyed with precise
amounts of two or three spectrally distinct fluorescent dyes to pro-
duce up to 100–500 different sets of microspheres, depending on
the system design. Any molecule, such as proteins, antibodies,
ligands, or nucleic acids, interacting with the desired target, can be
coupled to the beads [32]. Initially, the coupled beads are incu-
bated with the sample and bind to the desire target. Subsequently
the bead-target complex is incubated with a mixture of biotinyl-
ated antibodies, which are specific against the target analyte, asso-
ciated with a fluorescent bead. A fluorescent dye (phycoerythrin,
PE) is pre-conjugated to streptavidin molecules and is used to indi-
cate the occurrence of positive binding and quantify the amount of
target analytes (Fig. 1). The multiplexing detection is performed
on a Luminex analyzer, where two lasers (a 635-nm, 10-mW red
diode laser and a 532-nm, 13-mW yttrium aluminum garnet laser)
are used to identify and quantify fluorescence from each individual
microsphere bead-target analyte-streptavidin complex. The ana-
lyzer, simultaneously, identifies the target analyte through the spe-
cific bead-fluorescence spectral address and determines its quantity
(PE-fluorescence), as the sample passes through the analyzer [33].
Innogenetics manufactures one such xMAP-based test, the
INNO-BIA AlzBio3, which determines the amount of pTau, tTau,
and Aβ42 proteins in CSF samples. According to the test, a mix of
the microsphere beads of a specific region number is captured
selectively by a first monoclonal antibody [AT270 for pTau, AT120
for tTau, 4D7A3 for Aβ42)], coupled covalently. A mix of the
microspheres is added in a specific volume to the filter plates. CSF
samples or standards are added on the filter plate, together with a
mix of biotinylated antibodies. Each biotinylated detector anti-
body detects one or several parameters [e.g., 3D6 for Aβ42, HT7
for detection of pTau and tTau]. The antigen-antibody complex is
Established CSF Markers of Alzheimer’s Disease 99
3.2.2 Meso Scale In the MSD assay main steps that are followed are highly similar
Discovery’s Multi-Array to ELISA assay. The plate is initially coated with a capture reagent
(MSD) before the addition of the samples and followed by the addition of
detection reagents. However, MSD platform uses a different type
of uncoated plates, which have a working electrode surface that
absorbs capture reagent within the wells. The plates, used for the
MSD assay, can be coated with a variety of capture molecules and
reagents, including antibodies, virus-like particles, cells, peptides,
and lysates. After coating, the sample and a solution containing
detection antibodies conjugated with electrochemiluminescent
labels are added to the wells. The analytes in the sample bind to
the capture reagent immobilized on the electrode surface, and this
complex recruits the detection antibodies forming a “sandwich.”
An MSD read buffer that provides the appropriate chemical envi-
ronment for electrochemiluminescence is loaded to the plate and
analyzed into an MSD imager. The imager applies a voltage to the
plate electrodes, causing the SULFO-TAG in close proximity to
the bottom of the plate to emit light through a series of reduction
and oxidation reaction. The imager measures the intensity of
emitted light to provide a quantitative measure of analytes in the
sample [34].
Based on the above, at least two possible MSD immunoassay
formats may be developed for amyloid peptides (Aβ40 and Αβ42)
and Tau protein (tTau and pTau) identification and quantification.
First, a typical format of an immunoassay that employs a specific
antibody as the capture reagent (capture antibody) that coats the
well of a plate can be developed. In a second format of the MSD
assay, instead of using a capture antibody to coat a well-plate posi-
tion, the sample (CSF sample) is directly coated in the plate well as
the capture material. Then specific antibodies are used to identify
and quantify the under-study analyte. According to recently pub-
lished data, based on correlation studies between all the above-
described immunoassays, the data obtained with the MSD platform
for AD CSF biomarkers are sparse at this time compared to the
ELISA and xMAP platforms [31].
3.4.2 Analytical The measured CSF biomarker concentrations differ between stud-
Challenges ies which is the consequence of a number of preanalytical, analyti-
cal, and/or assay-related factors. The source of overall variability
can be attributed to within-assay run variability (between duplicate
samples), intralaboratory longitudinal variability, between-
laboratory variability, and within- and between-assay kit lot vari-
ability [36]. The overall variability is reported to be around
20%–30%, with lower numbers for ELISA than for xMAP and
MSD measurements. Interlaboratory variability is a major contrib-
utor, while intralaboratory longitudinal variability has a smaller and
the within-run variability an even smaller effect. For some of the
analytes, a significant impact from between-lot-dependent variabil-
ity has been observed. Hence, it is important that quality of kits is
improved, in order to minimize lot-to-lot variation, being caused
by matrix effects, variation in production of different kit compo-
nents, etc. Furthermore, the development of detailed experimental
protocols is an essential step for decreasing variability [31, 35, 36].
Interestingly, the development of a novel fully automated electro-
chemiluminescence assay for the quantitation of CSF Aβ42 has
been recently reported [45]. It has a markedly improved precision
in comparison to other available assays and possibly opens the road
to overcoming the shortcoming of the high variability. In addition,
several, already underway quality control initiatives aim to contrib-
ute to standardization efforts (harmonizing laboratory practices,
defining procedures on CSF collection and handling, establishing
reference measurement procedures) [31, 35]. The development of
the first reference procedure for CSF Aβ42 was an additional
important step toward standardization [46].
The differences in mean concentrations of CSF markers
between the analytical techniques ELISA, xMAP, and MSD are a
major limitation for establishing multicentric cooperation, gold-
standard protocols, and reference values to be shared by distinct
laboratories. This difference is attributable to the lack of certified
reference materials (CRMs), which are also called standard refer-
ence materials, and calibrators for CSF biomarkers. Metrology
institutes develop CRMs. Establishing a protein CRM is a complex
task, since heterogeneities engendered by post-transnational modi-
fications or contaminations make it difficult to establish the purity
of proteins. The development of CRMs for CSF AD biomarkers
embodies a major challenge. In order to achieve it, the coordinated
efforts of researchers, industry, and metrology are necessary. If suc-
cessful, full global traceability and comparability of biomarker
results yielded by different analytical techniques and at different
laboratories will be enabled [31, 36].
Established CSF Markers of Alzheimer’s Disease 103
4 Notes
References
1. De-Paula VJ, Radanovic M, Diniz BS et al 7. Forlenza OV, Radanovic M, Talib LL et al
(2012) Alzheimer's disease. Subcell Biochem (2015) Cerebrospinal fluid biomarkers in
65:329–352. https://doi.org/10.1007/ Alzheimer's disease: diagnostic accuracy and
978-94-007-5416-4_14 prediction of dementia. Alzheimers Dement
2. Kocahan S, Dogan Z (2017) Mechanisms of (Amst) 1(4):455–463. https://doi.org/10.
Alzheimer's disease pathogenesis and preven- 1016/j.dadm.2015.09.003
tion: the brain, neural pathology, N-methyl-D- 8. Albert MS, DeKosky ST, Dickson D et al
aspartate receptors, tau protein and other risk (2011) The diagnosis of mild cognitive impair-
factors. Clin Psychopharmacol Neurosci ment due to Alzheimer's disease: recommen-
15(1):1–8. https://doi.org/10.9758/cpn. dations from the National Institute on
2017.15.1.1 Aging-Alzheimer's Association workgroups on
3. Alexopoulos P, Kurz A (2015) The new con- diagnostic guidelines for Alzheimer's disease.
ceptualization of Alzheimer's disease under the Alzheimers Dement 7(3):270–279. https://
microscope of influential definitions of disease. doi.org/10.1016/j.jalz.2011.03.008
Psychopathology 48(6):359–367. https:// 9. Perneczky R, Pohl C, Sorg C et al (2006)
doi.org/10.1159/000441327 Complex activities of daily living in mild cogni-
4. McKhann GM (2011) Changing concepts of tive impairment: conceptual and diagnostic
Alzheimer disease. JAMA 305(23):2458–2459. issues. Age Ageing 35(3):240–245. https://
https://doi.org/10.1001/jama.2011.810 doi.org/10.1093/ageing/afj054
5. McKhann G, Drachman D, Folstein M et al 10. Dubois B, Hampel H, Feldman HH et al
(1984) Clinical diagnosis of Alzheimer's disease: (2016) Preclinical Alzheimer's disease: defini-
report of the NINCDS-ADRDA work group tion, natural history, and diagnostic criteria.
under the auspices of Department of Health and Alzheimers Dement 12(3):292–323. https://
Human Services Task Force on Alzheimer's dis- doi.org/10.1016/j.jalz.2016.02.002
ease. Neurology 34(7):939–944 11. Jack CR, Knopman DS, Jagust WJ et al (2013)
6. McKhann GM, Knopman DS, Chertkow H Tracking pathophysiological processes in
et al (2011) The diagnosis of dementia due to Alzheimer's disease: an updated hypothetical
Alzheimer's disease: recommendations from model of dynamic biomarkers. The Lancet
the National Institute on Aging- Alzheimer's Neurology 12(2):207–216. https://doi.
Association workgroups on diagnostic guide- org/10.1016/S1474-4422(12)70291-0
lines for Alzheimer's disease. Alzheimers 12. Martins-de-Souza D (2010) Is the word 'bio-
Dement 7(3):263–269. https://doi. marker' being properly used by proteomics
org/10.1016/j.jalz.2011.03.005 research in neuroscience? Eur Arch Psychiatry
Established CSF Markers of Alzheimer’s Disease 105
for Biological Analysis in China. Asia Pac Biotech 43. Blennow K, Wallin A, Hager O (1993) Low
14(10):26–30 frequency of post-lumbar puncture headache
34. Meso Scale Discovery®. MSD sector and in demented patients. Acta Neurol Scand
quickplex plates, 18075-v2-2014 Jul. www. 88(3):221–223
mesoscale.com 44. Zetterberg H, Tullhog K, Hansson O et al
35. Mattsson N, Andreasson U, Persson S et al (2010) Low incidence of post-lumbar punc-
(2013) CSF biomarker variability in the ture headache in 1,089 consecutive memory
Alzheimer's Association quality control pro- clinic patients. Eur Neurol 63(6):326–330.
gram. Alzheimers Dement 9(3):251–261. https://doi.org/10.1159/000311703
https://doi.org/10.1016/j.jalz.2013.01.010 45. Bittner T, Zetterberg H, Teunissen CE et al
36. Mattsson N, Andreasson U, Persson S et al (2016) Technical performance of a novel, fully
(2011) The Alzheimer’s Association external automated electrochemiluminescence immu-
quality control program for cerebrospinal fluid noassay for the quantitation of beta-amyloid
biomarkers. Alzheimers Dement 7(4):386– (1-42) in human cerebrospinal fluid.
395.e6. https://doi.org/10.1016/j.jalz. Alzheimers Dement 12(5):517–526. https://
2011.05.2243 doi.org/10.1016/j.jalz.2015.09.009
37. Lam B, Masellis M, Freedman M et al (2013) 46. Mattsson N, Zegers I, Andreasson U et al
Clinical, imaging, and pathological heteroge- (2012) Reference measurement procedures for
neity of the Alzheimer's disease syndrome. Alzheimer's disease cerebrospinal fluid bio-
Alzheimers Res Ther 5(1):1. https://doi. markers: definitions and approaches with focus
org/10.1186/alzrt155 on amyloid beta42. Biomark Med 6(4):409–
38. Alexopoulos P, Roesler J, Thierjung N et al 417. https://doi.org/10.2217/bmm.12.39
(2015) Mapping CSF biomarker profiles onto 47. Vanderstichele H, Bibl M, Engelborghs S et al
NIA-AA guidelines for Alzheimer's disease. Eur (2012) Standardization of preanalytical aspects
Arch Psychiatry Clin Neurosci 266(7):587–597. of cerebrospinal fluid biomarker testing for
https://doi.org/10.1007/s00406-015-0628-7 Alzheimer's disease diagnosis: a consensus
39. Perneczky R, Alexopoulos P, Kurz A (2014) paper from the Alzheimer's biomarkers stan-
Soluble amyloid precursor proteins and secre- dardization initiative. Alzheimers Dement
tases as Alzheimer's disease biomarkers. Trends 8(1):65–73. https://doi.org/10.1016/j.jalz.
Mol Med 20(1):8–15 2011.07.004
40. Maier W (2016) Searching biomarkers for 48. Cicognola C, Chiasserini D, Eusebi P et al
mental disorders-lessons from Alzheimer’s dis- (2016) No diurnal variation of classical and
ease. Eur Arch Psychiatry Clin Neurosci candidate biomarkers of Alzheimer's disease in
266(7):583–585. https://doi.org/10.1007/ CSF. Mol Neurodegener 11(1):65. https://
s00406-016-0732-3 doi.org/10.1186/s13024-016-0130-3
41. Perneczky R, Guo L-H (2016) Plasma pro- 49. Zimmermann R, Lelental N, Ganslandt O et al
teomics biomarkers in Alzheimer’s disease: lat- (2011) Preanalytical sample handling and sam-
est advances and challenges. Methods Mol Biol ple stability testing for the neurochemical
1303:521–529. https://doi.org/10.1007/ dementia diagnostics. J Alzheimers Dis
978-1-4939-2627-5_32 25(4):739–745. https://doi.org/10.3233/
JAD-2011-110212
42. Duits FH, Martinez-Lage P, Paquet C et al
(2016) Performance and complications of lum- 50. Bjerke M, Portelius E, Minthon L et al (2010)
bar puncture in memory clinics: results of the Confounding factors influencing amyloid Beta
multicenter lumbar puncture feasibility study. concentration in cerebrospinal fluid. Int
Alzheimers Dement 12(2):154–163. https:// J Alzheimers Dis 2010. https://doi.
doi.org/10.1016/j.jalz.2015.08.003 org/10.4061/2010/986310
Chapter 7
Abstract
Central nervous system diseases are usually associated with significant modifications in brain morphometry
and function. These alterations may be subtle, in particular at early stages of the disease progress, and thus
not evident by visual inspection alone in magnetic resonance imaging. Group-level statistical comparisons
have dominated neuroimaging studies for many years, leading to better insight into the patterns of regional
vulnerability in brain neurodegenerative pathologies. However, such group-level results have no diagnostic
value at the individual level. Recently, pattern recognition approaches using multivariate analyses have led
to a fundamental shift in paradigm, aiming to predict the cognitive fate of each individual on the basis of
MRI-based algorithms of structural parameters. We review here the state-of-the-art fundamentals of pat-
tern recognition including feature selection, cross validation, and classification techniques and discuss limi-
tations including interindividual variation in normal brain anatomy and neurocognitive reserve. We
conclude with a special reference to future trends including multimodal pattern recognition and multi-
center approaches with data sharing and cloud computing.
Key words MRI, Pattern recognition, Support vector machines (SVMs), Multivariate, Predictive
modeling
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_7, © Springer Science+Business Media, LLC 2018
107
108 Sven Haller et al.
2 Methods
Fig. 1 Schematic illustration of the data processing pipeline. Several types of data input such as T1, diffusion-
weighted imaging, or fMRI are in a first step preprocessed depending on the specific demands of spatial nor-
malization into a standard space, including field inhomogeneity correction, spatial (and temporal) smoothing,
atlasing, independent component decomposition, structural or functional connectivity analyses, and others. In
the next step, the preprocessed data is used for feature selection and classification
2.2 Feature Selection In most cases, the number of features (e.g., voxels in the case of
structural MRI) is much larger than the number of individuals.
The selection of the best features is a long-standing problem in
machine learning that can be addressed either explicitly (by a sepa-
rate feature selection step) or implicitly (by regularization in the
110 Sven Haller et al.
2.3 Classification Support vector machines (SVMs) have been widely applied to neu-
roimaging data, mostly because of their robustness against outliers,
but they are by far not the only choice available [18]. Typically,
SVMs are used for binary classification such as discriminating
patients from healthy controls. Recently, more advanced classifiers
have been developed (e.g., decision forests [19]) and should be
considered for multi-class classification. However, there is some
trade-off between increasing the complexity of the classifier and
improving the feature extraction. Usually, good features allow for
better classification with almost any classifier.
Another issue with classifiers arises when features are multimodal;
i.e., combining different types of imaging data with other measures
such as scores from neuropsychological tests or clinical parameters.
As the model of a single classifier will not allow the necessary flexibil-
ity, “ensemble learning” is a rich field in machine learning that deals
with combining multiple classifiers (and thus multiple models) to
integrate the richness of the data structure. One promising approach
in the future is the use of multilevel classifier algorithms to aggregate
information in a hierarchical way (see Fig. 2). This approach makes it
possible to take into account previously established knowledge about
the relative weight of each classifier.
Finally it is also important to mention the recent breakthrough
in artificial intelligence applied to neuroimaging classification: deep
learning [20]. This technique has been successfully applied to mul-
tiple fields including visual recognition, mostly with full supervi-
sion. A typical deep-learning architecture for visual recognition
builds upon convolutional neural network. Given large-scale
training data and the power of high-performance computational
infrastructure, deep learning has achieved tremendous improve-
ment in visual recognition with thousands of categories [21].
Structural Imaging in AD 111
Fig. 2 Simplified processing pipeline for image classification. Processing pipeline for a single domain classification
(a) from data input to feature selection/classification to output. In multimodal classification (b–d), all input data can
directly enter one single feature selection/classification (b). Alternatively, it is possible to have one feature selection/
classification per input data, followed by one single super-feature selection/classification at the second level (c).
Moreover, it is possible to introduce an additional feature selection/classification levels, for example, regrouping all
imaging modalities, followed by a super-feature selection/classification at the third level (d)
2.4 Cross Validation A key step to consider in multimodal prediction is the distinction
between training and validation of each classifier. This later phase
concerns separate parts of the data according to a cross validation
methodology. As already mentioned, the model of classification is
built using part of the data where both the features and the pre-
dicted variable are given. Then, the classifier is evaluated on the
remaining part of the data by comparing its outcome against the
ground-truth prediction. The procedure is repeated after removing
different parts of the data to produce an average performance across
the validation folds (e.g., in terms of specificity and sensitivity).
Ideally, the classifier should predict the main data structure and
ignore the noise, and its model should be applicable to unseen data.
3.2 Incorporating There is a substantial body of evidences accumulated over the past
Prior Knowledge 30 years on the structure/function relationship in a wide range of
different diseases. This extensive prior knowledge is largely ignored
in recent individual level pattern recognition analyses, although it
might potentially improve classification accuracy and robustness.
Similar to the discussion about the anatomic structure of the
brain above, it is likely beneficial to inject any available domain
knowledge to improve the information content of the features
despite the fact that feature selection and state-of-the-art classifiers
are designed to deal with high-dimensional learning. From this
viewpoint, well-documented case-control studies with a priori
hypotheses are needed to guide and enrich the technical perfor-
mance of multilevel classifiers. Purely data-driven methods could
lead to biologically irrelevant observations in dementia prediction.
In addition, the interpretation of the results could often become
easier when features represent domain-relevant information.
Modern methods have recently been proposed to exploit spatial
structure; e.g., based on hierarchical clustering to regroup similar
voxels and reinforce the robustness [26]. For some applications,
features can also be extracted from specific regions of interest
instead of the whole brain, or the “locality” of information can be
probed by a “searchlight” approach where classification perfor-
mance is reported in an information map for features extracted
from local neighborhoods [27].
Structural Imaging in AD 113
3.3 The Cognitive Pattern recognition analyses at the individual level (as well as uni-
Reserve variate group-level analyses) are based on the assumption that
there is a direct relationship between brain pathology and symp-
tomatology. In the example of cognitive decline, the assumption is
that decreasing cognitive functions are paralleled by progressive
brain atrophy. This assumption is, however, only rarely true. Due
to sociodemographic factors such as education and social integra-
tion, some individuals are able to compensate clinically the struc-
tural MRI changes even for one or two decades. The example of
AD is illustrative in this respect. The appearance of various biologi-
cal changes (including hippocampal volume loss or tau-related cor-
tical thinning) suggestive of AD pathology in controls has been
extensively documented. They may precede clinically overt demen-
tia by many years, or even decades, thus defining a temporally wide
preclinical phase of the disease [28]. This interindividual variation
in the neurocognitive reserve was described already in 1968 [29].
In the domain of pattern recognition, the same amount of struc-
tural brain alterations could be associated with clinically overt cog-
nitive decline only if the neurocognitive reserve is exhausted. This
interindividual variability in the neurocognitive reserve represents
a fundamental limitation for pattern recognition approaches espe-
cially in a clinical setting.
3.4 Disbalance As already mentioned, a typical feature set extracted from MRI
Between Number data can easily contain more than 100,000 features, which clearly
of Features exceeds the number of individuals in single-center studies. In most
and Number cases, the features are related (similarity of adjacent or homologous
of Subjects: The Risk voxels), and only a limited number will carry discriminative
of Over-Fitting information.
While the cross validation methodology is essential to train and
evaluate classifiers, it still has the risk of over-fitting the data as the
parameters can be tuned. Consequently, several nested levels of
cross validation should be used, as well as separate independent
test datasets to guarantee the quality of the multivariate recogni-
tion patterns. Often the sample size for single-center studies is
insufficient to estimate such real-world performance. In addition,
parameter tuning for both feature selection and classifiers requires
an additional inner cross validation loop that decreases the amount
of available data for learning and increase the risk of over-fitting.
In summary, selecting and determining the importance of fea-
tures is an essential processing step in classification analyses, yet
identifying the related parameters such as optimal number of fea-
tures or regularization tuning is not a trivial practice. This is still an
active field of research in machine learning; e.g., stability selection,
which refers to the consistency of features when subsampling the
feature space, including applications to neuroimaging [30].
114 Sven Haller et al.
3.5 Variability Related Additional potentially confounding factors include scanner hetero-
to Patient Selection, geneity [31, 32], variability in data preprocessing, and patient
Inter-Scanner selection.
Variability, and Data
Preprocessing
4 Conclusions
4.1 Toward New The simple classification of healthy subjects versus clinically overt
Biomarkers AD or even MCI is of marginal importance for clinical practice.
Modern AD research focuses on the prediction of cognitive decline
both in MCI and preclinical AD cases. The identification of predic-
tive biomarkers in AD research is one of the more promised and
challenging domains in human neurobiology. Although there is a
wide agreement about the need to identify in situ AD at the pre-
clinical stages and if possible long before the emergence of MCI,
passing from group differences to individual prediction is quite
difficult in the light of the impressive interindividual variability of
most biomarkers in cognitively intact cases that imply the need for
including a substantial number of cases in research cohorts, the
uncertainty regarding the cognitive fate of these cases, the diffi-
culty to propose curative treatments (at the level of primary pre-
vention) in the absence of any alert sign.
The definitions of MCI have substantially evolved and changed
over the past years, which goes beyond the scope of this review.
Depending on the MCI subtype, only about half of MCI subjects
will progress to clinically overt dementia, whereas the other half
may remain stable or evolve to other forms of dementia [33–35].
Assuming that only about half of unselected MCI individuals will
progress to clinically overt dementia, the prediction of individuals
at risk for consecutive cognitive decline is of paramount impor-
tance for early individual treatment as well as for clinical trials. In a
typical placebo-controlled pharmaceutical trial, 25% of unselected
MCI will remain stable despite being in the placebo group, while
only 25% of individuals will progress and obtain the active com-
pound. Therefore, preselection of at-risk individuals for future cog-
nitive decline would substantially improve the design of clinical
trials. As MRI is routinely performed in the clinical workup of cog-
nitive decline, advanced data analysis techniques as those by pat-
tern recognition tools make use of already existing data, which is
thus cost effective and without additional discomfort for the
patient. It is possible to predict future cognitive decline in MCI
using baseline MRI based on gray matter voxel-based morphome-
try (VBM) [1, 36, 37], white matter DTI [38, 39], or iron deposi-
tion [38]. It is further possible to combine diffusion tensor imaging
(DTI) and resting-state fMRI to identify MCI individuals [40] and
to predict MCI to AD conversion using multimodal measures also
Structural Imaging in AD 115
References
1. Fan Y, Batmanghelich N, Clark CM et al via high-dimensional pattern classification of
(2008) Spatial patterns of brain atrophy in MRI. Neuroimage 41:1220–1227. https://
MCI patients, identified via high-dimensional doi.org/10.1016/j.neuroimage.2008.03.050
pattern classification, predict subsequent cog- 5. Xekardaki A, Rodriguez C, Montandon M-L
nitive decline. Neuroimage 39:1731–1743. et al (2015) Arterial spin labeling may contrib-
https://doi.org/10.1016/j. ute to the prediction of cognitive deterioration
neuroimage.2007.10.031 in healthy elderly individuals. Radiology
2. Fan Y, Resnick SM, Wu X, Davatzikos C 274:490–499. https://doi.org/10.1148/
(2008) Structural and functional biomarkers of radiol.14140680
prodromal Alzheimer’s disease: a high- 6. Wang L, Benzinger TL, Hassenstab J et al
dimensional pattern classification study. (2015) Spatially distinct atrophy is linked to
Neuroimage 41:277–285. https://doi. β-amyloid and tau in preclinical Alzheimer dis-
org/10.1016/j.neuroimage.2008.02.043 ease. Neurology 84:1254–1260. https://doi.
3. Klöppel S, Stonnington CM, Chu C et al (2008) org/10.1212/WNL.0000000000001401
Automatic classification of MR scans in 7. Seeley WW, Crawford RK, Zhou J et al (2009)
Alzheimer’s disease. Brain J Neurol 131:681– Neurodegenerative diseases target large-scale
689. https://doi.org/10.1093/brain/awm319 human brain networks. Neuron 62:42–52.
4. Davatzikos C, Resnick SM, Wu X et al (2008) https://doi.org/10.1016/j.neuron.2009.
Individual patient diagnosis of AD and FTD 03.024
116 Sven Haller et al.
8. Haxby JV, Gobbini MI, Furey ML et al (2001) 20. Wu J, Yu Y, Huang C, Yu K (2015) Deep multiple
Distributed and overlapping representations of instance learning for image classification and
faces and objects in ventral temporal cortex. auto-annotation. IEEE, pp 3460–3469
Science 293:2425–2430. https://doi. 21. van der Burgh HK, Schmidt R, Westeneng H-J
org/10.1126/science.1063736 et al (2017) Deep learning predictions of sur-
9. Krishnan A, Williams LJ, McIntosh AR, Abdi vival based on MRI in amyotrophic lateral
H (2011) Partial Least Squares (PLS) methods sclerosis. Neuroimage Clin 13:361–369.
for neuroimaging: a tutorial and review. https://doi.org/10.1016/j.nicl.2016.10.008
Neuroimage 56:455–475. https://doi. 22. Suk H-I, Shen D (2013) Deep learning-based
org/10.1016/j.neuroimage.2010.07.034 feature representation for AD/MCI classifica-
10. Cox DD, Savoy RL (2003) Functional mag- tion. Med Image Comput Comput Assist
netic resonance imaging (fMRI) “brain read- Interv 16:583–590
ing”: detecting and classifying distributed 23. Haug H (1987) Brain sizes, surfaces, and neu-
patterns of fMRI activity in human visual cor- ronal sizes of the cortex cerebri: a stereological
tex. Neuroimage 19:261–270. https://doi. investigation of man and his variability and a
org/10.1016/S1053-8119(03)00049-1 comparison with some mammals (primates,
11. Pereira F, Mitchell T, Botvinick M (2009) whales, marsupials, insectivores, and one ele-
Machine learning classifiers and fMRI: a tuto- phant). Am J Anat 180:126–142. https://doi.
rial overview. Neuroimage 45:S199–S209. org/10.1002/aja.1001800203
https://doi.org/10.1016/j. 24. Haller S, Borgwardt SJ, Schindler C et al
neuroimage.2008.11.007 (2009) Can cortical thickness asymmetry anal-
12. Mitchell TM, Shinkareva SV, Carlson A et al ysis contribute to detection of at-risk mental
(2008) Predicting human brain activity associ- state and first-episode psychosis? A pilot study.
ated with the meanings of nouns. Science Radiology 250:212–221. https://doi.
320:1191–1195. https://doi.org/10.1126/ org/10.1148/radiol.2501072153
science.1152876 25. Hackmack K, Weygandt M, Wuerfel J et al
13. Ethofer T, Van De Ville D, Scherer K, (2012) Can we overcome the “clinico-
Vuilleumier P (2009) Decoding of emotional radiological paradox” in multiple sclerosis?
information in voice-sensitive cortices. Curr J Neurol 259:2151–2160. https://doi.
Biol 19:1028–1033. https://doi. org/10.1007/s00415-012-6475-9
org/10.1016/j.cub.2009.04.054 26. Cui Y, Sachdev PS, Lipnicki DM et al (2012)
14. Nishimoto S, Vu AT, Naselaris T et al (2011) Predicting the development of mild cognitive
Reconstructing visual experiences from brain impairment: a new use of pattern recognition.
activity evoked by natural movies. Curr Biol Neuroimage 60:894–901. https://doi.
21:1641–1646. https://doi.org/10.1016/j. org/10.1016/j.neuroimage.2012.01.084
cub.2011.08.031 27. Kriegeskorte N, Goebel R, Bandettini P (2006)
15. Horikawa T, Tamaki M, Miyawaki Y, Kamitani Information-based functional brain mapping.
Y (2013) Neural decoding of visual imagery Proc Natl Acad Sci U S A 103:3863–3868.
during sleep. Science 340:639–642. https:// https://doi.org/10.1073/pnas.0600244103
doi.org/10.1126/science.1234330 28. Lazarczyk MJ, Hof PR, Bouras C,
16. Tolosi L, Lengauer T (2011) Classification Giannakopoulos P (2012) Preclinical
with correlated features: unreliability of feature Alzheimer disease: identification of cases at risk
ranking and solutions. Bioinformatics among cognitively intact older individuals.
27:1986–1994. https://doi.org/10.1093/ BMC Med 10:127. https://doi.org/10.
bioinformatics/btr300 1186/1741-7015-10-127
17. Van De Ville D, Blu T, Unser M (2006) Surfing 29. Tomlinson BE, Blessed G, Roth M (1968)
the brain. IEEE Eng Med Biol Mag Q Mag Observations on the brains of non-demented
Eng Med Biol Soc 25:65–78 old people. J Neurol Sci 7:331–356
18. Cristianini N, Shawe-Taylor J (2000) An intro- 30. Langs G, Menze BH, Lashkari D, Golland P
duction to support vector machines: and other (2011) Detecting stable distributed patterns of
kernel-based learning methods. Cambridge brain activation using Gini contrast.
University Press, New York Neuroimage 56:497–507. https://doi.
19. Criminisi A, Shotton J (2013) Decision forests org/10.1016/j.neuroimage.2010.07.074
for computer vision and medical image analysis. 31. Kruggel F, Turner J, Muftuler LT, Alzheimer’s
Springer Publishing Company, Incorporated, Disease Neuroimaging Initiative (2010)
Berlin Impact of scanner hardware and imaging pro-
tocol on image quality and compartment
Structural Imaging in AD 117
volume precision in the ADNI cohort. and mild cognitive impairment. Neuroimage
Neuroimage 49:2123–2133. https://doi. 55:856–867. https://doi.org/10.1016/j.
org/10.1016/j.neuroimage.2009.11.006 neuroimage.2011.01.008
32. Abdulkadir A, Mortamet B, Vemuri P et al 43. Zhang D, Shen D, Alzheimer’s Disease
(2011) Effects of hardware heterogeneity on Neuroimaging Initiative (2012) Multi-modal
the performance of SVM Alzheimer’s disease multi-task learning for joint prediction of mul-
classifier. Neuroimage 58:785–792. https:// tiple regression and classification variables in
doi.org/10.1016/j.neuroimage.2011.06.029 Alzheimer’s disease. Neuroimage 59:895–907.
33. Petersen RC (2004) Mild cognitive impair- https://doi.org/10.1016/j.
ment as a diagnostic entity. J Intern Med neuroimage.2011.09.069
256:183–194. https://doi. 4 4. Zhang D, Shen D, Initiative ADN (2012)
org/10.1111/j.1365-2796.2004.01388.x Predicting future clinical changes of MCI
34. Mariani E, Monastero R, Mecocci P (2007) patients using longitudinal and multi-
Mild cognitive impairment: a systematic review. modal biomarkers. PLoS One 7:e33182.
J Alzheimers Dis 12:23–35 https://doi.org/10.1371/journal.pone.
35. Forlenza OV, Diniz BS, Nunes PV et al (2009) 0033182
Diagnostic transitions in mild cognitive impair- 45. Haller S, Missonnier P, Herrmann FR et al
ment subtypes. Int Psychogeriatr 21:1088– (2013) Individual classification of mild cogni-
1095. https://doi.org/10.1017/S10416102 tive impairment subtypes by support vector
09990792 machine analysis of white matter DTI. AJNR
36. Misra C, Fan Y, Davatzikos C (2009) Baseline Am J Neuroradiol 34:283–291. https://doi.
and longitudinal patterns of brain atrophy in org/10.3174/ajnr.A3223
MCI patients, and their use in prediction of 46. Haller S, Barkhof F (2017) Interaction of vas-
short-term conversion to AD: results from cular damage and Alzheimer dementia: focal
ADNI. Neuroimage 44:1415–1422. https:// damage and disconnection. Radiology
doi.org/10.1016/j.neuroimage.2008.10.031 282:311–313. https://doi.org/10.1148/
37. Plant C, Teipel SJ, Oswald A et al (2010) radiol.2016161564
Automated detection of brain atrophy patterns 47. Saito S, Ihara M (2016) Interaction between
based on MRI for the prediction of Alzheimer’s cerebrovascular disease and Alzheimer pathol-
disease. Neuroimage 50:162–174. https:// ogy. Curr Opin Psychiatry 29:168–173.
doi.org/10.1016/j.neuroimage.2009.11.046 https://doi.org/10.1097/YCO.00000000
38. Haller S, Bartsch A, Nguyen D et al (2010) 00000239
Cerebral microhemorrhage and iron deposi- 48. Kapasi A, Schneider JA (2016) Vascular contri-
tion in mild cognitive impairment: butions to cognitive impairment, clinical
susceptibility-
weighted MR imaging assess- Alzheimer’s disease, and dementia in older per-
ment. Radiology 257:764–773. https://doi. sons. Biochim Biophys Acta 1862:878–886.
org/10.1148/radiol.10100612 https://doi.org/10.1016/j.bbadis.2015.
39. O’Dwyer L, Lamberton F, Bokde ALW et al 12.023
(2012) Using support vector machines with 49. Madigan JB, Wilcock DM, Hainsworth AH
multiple indices of diffusion for automated (2016) Vascular contributions to cognitive
classification of mild cognitive impairment. impairment and dementia: topical review of
PLoS One 7:e32441. https://doi. animal models. Stroke 47:1953–1959.
org/10.1371/journal.pone.0032441 https://doi.org/10.1161/STROKEAHA.
40. Wee C-Y, Yap P-T, Zhang D et al (2012) 116.012066
Identification of MCI individuals using struc- 50. Brickman AM (2013) Contemplating
tural and functional connectivity networks. Alzheimer’s disease and the contribution of
Neuroimage 59:2045–2056. https://doi. white matter hyperintensities. Curr Neurol
org/10.1016/j.neuroimage.2011.10.015 Neurosci Rep 13:415. https://doi.
41. Cui Y, Liu B, Luo S et al (2011) Identification org/10.1007/s11910-013-0415-7
of conversion from mild cognitive impairment 51. Mueller SG, Weiner MW, Thal LJ et al (2005)
to Alzheimer’s disease using multivariate pre- Ways toward an early diagnosis in Alzheimer’s
dictors. PLoS One 6:e21896. https://doi. disease: the Alzheimer’s Disease Neuroimaging
org/10.1371/journal.pone.0021896 Initiative (ADNI). Alzheimers Dement 1:55–
42. Zhang D, Wang Y, Zhou L et al (2011) 66. https://doi.org/10.1016/j.
Multimodal classification of Alzheimer’s disease jalz.2005.06.003
Chapter 8
Abstract
In this chapter, we summarize the most important methods and techniques of functional brain imaging
(FBI) in the context of biomarkers in preclinical Alzheimer’s disease (pAD). We analyze putative models of
pAD, covering both genetic and clinical aspects of pAD. Studies on cognitively healthy individuals carrying
the APOE-ε4 allele and patients with mild cognitive impairment have been included. To address method-
ologies, studies on metabolism and cerebral perfusion are analyzed. Although perfusion is not part of the
criteria for pAD, there is increasing evidence that perfusion imaging techniques, including PET/SPECT
methods, and magnetic resonance imaging (MRI) techniques, such as functional MRI (fMRI) and arterial
spin labeling (ASL), can be useful for elucidating the underlying pathophysiological changes in pAD.
However, metabolic imaging with glucose PET still remains the most established FBI biomarker of pAD.
1 Introduction
1.1 Definition Actually, there is only sparse knowledge about changes occurring
and Potential Models in the brain during the years preceding the cognitive and func-
of Preclinical AD tional impairment associated with Alzheimer’s disease (AD).
Normal cognitive status in the elderly (i.e., incidental forgetfulness
of aging) can be described as aging-associated cognitive decline
without relevant memory complaints.
In 2011, the National Institute on Aging-Alzheimer’s
Association (NIA-AA) published criteria to stage preclinical
Alzheimer’s disease (pAD), mainly based on the amyloid cascade
hypothesis of AD. These criteria [1] define three consecutive stages:
●● Stage 1: Asymptomatic cerebral amyloidosis
●● Stage 2: Cerebral amyloid deposition + downstream neurode-
generation with pathological CSF/PET or MRI markers
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_8, © Springer Science+Business Media, LLC 2018
119
120 Peter Häussermann et al.
(tau/phospho-tau; 18
F-FDG-PET, atrophy as visualized by
structural MRI)
●● Stage 3: Cerebral amyloidosis + neurodegeneration + subtle
cognitive or behavioral decline, not meeting the criteria for
MCI
Clinically, elderly patients reporting cognitive problems have
recently been classified as suffering from subjective cognitive
decline (SCD). SCD is defined as a subjectively experienced persis-
tent continuous cognitive decline, or decline in cognitive capacity,
compared with a previously normal state, not related to an acute
event. Performance on standard tests used to define mild cognitive
impairment (MCI) is normal. Patients with SCD expressing con-
cern about these deficits have a threefold increased risk of later
developing dementia and a sixfold increased risk of later develop-
ing AD. In models of the very early stages of AD, SCD can be
considered to occur in late stages of pAD, characterized by increas-
ing compensatory cognitive efforts and subtle cognitive changes
before the threshold of MCI or prodromal AD is reached [2].
There are several human models of pAD. The clinically most
important human model is the mild cognitive impairment (MCI)
stage of cognitive decline in the elderly. In MCI, patients are par-
tially symptomatic with substantiated cognitive impairment that
does not significantly interfere with normal daily functioning. The
amnestic MCI subgroup (aMCI) has the highest risk to develop
AD in the future [3]. The annual conversion rate from MCI to
dementia is approximately between 5–25%, depending on MCI
criteria and subtype [3, 4]. MCI can, therefore, be considered as a
transitional period between normal aging and dementia.
Another approach to better understand presymptomatic AD is
to study cognitively healthy individuals with known genetic risk
factors. This includes nondemented individuals who are at risk of
later developing AD, such as subjects with recognized genetic
traits. Individuals carrying the ε4 allele of the apolipoprotein E
(APOE-ε4) gene have an increased risk of later developing AD. The
APOE gene is thought to play a role in cerebrovascular lesions in
dementia [5] and is considered to be the single most important
genetic factor in late onset AD [6]. Carriage of the APOE-ε4 allele
significantly increases AD risk in a gene-dose-dependent manner
and lowers the age of AD onset [7, 8].
2.1.3 Functional MRI Functional magnetic resonance imaging (fMRI) is a FBI method
(fMRI) that uses the phenomenon of nuclear magnetic resonance to indi-
rectly measure and map brain function (not anatomy) by detecting
cerebral blood flow (CBF) changes following alterations in neural
activity. fMRI is based on the so-called blood-oxygen-level-
dependent (BOLD) contrast. This method quantifies circum-
scribed changes in deoxyhemoglobin level, which exhibits an
interwoven dependency on CBF, cerebral metabolic rate of O2, as
well as on cerebral blood volume [22]. To understand the fMRI
signal, two effects need to be considered. First, there is a difference
in magnetic properties between oxygen-rich arterial (oxygenated)
and oxygen-poor venous (deoxygenated) blood. Second, neural
activity paradoxically leads to a much greater increase in CBF than
oxygen consumption, which in turn leads to the blood being more
oxygenated with increasing neural activity. An increase in neural
activity in certain brain areas thereby leads to a small but detectable
change in the magnetic resonance signal. The fMRI signal, there-
fore, depends on physiological changes associated with CBF and
oxygen metabolism [22].
fMRI is widely available and relatively cheap and does not use
ionizing radiation, being able to localize cerebral activity in a milli-
meter range. fMRI can be combined with electrophysiological tech-
niques such as EEG and transcranial magnetic stimulation [22].
fMRI acquisition is usually coupled to a carefully designed
experiment, carried out by the subject during imaging. Experimental
paradigms trigger an activation of functional brain networks or
regions coupled. Limitations concern the reliance on cooperation
of the patient; children and dementia patients often cannot per-
form complex fMRI tasks. These experiments are time-consuming
and require a complex experimental setup with MRI-compatible
equipment. In contrast to the very controlled task-based fMRI
experiments, the so-called resting-state fMRI (rs-fMRI) is much
easier to perform and independent from a specific task. By analyz-
ing correlations in the BOLD signal over time between different
regions or voxels, rs-fMRI tries to assess the functional connectivity
(fc) patterns of the brain during rest. The term functional connec-
tivity is used to describe the specific connectivity between brain
areas sharing functional properties (i.e., the temporal correlation
between spatially remote neurophysiological events in distributed
neuronal networks). Slow fluctuations (<0.08 Hz) of the BOLD
signal with a high degree of temporal correlation between func-
tionally connected brain regions during rest and intrinsic process-
ing represent the neuronal baseline activity. These fluctuations
correspond to inter- and intraindividually stable resting-state net-
works (RSNs). Resting-state fc analyses measure task-independent
changes of brain function, thereby providing important insights
into brain plasticity [23].
124 Peter Häussermann et al.
2.1.4 Arterial Spin Arterial spin labeling (ASL) is a noninvasive MRI technique, mag-
Labeling: MRI netically labeling arterial water and using it as an endogenous tracer
to measure CBF changes associated with neuronal activity [10, 29].
The BOLD signal, which is the usual basis for the fMRI signal, is
linked to a signal change between two or more conditions. In
fMRI, there is no information about the absolute level of blood
flow. In contrast to this, the ASL technique has the ability to quan-
tify the absolute level of CBF by subtracting the signal of magneti-
cally tagged and untagged arterial blood. Thereby, the static signal
from all hydrogen nuclei subtracts out, ideally leaving only the sig-
nal arising from arterial blood flow [29, 30]. The ASL signal is
therefore independent from changes in neuronal activity. However,
the disadvantage is an increase in acquisition time, since the ASL
technique requires the acquisition of two images (one with the
labeled and one with non-labeled blood). ASL MRI has the poten-
tial to precisely estimate both extent and site of neural function
[29]. Changes in CBF are more localized to the brain parenchyma
(capillary bed), whereas the BOLD signal is more localized to
Functional Imaging in AD 125
2.2 Imaging Besson et al. [32] characterized a group of cognitively healthy sub-
of Metabolism jects by structural MRI, 18F-FDG-PET, and amyloid PET (florbeta-
in Preclinical AD pir) and dichotomized these individuals into positive or negative for
each neuroimaging marker. The 18F-FDG-positive group showed
2.2.1 F-FDG-PET
18
an AD typical hypometabolic pattern. Furthermore, in the “neuro-
degeneration” group with both structural MRI and FDG-PET
AD-like changes, the authors found a trend for an inverse relation-
ship with Aß deposition (i.e., subjects with neurodegeneration
exhibited less Aß and vice versa). Therefore, Besson et al. suggested
to combine structural MRI and amyloid/glucose PET biomarkers.
These methods offer unique insight into the time course of Aß
deposition and neurodegeneration in pAD. Furthermore, neurode-
generative changes seen in this study were largely independent from
cerebral Aß deposition.
Teipel et al. [33] analyzed hippocampal and posterior cingu-
late cortex (PCC) volume, PCC glucose metabolism, and Aß load
in a cohort of 667 subjects participating in the Alzheimer’s Disease
Neuroimaging Initiative (ADNI). Controls and early MCI patients
exhibited hypometabolism in PCC which was associated with hip-
pocampal atrophy. In late stages of MCI, hypometabolism in PCC
was associated both with PCC and hippocampal atrophy as well as
PCC Aß deposition. In patients suffering from AD, PCC hypome-
tabolism was associated with PCC atrophy.
In another study, Ewers et al. [34] showed that temporopari-
etal and prefrontal hypometabolism as well as decline in executive
function predicted conversion of cognitively healthy elderly to
MCI or dementia during a follow-up period of 3–4 years. Best
predicting results for conversion to MCI/dementia were obtained
when combining executive function assessment with rCMRglc in
the above-mentioned areas [34]. The same group examined the
effects of cognitive reserve capacity (CRC) on glucose metabolism
in cognitively healthy individuals with (Aß+) and without (Aß−)
CSF biomarkers of preclinical AD [35]. When using a ROI
approach, higher CRC was associated with lower glucose metabo-
lism in the PCC and angular gyrus in the Aß+ group, while higher
CRC was associated with PCC hypermetabolism in the Aß− group.
CRC may, therefore, play an important compensatory role in sus-
taining cognition in the presence of cerebral Aß deposition and
altered glucose metabolism.
Another study [36] analyzed the longitudinal changes of Aß
deposition, glucose metabolism, and medial temporal lobe (MTL)
atrophy in three different cohorts (pAD, MCI and dementia).
Patients with initial pAD stages II and III showed greater MTL
126 Peter Häussermann et al.
2.2.2 18F-FDG-PET Seo et al. [40] analyzed APOE-ɛ4, rCMRglc, and cerebral amyloid
in APOE-ɛ4 Carriers load (18F-florbetapir PET) in cognitively healthy controls, MCI
patients, and AD patients. In controls, carriage of the APOE-ɛ4
allele was associated with hypometabolism in the bilateral frontal,
temporal, and left parietal regions. In MCI, the same comparison
showed hypometabolism in the bilateral posterior parietal, temporal,
and left frontal regions. In AD, APOE-ɛ4 carriers exhibited hypo-
metabolism in the left hippocampus, right insular, and right tempo-
ral gyrus. After adjustment for Aß, the significant differences within
temporal areas disappeared in controls and MCI, whereas in AD, all
significant metabolic differences disappeared. The authors deducted
that Aß-independent APOE-ɛ4 influence on glucose metabolism is
restricted to frontal and parietal brain regions, mainly in the early
stages of cognitive decline. The same group [41] also showed that
temporoparietal hypometabolism disappears when controlling for
Aß in APOE-ɛ4 carriers without cognitive symptoms as compared to
healthy noncarriers [41], while medial frontal and anterior temporal
hypermetabolism seen in the abovementioned comparison persists
Functional Imaging in AD 127
2.2.3 F-FDG-PET
18
MCI is clinically the most important model of pAD. The amnestic
in MCI subtype has the highest risk of conversion to AD. The metabolic
pattern of aMCI resembles that seen in early stages of AD.
128 Peter Häussermann et al.
Fig. 1 Visualization of the statistical maps used to calculate the expression values corresponding to the group
t-contrasts. (Panel a) AD versus CON. (Panel b) PD versus CON. Statistical maps were shown with a threshold
of punc < 0.001 and overlaid onto the single-subject MR T1 template as delivered with the SPM software.
2.3 Imaging Perfusion describes the process by which oxygenated arterial blood
of Perfusion is provided to the capillary bed in a predefined mass of brain tissue.
in Preclinical AD It is measured in milliliters of blood per 100 g of tissue per min
[22]. CBF values in human being approach 50 mL/100 g per min,
and gray matter CBF is approximately three times higher than
white matter CBF [22]. Regional CBF can be assessed using PET/
SPECT methods, the more ancient 133Xe inhalation technique, and
MRI-based techniques such as fMRI and ASL.
In normal aging, cognitively healthy adults exhibit grey matter
perfusion reductions of about 0.45% per year [31]. Hypoperfusion
in aging tends to be widespread, affecting frontal, medial temporal,
parietal, and subcortical regions [13, 31, 65, 66]. In the stage of
clinically manifest dementia, perfusion in patients suffering from
AD correlates negatively with disease severity [67]. A p
henotypically
normal cognitive status in the elderly despite perfusion deficits may
reflect cognitive reserve capacity (CRC) of the aging brain [68].
As neither oxygen nor glucose can be stored within the brain,
neuronal energy supply strongly depends on a constant delivery of
oxygen and glucose by CBF. Therefore, there is a strong link
between neural activity, energy metabolism, and CBF, which rep-
resents the rationale of FBI methods using perfusion, not only in
pAD [10]. Several factors mediate the relationship between CBF
and cognition, including CRC, age, sex, genotype, and vascular
risk factors.
Functional Imaging in AD 131
2.3.1 PET/SPECT To date, there are no recent studies applying SPECT or PET for
Perfusion Imaging perfusion imaging in pAD, as defined by Sperling et al. [1]. However,
one study [69] investigated the relation between rCBF (as measured
with 99mTc-exametazime-SPECT) and CSF biomarkers in cogni-
tively healthy elderly. The authors found that high CSF p-tau and
total tau levels are correlated with decreased rCBF in the right medial
frontal lobe and that high CSF p-tau levels are correlated with
increased rCBF in the left frontotemporal border zone. There were
no significant associations between rCBF and CSF Aß1–42 levels.
These results indicate a correlation between tau pathology rather
than Aß deposition and CBF abnormalities in pAD.
Another study from the Baltimore Longitudinal Study of
Aging using repeated resting-state 15O-H2O PET scans found
rCBF differences between cognitively normal elderly with high and
low cerebral amyloid load [70]. Greater longitudinal reductions of
CBF in the group exhibiting high amyloid values were found in the
anterior as well as middle cingulate cortex, left thalamus, right
supramarginal gyrus, and midbrain. Greater longitudinal gains in
rCBF in the group exhibiting high amyloid values were encoun-
tered in left medial and inferior frontal gyri, left inferior parietal
gyrus, right precuneus, and left postcentral gyrus.
2.3.2 fMRI, Resting-State Edelman et al. [71] combined fMRI and Pittsburgh compound
fMRI and Functional B-PET (PIB-PET) in a face-name memory task. In their study,
Connectivity Aß-positive healthy elderly displayed an increased (possibly com-
in Preclinical AD pensatory) hippocampal activation during the task. In another
study, Gordon et al. [72] combined fMRI and CSF, respectively,
PET biomarkers of AD in two attentional control tasks to examine
changes related to Aß and tau pathology in cognitively healthy
subjects. They found that increased levels of tau and p-tau are
linked to overactivations of attentional control areas, indicating
that alterations of attentional networks are associated with AD
neuropathology.
Sheline et al. [73] applied rs-fMRI and PIB-PET in AD patients
and two groups of cognitively healthy individuals, one with evi-
dence of brain Aß (Aß+) and one without Aß (Aß−). The Aß+
group as well as the AD group showed significant differences in
functional connectivity (fc) within the DMN. The authors con-
cluded that brain Aß deposition leads to alterations in fc of the
DMN in pAD, in the same anatomical regions and in the same
direction as seen in AD.
Schultz et al. [74] examined fc in several cortical association
networks, including the DMN, in cognitively healthy individuals.
132 Peter Häussermann et al.
2.3.3 Arterial Spin As described above, reductions of CBF mirror the pathophysiologi-
Labeling MRI cal changes seen in pAD. Hays et al. [10] summarized ASL MRI
in Preclinical AD studies in several models of pAD. In the early preclinical stage, there
is evidence of hyperperfusion, whereas in later preclinical stages,
there exist both hyper- and hypoperfusion. Later, in manifest AD,
there is evidence for widespread reductions of CBF. Cerebral amy-
loid angiopathy may be an important contributing factor.
A recent study [14] found continuous CBF decreases in pAD,
mainly in temporal and parietal regions, along the stages as defined
by Sperling et al. [1]. Other studies have found CBF decreases
related to Aß deposition in the inferior temporal and parietal
cortex, In control subjects, high Aß levels were associated with
greater CBF reductions [70, 79].
2.4 APOE-ɛ4 Carriers Generally, there have been few PET/SPECT perfusion studies
focusing on cognitively normal APOE-ɛ4 carriers. Thambisetty
2.4.1 PET/SPECT
et al. [80] studied longitudinal CBF differences between two
Perfusion Imaging
groups of nondemented elderly from the Baltimore Longitudinal
Study of Aging using 15O-PET. One group contained APOE-ɛ4
carriers and the other contained noncarriers. APOE-ɛ4 carriers
exhibited widespread and greater regional CBF decline in frontal,
temporal, and parietal cortices as compared to noncarriers.
In patients with manifest AD, there was a dose-dependent
effect of the APOE-ɛ4 allele on rCBF, with more hypoperfusion
seen in frontal, temporoparietal, MTL, and occipital regions in the
APOE-ɛ4 carrier group. AD patients homozygous for the ɛ4 allele
also exhibit more severe MTL atrophy than noncarriers or AD
patients heterozygous for the ɛ4 allele [19, 81–83]. Furthermore,
there is a more rapid decline of CBF in the APOE-ɛ4 group com-
pared to noncarriers [19].
MCI patients carrying the APOE-ɛ4 allele have a greater
chance to convert to AD. The converters show hypoperfusion in
the MTL and parietal regions [84] as well as within the postcentral
region, as compared to nonconverters [85].
2.4.2 fMRI, Resting-State Fleisher et al. [86] compared resting CBF (DMN signal differ-
fMRI, and Functional ences) and fMRI BOLD response during an associative memory-
Connectivity encoding task in APOE-ɛ4 carriers with a positive familial history
of AD versus healthy controls without these risk factors. BOLD
activations during encoding did not differ between both groups.
Deactivations during encoding were more pronounced in the low
risk group within the parietal cortex. However, resting state DMN
analysis better discriminated both groups than encoding associated
fMRI. The authors conclude that resting state imaging is a more
effective and promising tool in assessing AD risk as compared to
activation fMRI.
Harrison et al. [87] analyzed fc of the hippocampus in cogni-
tively normal elderly, subdivided into two groups, one consisting
134 Peter Häussermann et al.
2.4.3 Arterial Spin Michels et al. [89] assessed APOE-ε4 genotype, Aß load, and CBF
Labeling MRI in cognitively healthy (CH) individuals and aMCI patients. The
global CBF was lower in the group of APOE-ε4-positive subjects
as compared to noncarriers. In aMCI, the global CBF was lower
compared to CH, and participants with increased Aß showed a
trend for lower global CBF. Therefore, the authors concluded that
the APOE-ε4 allele has an impact on CBF, which is at least par-
tially independent from Aß.
Bangen et al. [90] used ASL MRI and fMRI to assess resting
CBF as well as task-dependent CBF and BOLD response, in a
memory-encoding paradigm in cognitively healthy elderly (with
and without the APOE-ε4 allele) and in MCI patients (also with
and without the APOE-ε4 allele). In cognitively healthy APOE-ε4
carriers, there is evidence of hyperperfusion with increased resting-
state CBF in the MTL compared to ε4 noncarriers, while MCI
patients have decreased resting-state CBF within MTL structures
relative to controls. There were no significant group differences for
task-dependent BOLD response (MCI versus controls). These
findings suggest that alterations in resting-state CBF and CBF
response to memory encoding may serve as early markers of brain
dysfunction in different models of pAD.
Functional Imaging in AD 135
2.5 MCI Using serial 15O-water PET imaging data from the Baltimore
Longitudinal Study of Aging, one study [93] found alterations of
2.5.1 PET/SPECT rCBF in elderly individuals later converting to MCI. Compared to
Perfusion Imaging cognitively stable elderly, MCI converters displayed greater longi-
tudinal rCBF decreases in temporal, parietal, and thalamic regions
and rCBF increases in orbitofrontal, MTL, and ACC regions.
These rCBF changes were seen within brain regions that show
early accumulation of pathology of AD, suggesting a link between
early neuropathological changes and rCBF alterations.
2.5.2 fMRI, Resting-State Adriaanse et al. [94] analyzed fc using rs-fMRI in the DMN in
fMRI, and Functional cognitively healthy elderly and patients with MCI and AD. Amyloid
Connectivity deposition was assessed with PIB-PET. The authors found no asso-
ciation between fc of the DMN and Aß load in DMN areas in any
of the groups or between groups. The same group described
decreased fc within the DMN in AD in the precuneus and PCC
compared to controls. MCI patients exhibited fc changes that were
situated between patients suffering from AD and controls [95].
Liang et al. [96] examined fc in MCI patients in three different
areas of the inferior parietal cortex, notably the intraparietal sulcus
(IPS), the angular gyrus (AG), and the supramarginal gyrus (SG).
136 Peter Häussermann et al.
2.5.3 Arterial Spin Perfusion changes seen in MCI are suggestive of neurodegeneration-
Labeling MRI induced CBF dysregulation, which may result from altered metabolic
requests to preserve cognitive function [97]. Mild cognitive impair-
ment is linked to increases and also decreases in resting-state CBF.
Kim et al. [92] used ASL MRI to study CBF in patients suffer-
ing from AD and MCI as well as in cognitively normal controls.
Each group was further divided into carriers or noncarriers of the
APOE-ɛ4 allele. In the MCI APOE-ɛ4 carriers, hyperperfusion
was detected in the right parahippocampal gyrus, bilateral cingu-
late gyri, and right PCC as compared to noncarriers.
Michels et al. [89] assessed Aß, CBF, and APOE genotype in
cognitively healthy individuals and aMCI patients. They found
widespread CBF reductions, largely independent from Aß load, in
elderly APOE-ε4 carriers. Global CBF was lower in patients with
aMCI compared with cognitively healthy elderly. Interestingly, car-
riage of the APOE-ε4 allele had a more deleterious effect on both
local and global CBF than cerebral Aß deposition.
However, another study found that cerebral amyloid pathol-
ogy affected CBF across the span from cognitively healthy elderly
(with cerebral amyloidosis) to clinically manifest AD. In their
study, the authors [79] showed Aß pathology to be associated with
hypoperfusion in pAD, whereas in later stages of neurodegenera-
tion, atrophy becomes more prominent.
Other studies found that CBF increases within the temporal
lobe, ACC, insula, putamen, left hippocampus, right amygdala,
and basal ganglia. Decreases of CBF have been described in frontal
and temporoparietal areas (extending to the MTL), PCC, precu-
neus, and left middle occipital lobe [90, 98–101].
The PCC seems to play an important role as indicator of cog-
nitive decline, even in cognitively normal elderly [68]. This brain
region, known to be involved in early AD changes, exhibits
Functional Imaging in AD 137
3 Discussion
The rationale for defining pAD is that the best time for preventing
neurodegeneration in elderly subjects is early in the predementia
stage. Innovative pharmacological and non-pharmacological pre-
ventive interventions can best be assessed in pAD [1, 103]. Hence,
there is a need for personalized, biomarker-based approaches to
identify future dementia patients.
In recent years, functional brain imaging (FBI) has increas-
ingly been recognized as having the potential to represent such
biomarkers of pAD. Table 1 summarizes the most important FBI
findings in pAD, MCI, and nondemented APOE-ε4 allele carriers.
Metabolic imaging (i.e., 18F-FDG-PET) has significantly con-
tributed to our understanding of pAD, showing hypometabolic
changes within the PCC, precuneus, MTL, and temporoparietal
regions that resemble metabolic alterations observed in AD.
Concerning perfusion imaging, more heterogeneous results
have been obtained. In AD, there is evidence for widespread reduc-
tions of CBF, with a negative correlation between CBF and disease
severity [67]. In pAD, both hypo- and hyperperfusion have been
reported in various brain regions in different stages of cognitive
decline [14]. Hays et al. [10] summarized ASL MRI studies in
several models of pAD. In the preclinical phase, hyperperfusion
prevails, mainly in MTL structures, while later in the subclinical
phase, both hyper- and hypoperfusion coexist, with some evidence
of initial hyperperfusion and later hypoperfusion.
CBF dysregulation in pAD and its models suggests the pres-
ence of vascular regulative mechanisms, putatively in response to
an increased need for oxygen and glucose, or changes in cerebral
metabolism to maintain cognitive function [97]. However, the
direction of CBF alterations is not specified. Hyper- as well as
138 Peter Häussermann et al.
Abbreviations: FBI functional brain imaging, pAD preclinical Alzheimer’s disease, MCI mild cognitive impairment, Aß amyloid-ß1–42, rCBF regional cerebral blood flow, RSNs resting-state
networks, DMN default mode network, MTL medial temporal lobe, PCC posterior cingulate cortex, SPECT single photon emission computer tomography, 18F-FDG-PET 18F-fluoro-
desoxy-glucose positron emission tomography, MRI magnetic resonance imaging, fMRI functional magnetic resonance imaging, rs-fMRI resting-state fMRI, ASL arterial spin labeling
a
Preclinical AD as defined by Sperling et al. [1]
b
Only results of nondemented patients are summarized
140 Peter Häussermann et al.
References
1. Sperling RA, Aisen PS, Beckett LA et al lar disorder? Am J Cardiovasc Dis 3(4):
(2011) Toward defining the preclinical stages 197–226
of Alzheimer’s disease: recommendations 12. Trojanowski JQ, Growdon JH (1998) A new
from the National Institute on Aging- consensus report on biomarkers for the early
Alzheimer’s Association workgroups on diag- antemortem diagnosis of Alzheimer disease:
nostic guidelines for Alzheimer’s disease. current status, relevance to drug discovery,
Alzheimers Dement 7(3):280–292. https:// and recommendations for future research. J
doi.org/10.1016/j.jalz.2011.03.003 Neuropathol Exp Neurol 57(6):643–644
2. Jessen F, Amariglio RE, van Boxtel M et al 13. Wierenga CE, Hays CC, Zlatar ZZ (2014)
(2014) A conceptual framework for research Cerebral blood flow measured by arterial spin
on subjective cognitive decline in preclinical labeling MRI as a preclinical marker of
Alzheimer’s disease. Alzheimers Dement Alzheimer’s disease. J Alzheimers Dis
10(6):844–852. https://doi.org/10.1016/j. 42(Suppl 4):S411–S419. https://doi.
jalz.2014.01.001 org/10.3233/JAD-141467
3. Fischer P, Jungwirth S, Zehetmayer S et al 14. Binnewijzend MAA, Benedictus MR, Kuijer
(2007) Conversion from subtypes of mild cog- JPA et al (2016) Cerebral perfusion in the
nitive impairment to Alzheimer dementia. predementia stages of Alzheimer’s disease.
Neurology 68(4):288–291. https://doi. Eur Radiol 26(2):506–514. https://doi.
org/10.1212/01.wnl.0000252358.03285.9d org/10.1007/s00330-015-3834-9
4. Mitchell AJ, Shiri-Feshki M (2009) Rate of 15. Maier FC, Wehrl HF, Schmid AM et al (2014)
progression of mild cognitive impairment to Longitudinal PET-MRI reveals beta-amyloid
dementia—meta-analysis of 41 robust incep- deposition and rCBF dynamics and connects
tion cohort studies. Acta Psychiatr Scand vascular amyloidosis to quantitative loss of
119(4):252–265. https://doi. perfusion. Nat Med 20(12):1485–1492.
org/10.1111/j.1600-0447.2008.01326.x https://doi.org/10.1038/nm.3734
5. Yip AG, McKee AC, Green RC et al (2005) 16. Kato T, Inui Y, Nakamura A et al (2016)
APOE, vascular pathology, and the AD brain. Brain fluorodeoxyglucose (FDG) PET in
Neurology 65(2):259–265. https://doi. dementia. Ageing Res Rev 30:73–84.
o r g / 1 0 . 1 2 1 2 / 0 1 . https://doi.org/10.1016/j.arr.2016.02.003
wnl.0000168863.49053.4d 17. Granert O, Drzezga AE, Boecker H et al
6. Coon KD, Myers AJ, Craig DW et al (2007) (2015) Metabolic topology of neurodegen-
A high-density whole-genome association erative disorders: influence of cognitive and
study reveals that APOE is the major suscep- motor deficits. J Nucl Med 56(12):1916–
tibility gene for sporadic late-onset 1921. https://doi.org/10.2967/
Alzheimer’s disease. J Clin Psychiatry jnumed.115.156067
68(4):613–618 18. Matsuda H (2007) Role of neuroimaging in
7. Saunders AM, Strittmatter WJ, Schmechel D Alzheimer’s disease, with emphasis on brain
et al (1993) Association of apolipoprotein E perfusion SPECT. J Nucl Med 48(8):1289–
allele epsilon 4 with late-onset familial and 1300. https://doi.org/10.2967/
sporadic Alzheimer’s disease. Neurology jnumed.106.037218
43(8):1467–1472 19. Sakamoto S, Matsuda H, Asada T et al (2003)
8. Corder EH, Saunders AM, Strittmatter WJ et Apolipoprotein E genotype and early
al (1993) Gene dose of apolipoprotein E type Alzheimer’s disease: a longitudinal SPECT
4 allele and the risk of Alzheimer’s disease in study. J Neuroimaging 13(2):113–123
late onset families. Science 20. Knopman DS, Jack CR, Wiste HJ et al (2014)
261(5123):921–923 18F-fluorodeoxyglucose positron emission
9. Biomarkers Definitions Working Group tomography, aging, and apolipoprotein E
(2001) Biomarkers and surrogate endpoints: genotype in cognitively normal persons.
preferred definitions and conceptual frame- Neurobiol Aging 35(9):2096–2106. https://
work. Clin Pharmacol Ther 69(3):89–95. d o i . o r g / 1 0 . 1 0 1 6 / j .
https://doi.org/10.1067/mcp.2001.113989 neurobiolaging.2014.03.006
10. Hays CC, Zlatar ZZ, Wierenga CE (2016) 21. Nasrallah I, Dubroff J (2013) An overview of
The utility of cerebral blood flow as a bio- PET neuroimaging. Semin Nucl Med
marker of preclinical Alzheimer’s disease. Cell 43(6):449–461. https://doi.org/10.1053/j.
Mol Neurobiol 36(2):167–179. https://doi. semnuclmed.2013.06.003
org/10.1007/s10571-015-0261-z 22. Buxton RB (2009) Introduction to func-
11. Kelleher RJ, Soiza RL (2013) Evidence of tional magnetic resonance imaging: principles
endothelial dysfunction in the development and techniques, 2nd edn. Cambridge
of Alzheimer’s disease: is Alzheimer’s a vascu- University Press, Cambridge
Functional Imaging in AD 143
23. Damoiseaux JS, Rombouts SARB, Barkhof F 34. Ewers M, Brendel M, Rizk-Jackson A et al
et al (2006) Consistent resting-state networks (2014) Reduced FDG-PET brain metabolism
across healthy subjects. Proc Natl Acad Sci U and executive function predict clinical pro-
S A 103(37):13848–13853. https://doi. gression in elderly healthy subjects.
org/10.1073/pnas.0601417103 Neuroimage Clin 4:45–52. https://doi.
24. Guerra-Carrillo B, Mackey AP, Bunge SA org/10.1016/j.nicl.2013.10.018
(2014) Resting-state fMRI: a window into 35. Ewers M, Insel PS, Stern Y et al (2013)
human brain plasticity. Neuroscientist Cognitive reserve associated with FDG-PET
20(5):522–533. https://doi.org/10.1177/ in preclinical Alzheimer disease. Neurology
1073858414524442 80(13):1194–1201. https://doi.
25. Chuang K-H, van Gelderen P, Merkle H et al org/10.1212/WNL.0b013e31828970c2
(2008) Mapping resting-state functional con- 36. Knopman DS, Jack CR, Wiste HJ et al (2013)
nectivity using perfusion MRI. Neuroimage Selective worsening of brain injury biomarker
40(4):1595–1605. https://doi. abnormalities in cognitively normal elderly
org/10.1016/j.neuroimage.2008.01.006 persons with β-amyloidosis. JAMA Neurol
26. Biswal BB, van Kylen J, Hyde JS (1997) 70(8):1030–1038. https://doi.
Simultaneous assessment of flow and BOLD org/10.1001/jamaneurol.2013.182
signals in resting-state functional connectivity 37. Wirth M, Oh H, Mormino EC et al (2013)
maps. NMR Biomed 10(4–5):165–170 The effect of amyloid β on cognitive decline is
27. Watanabe T, Kan S, Koike T et al (2014) modulated by neural integrity in cognitively
Network-dependent modulation of brain normal elderly. Alzheimers Dement 9(6):687–
activity during sleep. Neuroimage 98:1–10. 698.e1. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.neuroimage. jalz.2012.10.012
2014.04.079 38. Ossenkoppele R, van der Flier WM, Verfaillie
28. Greicius MD, Srivastava G, Reiss AL et al SCJ et al (2014) Long-term effects of amyloid,
(2004) Default-mode network activity distin- hypometabolism, and atrophy on neuropsy-
guishes Alzheimer’s disease from healthy chological functions. Neurology 82(20):
aging: evidence from functional MRI. Proc 1768–1775. https://doi.org/10.1212/
Natl Acad Sci U S A 101(13):4637–4642. WNL.0000000000000432
https://doi.org/10.1073/pnas.0308627101 39. Knopman DS, Jack CR, Wiste HJ et al (2013)
29. Liu TT, Brown GG (2007) Measurement of Brain injury biomarkers are not dependent on
cerebral perfusion with arterial spin labeling: β-amyloid in normal elderly. Ann Neurol
Part 1. Methods. J Int Neuropsychol Soc 73(4):472–480. https://doi.org/10.1002/
13(3):517–525. https://doi.org/10.1017/ ana.23816
S1355617707070646 40. Seo EH, Kim SH, Park SH et al (2016)
30. Lee SP, Duong TQ, Yang G et al (2001) Topographical APOE ɛ4 genotype influence
Relative changes of cerebral arterial and on cerebral metabolism in the continuum of
venous blood volumes during increased cere- Alzheimer’s disease: amyloid burden adjusted
bral blood flow: implications for BOLD analysis. J Alzheimers Dis 54(2):559–568.
fMRI. Magn Reson Med 45(5):791–800 https://doi.org/10.3233/JAD-160395
31. Parkes LM, Rashid W, Chard DT et al (2004) 41. Yi D, Lee DY, Sohn BK et al (2014) Beta-
Normal cerebral perfusion measurements amyloid associated differential effects of
using arterial spin labeling: reproducibility, APOE ε4 on brain metabolism in cognitively
stability, and age and gender effects. Magn normal elderly. Am J Geriatr Psychiatr
Reson Med 51(4):736–743. https://doi. 22(10):961–970. https://doi.
org/10.1002/mrm.20023 org/10.1016/j.jagp.2013.12.173
32. Besson FL, La Joie R, Doeuvre L et al (2015) 42. Krell-Roesch J, Ruider H, Lowe VJ et al
Cognitive and brain profiles associated with (2016) FDG-PET and neuropsychiatric
current neuroimaging biomarkers of preclini- symptoms among cognitively normal elderly
cal Alzheimer’s disease. J Neurosci persons: the Mayo Clinic study of aging. J
35(29):10402–10411. https://doi. Alzheimers Dis 53(4):1609–1616. https://
org/10.1523/JNEUROSCI.0150-15.2015 doi.org/10.3233/JAD-160326
33. Teipel S, Grothe MJ (2016) Does posterior 43. Didic M, Felician O, Gour N et al (2015)
cingulate hypometabolism result from dis- Rhinal hypometabolism on FDG PET in
connection or local pathology across preclini- healthy APO-E4 carriers: impact on memory
cal and clinical stages of Alzheimer’s disease? function and metabolic networks. Eur J Nucl
Eur J Nucl Med Mol Imaging 43(3): Med Mol Imaging 42(10):1512–1521.
526–536. https://doi.org/10.1007/ https://doi.org/10.1007/s00259-015-
s00259-015-3222-3 3057-y
144 Peter Häussermann et al.
Alzheimer’s disease: relation to apolipopro- 92. Kim SM, Kim MJ, Rhee HY et al (2013)
tein E polymorphism. J Neurol Neurosurg Regional cerebral perfusion in patients with
Psychiatry 64(6):742–746 Alzheimer’s disease and mild cognitive
83. Lehtovirta M, Soininen H, Laakso MP et al impairment: effect of APOE epsilon4 allele.
(1996) SPECT and MRI analysis in Neuroradiology 55(1):25–34. https://doi.
Alzheimer’s disease: relation to apolipopro- org/10.1007/s00234-012-1077-x
tein E epsilon 4 allele. J Neurol Neurosurg 93. Beason-Held LL, Goh JO, An Y et al (2013)
Psychiatry 60(6):644–649 Changes in brain function occur years before
84. Devanand DP, van Heertum RL, Kegeles LS the onset of cognitive impairment. J
et al (2010) (99m)Tc hexamethyl-propylene- Neurosci 33(46):18008–18014. https://
aminoxime single-photon emission computed doi.org/10.1523/JNEUROSCI.
tomography prediction of conversion from 1402-13.2013
mild cognitive impairment to Alzheimer dis- 94. Adriaanse SM, Sanz-Arigita EJ, Binnewijzend
ease. Am J Geriatr Psychiatr 18(11):959–972. MAA et al (2014) Amyloid and its association
https://doi.org/10.1097/ with default network integrity in Alzheimer’s
JGP.0b013e3181ec8696 disease. Hum Brain Mapp 35(3):779–791.
85. Alegret M, Cuberas-Borrós G, Espinosa A et https://doi.org/10.1002/hbm.22213
al (2014) Cognitive, genetic, and brain perfu- 95. Binnewijzend MAA, Schoonheim MM, Sanz-
sion factors associated with four year inci- Arigita E et al (2012) Resting-state fMRI
dence of Alzheimer’s disease from mild changes in Alzheimer’s disease and mild cog-
cognitive impairment. J Alzheimers Dis nitive impairment. Neurobiol Aging
41(3):739–748. https://doi.org/10.3233/ 33(9):2018–2028. https://doi.
JAD-132516 org/10.1016/j.neurobiolaging.2011.07.003
86. Fleisher AS, Sherzai A, Taylor C et al (2009) 96. Liang P, Wang Z, Yang Y et al (2012) Three
Resting-state BOLD networks versus task- subsystems of the inferior parietal cortex are
associated functional MRI for distinguishing differently affected in mild cognitive impair-
Alzheimer’s disease risk groups. Neuroimage ment. J Alzheimers Dis 30(3):475–487.
47(4):1678–1690. https://doi. https://doi.org/10.3233/
org/10.1016/j.neuroimage.2009.06.021 JAD-2012-111721
87. Harrison TM, Burggren AC, Small GW et al 97. Ostergaard L, Aamand R, Gutierrez-Jimenez
(2016) Altered memory-related functional E et al (2013) The capillary dysfunction
connectivity of the anterior and posterior hip- hypothesis of Alzheimer’s disease. Neurobiol
pocampus in older adults at increased genetic Aging 34(4):1018–1031. https://doi.
risk for Alzheimer’s disease. Hum Brain Mapp org/10.1016/j.neurobiolaging.2012.09.011
37(1):366–380. https://doi.org/10.1002/ 98. Alexopoulos P, Sorg C, Forschler A et al
hbm.23036 (2012) Perfusion abnormalities in mild cog-
88. Sheline YI, Morris JC, Snyder AZ et al (2010) nitive impairment and mild dementia in
APOE4 allele disrupts resting state fMRI con- Alzheimer’s disease measured by pulsed arte-
nectivity in the absence of amyloid plaques or rial spin labeling MRI. Eur Arch Psychiatry
decreased CSF Aβ42. J Neurosci Clin Neurosci 262(1):69–77. https://doi.
30(50):17035–17040. https://doi.org/ org/10.1007/s00406-011-0226-2
10.1523/JNEUROSCI.3987-10.2010 99. Dai W, Lopez OL, Carmichael OT et al
89. Michels L, Warnock G, Buck A et al (2016) (2009) Mild cognitive impairment and
Arterial spin labeling imaging reveals wide- alzheimer disease: patterns of altered cerebral
spread and Abeta-independent reductions in blood flow at MR imaging. Radiology
cerebral blood flow in elderly apolipoprotein 250(3):856–866. https://doi.org/10.1148/
epsilon-4 carriers. J Cereb Blood Flow Metab radiol.2503080751
36(3):581–595. https://doi.org/10.1177/ 100. Johnson NA, Jahng G-H, Weiner MW et al
0271678X15605847 (2005) Pattern of cerebral hypoperfusion in
90. Bangen KJ, Restom K, Liu TT et al (2012) Alzheimer disease and mild cognitive impair-
Assessment of Alzheimer’s disease risk with ment measured with arterial spin-labeling MR
functional magnetic resonance imaging: an imaging: initial experience. Radiology
arterial spin labeling study. J Alzheimers Dis 234(3):851–859. https://doi.org/10.1148/
31(Suppl 3):S59–S74. https://doi.org/ radiol.2343040197
10.3233/JAD-2012-120292 101. Xu G, Antuono PG, Jones J et al (2007)
91. Wierenga CE, Clark LR, Dev SI et al (2013) Perfusion fMRI detects deficits in regional
Interaction of age and APOE genotype on CBF during memory-encoding tasks in MCI
cerebral blood flow at rest. J Alzheimers Dis subjects. Neurology 69(17):1650–1656.
34(4):921–935. https://doi.org/10.3233/ https://doi.org/10.1212/01.
JAD-121897 wnl.0000296941.06685.22
Functional Imaging in AD 147
102. Xie L, Dolui S, Das SR et al (2016) A brain and frontotemporal dementia: two sides of
stress test: cerebral perfusion during memory the same coin? Eur Radiol 25(10):
encoding in mild cognitive impairment. 3050–3059. https://doi.org/10.1007/
Neuroimage Clin 11:388–397. https://doi. s00330-015-3696-1
org/10.1016/j.nicl.2016.03.002 107. Wang L, Day J, Roe CM et al (2014) The
103. Vanitallie TB (2013) Preclinical sporadic
effect of APOE ε4 allele on cholinesterase
Alzheimer’s disease: target for personalized inhibitors in patients with Alzheimer disease:
diagnosis and preventive intervention. Metab evaluation of the feasibility of resting state
Clin Exp 62(Suppl 1):S30–S33. https://doi. functional connectivity magnetic resonance
org/10.1016/j.metabol.2012.08.024 imaging. Alzheimer Dis Assoc Disord
104. Lee IH, Kim ST, Kim H-J et al (2010)
28(2):122–127. https://doi.org/10.1097/
Analysis of perfusion weighted image of CNS WAD.0b013e318299d096
lymphoma. Eur J Radiol 76(1):48–51. 108. Sheline YI, Raichle ME (2013) Resting state
https://doi.org/10.1016/j. functional connectivity in preclinical
ejrad.2009.05.013 Alzheimer’s disease. Biol Psychiatry
105. Jack CR, Wiste HJ, Weigand SD et al (2013) 74(5):340–347. https://doi.org/10.1016/j.
Amyloid-first and neurodegeneration-first biopsych.2012.11.028
profiles characterize incident amyloid PET 109. Morbelli S, Brugnolo A, Bossert I et al (2015)
positivity. Neurology 81(20):1732–1740. Visual versus semi-quantitative analysis of
https://doi.org/10.1212/01. 18F-FDG-PET in amnestic MCI: an
wnl.0000435556.21319.e4 European Alzheimer’s Disease Consortium
106. Verfaillie SCJ, Adriaanse SM, Binnewijzend (EADC) project. J Alzheimers Dis 44(3):
MAA et al (2015) Cerebral perfusion and 815–826. https://doi.org/10.3233/
glucose metabolism in Alzheimer’s disease JAD-142229
Chapter 9
Abstract
The content of this chapter deals with the usefulness and indications of amyloid PET for the diagnosis of
patients with Alzheimer’s disease (AD). In addition to a description of amyloid PET itself, topics such as
early and differential diagnosis of AD are discussed. Measuring amyloid in CSF as an alternative method to
amyloid PET and the associations between cerebral amyloid load and the apolipoprotein E genotype are
also covered in this section.
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_9, © Springer Science+Business Media, LLC 2018
149
150 Timo Grimmer and Jennifer Grace Perryman
2.1 Differential The value of amyloid PET for differential diagnosis varies between
Diagnosis the particular causes of dementia, depending on the occurrence of
cerebral amyloid load [7]. The first study that successfully used
amyloid PET to differentiate between semantic dementia, a sub-
type of the frontotemporal lobar degenerations (FTLD) [8] usu-
ally caused by non-amyloid pathologies, and AD yielded a perfect
group discrimination [9]. A similar result was achieved differenti-
ating AD from the behavioral variant of frontotemporal dementia
(bvFTD) [10], another FTLD subtype. However, distinguishing
AD from dementias associated with Parkinson’s disease, such as
Lewy body disease (LBD), may be difficult as heightened cerebral
amyloid plaque load can be found in both [11]. The usefulness of
amyloid PET for differential diagnosis is supported by a recent
meta-analysis confirming the associations between amyloid positiv-
ity as shown by amyloid imaging and clinical diagnosis [12]:
Patients with the clinical diagnosis of dementia due to AD were
93% amyloid positive at 50 years of age and 79% at 90 years of age.
Although this finding does not seem intuitive at first glance, there
are several explanations as to why a smaller percentage of amyloid
positivity prevalence can be found in older patients. The primary
cause may be the occurrence of rivaling causes for dementia in
older patients, resembling the clinical picture of AD. Although less
likely, it may be possible that either AD in elderly patients is caused
less by cerebral amyloid load or that the tracer binds less efficiently
to amyloid of older patients. The mean prevalence estimate for
patients with the clinical diagnosis of dementia due to AD was
88%. Patients with dementia due to Lewy bodies (DLB) were amy-
loid positive in 51% of cases, a figure compatible with the histo-
pathological finding that around 50% of patients with DLB show
amyloid depositions as discussed above. Patients with FTLD were
amyloid positive only in 12% of cases, which is compatible with the
histopathological finding that FTLD are caused by non-amyloid
pathology only.
Imaging of Brain Amyloid Load in Early Alzheimer’s Disease 151
2.2 Early Diagnosis Amyloid PET may also be used as a means to diagnose AD at a very
early stage as amyloid deposition precedes initial clinical symptoms
by years if not decades [13], with estimates giving a time period of
20–30 years between the first discovery of an abnormal amyloid
load and the beginning of dementia. In patients with mild cogni-
tive impairment (MCI), a predisposing factor and frequent precur-
sor of AD, a positive amyloid PET, for example, using the [11C]
PiB compound, can be found in 50% [1]. In accordance with the
assumption that amyloid precedes clinical symptoms, it was shown
that amyloid positivity in clinically asymptomatic persons increases
with age [12]. Usage of amyloid PET could therefore help dis-
cover the ailment and warn patients many decades prior to the
outbreak of symptoms.
As there is currently no treatment available for AD at an asymp-
tomatic stage, however, the advantages of early diagnosis are debat-
able. Nevertheless, it should be mentioned that early diagnosis
would allow patients to plan better for their future circumstances
regarding financial matters and caregiving and also inform them-
selves and relevant people about matters such as safety in everyday
life, driving, and counseling options [14].
2.3 Excluding As well as indicating a progression to AD, amyloid PET may also
Alzheimer’s Disease be used to rule out the disease as the cause of dementia: According
to the guidance documents of the German Society of Nuclear
Medicine and the American Society of Nuclear Medicine and
Molecular Imaging [15, 16], it is highly unlikely to suffer from
Alzheimer’s disease without cerebral amyloid load in the form of
plaques. Should the amyloid PET therefore be negative, one may
assume with a high probability that AD is not the cause for the
patient’s dementia.
It may also help reassure patients who are concerned about
developing the illness in the future. A survey by the Harvard School
of Public Health showed that around 89% of patients stated that
they would like to be informed whether or not AD was the cause
for their impairments [14].
2.4 Limitations Negative amyloid PET findings may be due to misdiagnosis; unsuc-
cessful tracer production may also be possible in some cases [7].
Another potential limitation is the availability of tracer produc-
tion/delivery and PET cameras. Furthermore, one should keep in
mind that the examination involves radiation exposure
(5.8–7.0 mSv).
152 Timo Grimmer and Jennifer Grace Perryman
2.5 Appropriate Use Amyloid PET is already commonly clinically used and recom-
Criteria mended in several guidelines (German Society of Nuclear
Medicine (DGN) [16], European Association of Nuclear
Medicine (EANM) [17], the American Society of Nuclear
Medicine and Molecular Imaging (SNMMI) [15], German S3
Guideline Dementia [18]). In order to provide a framework for
guiding clinicians who intend to apply amyloid PET, appropriate
use criteria for this particular method were published in 2013
[15]. For patients suffering from a subjectively and objectively
identified cognitive impairment, with AD as a likely, but as yet
undecided by experts, diagnosis, and assuming that a positive or
negative amyloid PET would provide assurance for the clinical
diagnosis, the authors concluded that amyloid PET is a useful
tool to draw on in the following situations:
1. The patient suffers from MCI in a persistent or progressively
unexplained form.
2. Patients with the clinical criteria for possible AD but with
unclear clinical presentation, e.g., an atypical clinical course or
etiologically mixed.
3. Patient with early-onset dementia.
The following scenarios are deemed as inappropriate for the
use of amyloid PET:
1. Patient fulfilling the clinical criteria for probable AD including
typical age of onset.
2. To determine the severity of the dementing syndrome.
3. Asymptomatic persons with a positive family history of demen-
tia or presence of apolipoprotein E (APOE) ε4 only.
4. The patient reports cognitive complaints which are, however,
without substantiation on clinical examination (memory
complainer).
5. In asymptomatic individuals.
6. For nonmedical use (e.g., legal insurance coverage or employ-
ment screening).
Unfortunately, the authors missed one important indication:
identifying participants for anti-amyloid therapy. In persons or
patients interested in participating in an anti-amyloid drug trial,
amyloid positivity needs to be confirmed in advance. This may be
achieved in vivo and noninvasively by amyloid PET.
Association of cerebral amyloid load and the apolipoprotein
E 4 genotype and its implications, as well as an alternative method
for assessing amyloid in vivo, shall be discussed in the notes sec-
tion below.
Imaging of Brain Amyloid Load in Early Alzheimer’s Disease 153
3 Materials
4 Method
Fig. 1 Amyloid PET using 11C-PiB in a patient with dementia due to Alzheimer’s
disease overlaid on an MRI scan, sagittal view
References
1. Jansen WJ, Ossenkoppele R, Knol DL et al totemporal dementia. The Lund and
(2015) Prevalence of cerebral amyloid pathol- Manchester Groups. J Neurol Neurosurg
ogy in persons without dementia: a meta- Psychiatry 57:416–418
analysis. JAMA 313:1924–1938 9. Drzezga A, Grimmer T, Henriksen G et al
2. Braak H, .Braak E (1991) Neuropathological (2008) Imaging of amyloid plaques and cere-
stageing of Alzheimer-related changes. Acta bral glucose metabolism in semantic dementia
Neuropathol 82:239–259 and Alzheimer’s disease. Neuroimage
3. Thal DR, Rüb U, Orantes M, Braak H (2002) 39:619–633
Phases of Aß-deposition in the human brain 10. Engler H, Santillo AF, Wang SX et al (2008) In
and its relevance for the development of vivo amyloid imaging with PET in frontotem-
AD. Neurology 58:1791–1800 poral dementia. Eur J Nucl Med Mol Imaging
4. Grimmer T, Tholen S, Yousefi BH et al (2010) 35:100–106
Progression of cerebral amyloid load is associ- 11. McKeith IG, Dickson DW, Lowe J et al (2005)
ated with the apolipoprotein E epsilon4 geno- Diagnosis and management of dementia with
type in Alzheimer’s disease. Biol Psychiatry Lewy bodies: third report of the DLB
68:879–884 Consortium. Neurology 65:1863–1872
5. Klunk WE, Engler H, Nordberg A et al (2004) 12. Ossenkoppele R, Jansen WJ, Rabinovici GD
Imaging brain amyloid in Alzheimer’s disease et al (2015) Prevalence of amyloid PET positiv-
with Pittsburgh Compound-B. Ann Neurol ity in dementia syndromes: a meta-analysis.
55:306–319 JAMA 313:1939–1949
6. Ikonomovic MD, Klunk WE, Abrahamson EE 13. Bateman RJ, Xiong C, Benzinger TL et al
et al (2008) Post-mortem correlates of in vivo (2012) Clinical and biomarker changes in
PiB-PET amyloid imaging in a typical case of dominantly inherited Alzheimer’s disease. N
Alzheimer’s disease. Brain 131:1630–1645 Engl J Med 367:795–804
7. Grimmer T, Drzezga A, Kurz A (2010) 14. Blendon RJ, Benson JM, Wikler EM et al
Visualization of amyloid with positron emis- (2012) The impact of experience with a family
sion tomography. Useful improvement in the member with Alzheimer’s disease on views
diagnosis of dementia? Nervenarzt about the disease across five countries. Int
81:602–606 J Alzheimers Dis 2012:903645
8. Brun A, Englund B, Gustafson L et al (1994) 15. Johnson KA, Minoshima S, Bohnen NI et al
Clinical and neuropathological criteria for fron- (2013) Appropriate use criteria for amyloid
156 Timo Grimmer and Jennifer Grace Perryman
PET: a report of the Amyloid Imaging Task Compound B amyloid imaging PET studies: a
Force, the Society of Nuclear Medicine and comparative analysis. J Nucl Med
Molecular Imaging, and the Alzheimer’s 46:1959–1972
Association. J Nucl Med 54:476–490 27. Friston KJ, Holmes AP, Worsley KJ (1994)
16. Barthel, H., Meyer, P. T., Drzezga, A., Statistical parametric mapping in functional
Bartenstein, P., Boecker, H., Brust, P., Buchert, imaging: a general linear approach. Hum Brain
R., Coenen, H. H., La Fourgère, C., Gründer, Mapp 2:189–210
G., Grünwald, F., Krause, B. J., Kuwert, T., 28. Ziolko SK, Weissfeld LA, Klunk WE et al
Schreckenberger, M., Tatsch, K., Langen, K. J., (2006) Evaluation of voxel-based methods for
and Sabri, O. (2015) Beta- amyloid- PET- the statistical analysis of PIB PET amyloid
Bildgebung des Gehirns – DGN- imaging studies in Alzheimer’s disease.
Handlungsempfehlung (S1-Leitlinie). www. Neuroimage 33:94–102
awmf.org/uploads/tx_szleitlinien/031-052l_ 29. Minoshima S, Frey KA, Koeppe RA, Foster
S 1 _ B e t a _ A m y l o i d - P E T- B i l d g e b u n g _ NL, Kuhl DE (1995) A diagnostic approach in
Gehirn_2016-02.pdf Alzheimer’s disease using three-dimensional
17. Minoshima S, Drzezga AE, Barthel H et al stereotactic surface projections of fluorine-18-
(2016) SNMMI procedure standard/EANM FDG PET. J Nucl Med 36:1238–1248
practice guideline for amyloid PET imaging of 30. Grimmer T, Wutz C, Alexopoulos P et al (2016)
the brain 1.0. J Nucl Med 57:1316–1322 Visual versus fully automated analyses of 18F-
18. Deutsche Gesellschaft für Psychiatrie und FDG and amyloid PET for prediction of
Psychotherapie, Psychosomatik und dementia due to Alzheimer disease in mild cog-
Nervenheilkunde DGPPN and Deutsche nitive impairment. J Nucl Med 57:204–207
Gesellschaft für Neurologie (DGN) (2016) 31. Palmqvist S, Mattsson N, Hansson O (2016)
S3-Leitlinie “Demenzen”. https://www. Cerebrospinal fluid analysis detects cerebral
dgppn.de/fileadmin/user_upload/_medien/ amyloid-beta accumulation earlier than positron
download/pdf/kurzversion-leitlinien/REV_ emission tomography. Brain 139:1226–1236
S3-leiltlinie-demenzen.pdf
32. Fagan AM, Mintun MA, Shah AR et al (2009)
19. https://www.accessdata.fda.gov/drugsatfda_ Cerebrospinal fluid tau and ptau(181) increase
docs/label/2012/202008s000lbl.pdf. 18 Apr with cortical amyloid deposition in cognitively
2017 normal individuals: implications for future clin-
20. https://www.accessdata.fda.gov/drugsatfda_ ical trials of Alzheimer’s disease. EMBO Mol
docs/label/2016/203137s005lbl.pdf. 18 Apr Med 1:371–380
2017 33. Mattsson N, Insel PS, Landau S et al (2014)
21. https://www.accessdata.fda.gov/drugsatfda_ Diagnostic accuracy of CSF Ab42 and florbeta-
docs/label/2014/204677s000lbl.pdf. 18 Apr pir PET for Alzheimer’s disease. Ann Clin
2017 Transl Neurol 1:534–543
22. Landau SM, Breault C, Joshi AD et al (2013) 34. Grimmer T, Riemenschneider M, Forstl H et al
Amyloid-beta imaging with Pittsburgh com- (2009) Beta amyloid in Alzheimer’s disease:
pound B and florbetapir: comparing radiotrac- increased deposition in brain is reflected in
ers and quantification methods. J Nucl Med reduced concentration in cerebrospinal fluid.
54:70–77 Biol Psychiatry 65:927–934
23. Vandenberghe R, Van Laere K, Ivanoiu A et al 35. Drzezga A, Grimmer T, Henriksen G et al
(2010) 18F-flutemetamol amyloid imaging in (2009) Effect of APOE genotype on amyloid
Alzheimer disease and mild cognitive impair- plaque load and gray matter volume in
ment: a phase 2 trial. Ann Neurol 68:319–329 Alzheimer disease. Neurology 72:1487–1494
24. Villemagne VL, Mulligan RS, Pejoska S et al 36. Cosentino S, Scarmeas N, Helzner E et al
(2012) Comparison of 11C-PiB and (2008) APOE epsilon 4 allele predicts faster
18F-florbetaben for Abeta imaging in ageing cognitive decline in mild Alzheimer disease.
and Alzheimer’s disease. Eur J Nucl Med Mol Neurology 70:1842–1849
Imaging 39:983–989 37. Deane R, Sagare A, Hamm K et al (2008) apoE
25. Grimmer T, Henriksen G, Wester HJ et al isoform-specific disruption of amyloid beta
(2009) Pittsburgh Compound B(PIB) uptake peptide clearance from mouse brain. J Clin
in positron emission tomography is associated Invest 118:4002–4013
with clinical severity of Alzheimer’s disease. 38. Craft S, Teri L, Edland SD et al (1998)
Neurobiol Aging 30(12):1902–1909 Accelerated decline in apolipoprotein
26. Lopresti BJ, Klunk WE, Mathis CA et al (2005) E-epsilon4 homozygotes with Alzheimer’s dis-
Simplified quantification of Pittsburgh ease. Neurology 51:149–153
Chapter 10
Abstract
Quantitative EEG analyses, especially connectivity and graph analyses, are promising biomarkers in
neurodegenerative disorders such as Alzheimer’s disease. To develop reliable and valid biomarkers,
many aspects in pre- and post-processing of EEG data are important.
Artifact detection and segment selection are done by visual inspection or automated routines, both
with specific advantages and difficulties. Inverse solution is an elegant way of solving the issue of common
source but may be somewhat imprecise in the absence of defined graphoelements such as epileptiform
discharges. Connectivity analyses allow a description of interactions between brain regions, which may
represent one of the earliest signs of cognitive decline.
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_10, © Springer Science+Business Media, LLC 2018
157
158 Florian Hatz and Peter Fuhr
Fig. 1 (continued) from the resulting values, and signals with a z-value >3 are labeled as “bad.” Results are
summarized in a matrix (lower part of the figure). All signal periods labeled as “bad” by one of the methods are
represented in red, the others in green color. For selection of bad channels or activations, the resulting matrix
is evaluated horizontally (lower right matrix in figure, threshold: >70% bad signals). For selection of segments,
the green squares (= good signals) in the resulting matrix are replaced by Peak2Min values (brighter green
colors represent higher Peak2Min values), the red squares by zero values. Maximizing the summed values of
the squares allows the automated selection of the optimal segments for further processing (lower left matrix
in figure; blue frame represents a selected segment)
Fig. 1 Automated selection of segments, bad channels, and ICA activations in TAPEEG. TAPEEG includes
routines to automatically select segments of EEG for processing and the detection of channels with high arti-
fact load (bad channels). For artifact reduction, the automated selection of bad ICA activations is possible. For
all three tasks, TAPEEG processes the EEG data similarly, using the workflow depicted in the figure. EEG data
is first splitted into periods (e.g. 4 s). In every period, the signals (= channels) are analyzed using a set of
predefined methods as listed in the central boxes of the figure. Every method evaluates the signals
independently. FFT = Fast Fourier transformation using an 80% Hanning window. Band power 0.5–2 Hz > 70%,
band power 15–30 Hz > 50%, and 50 Hz-power > 50% are labeled as bad. Hurst exponent = Hurst exponent
is a measure of long-range dependence within a signal, with a range of 0–1. Human EEG has values around 0.7
[41]. Topo correlation = Correlation to surrounding channels (high correlation values are expected for high density
EEG). Median gradient = Median value of all 1 timeframe differences in amplitude of every channel.
Kurtosis = Kurtosis measures the peakedness of data and is high for ICA activations loading on one single elec-
trode only. EOG correlation = Correlation to electrooculogram. ECG correlation = Correlation of peaks in signals to
peaks in electrocardiogram. Peak2Min = Ratio of amplitude of peak frequency and minimal amplitude in lower
frequency for every period in parieto-occipital electrodes (amplitude of IAF divided by amplitude of TF in ).
Peak2Min represents the amount of alpha activity and is positively correlated to alertness [10]. For Hurst expo-
nent, variance, topo correlation, median gradient, kurtosis, ECG, and EOG correlation, z-values are c alculated
160 Florian Hatz and Peter Fuhr
Fig. 2 Calculation of individual frequency bands according to Klimesch 1999. IAF = individual alpha frequency,
TF = transition frequency between theta and alpha
3 Connectivity Analyses
4 Inverse Solutions
5 Graph Analysis
7 Conclusions
References
1. Caviness JN, Hentz JG, Evidente VG, 5. Wallstrom GL, Kass RE, Miller A, Cohn JF,
Driver-Dunckley E, Samanta J, Mahant P,
Fox NA (2004) Automatic correction of
Connor DJ, Sabbagh MN, Shill HA, Adler CH ocular artifacts in the EEG: a comparison of
(2007) Both early and late cognitive dysfunction regression-based and component-based m ethods.
affects the electroencephalogram in Parkinson’s Int J Psychophysiol 53:105–119. https://doi.
disease. Parkinsonism Relat Disord 13:348–354 org/10.1016/j.ijpsycho.2004.03.007
2. Fonseca LC, Tedrus GMAS, Letro GH, Bossoni 6. Nolan H, Whelan R, Reilly RB (2010) FASTER:
AS (2009) Dementia, mild cognitive impairment fully automated statistical thresholding for
and quantitative EEG in patients with Parkinson’s EEG artifact rejection. J Neurosci Methods
disease. Clin EEG Neurosci 40:168–172 192:152–162. https://doi.org/10.1016/j.
3. Roh JH, Park MH, Ko D, Park K-W, Lee D-H, jneumeth.2010.07.015
Han C, Jo SA, Yang K-S, Jung K-Y (2011) 7. Brunet D, Murray MM, Michel CM (2011)
Region and frequency specific changes of spectral Spatiotemporal analysis of multichannel
power in Alzheimer’s disease and mild cognitive EEG: CARTOOL. Comput Intell
impairment. Clin Neurophysiol 122:2169–2176. Neurosci 2011:813870. https://doi.
https://doi.org/10.1016/j.clinph.2011.03.023 org/10.1155/2011/813870
4. Gross J, Baillet S, Barnes GR, Henson RN, 8. Tadel F, Baillet S, Mosher JC, Pantazis D,
Hillebrand A, Jensen O, Jerbi K, Litvak V, Leahy RM (2011) Brainstorm: a user-friendly
Maess B, Oostenveld R, Parkkonen L, Taylor application for MEG/EEG analysis. Comput
JR, van Wassenhove V, Wibral M, Schoffelen Intell Neurosci 2011:879716. https://doi.
J-M (2013) Good practice for conducting org/10.1155/2011/879716
and reporting MEG research. Neuroimage 9. Oostenveld R, Fries P, Maris E, Schoffelen JM
65:349–363. https://doi.org/10.1016/j. (2011) FieldTrip: open source software for
neuroimage.2012.10.001 advanced analysis of MEG, EEG, and invasive
EEG Connectivity in AD 165
33. Vecchio F, Miraglia F, Piludu F, Granata G, cortical sources of resting state EEG rhythms.
Romanello R, Caulo M, Onofrj V, Bramanti Front Neurosci. https://doi.org/10.3389/
P, Colosimo C, Rossini PM (2017) “Small fnins.2016.00047
World” architecture in brain connectivity and 38. Ranasinghe KG, Hinkley LB, Beagle AJ,
hippocampal volume in Alzheimer’s disease: a Mizuiri D, Dowling AF, Honma SM, Finucane
study via graph theory from EEG data. Brain MM, Scherling C, Miller BL, Nagarajan SS,
Imaging Behav 11:473–485. https://doi. Vossel KA (2014) Regional functional connec-
org/10.1007/s11682-016-9528-3 tivity predicts distinct cognitive impairments
34. Hatz F, Hardmeier M, Benz N, Ehrensperger M, in Alzheimer’s disease spectrum. Neuroimage
Gschwandtner U, Rüegg S, Schindler C, Monsch Clin 5:385–395. https://doi.org/10.1016/j.
AU, Fuhr P (2015) Microstate connectivity nicl.2014.07.006
alterations in patients with early Alzheimer’s 39. Triggiani AI, Bevilacqua V, Brunetti A, Lizio
disease. Alzheimers Res Ther 7:78. https://doi. R, Tattoli G, Cassano F, Soricelli A, Ferri R,
org/10.1186/s13195-015-0163-9 Nobili F, Gesualdo L, Barulli MR, Tortelli
35. Suárez-Revelo JX, Ochoa-Gómez JF, R, Cardinali V, Giannini A, Spagnolo P,
Duque-Grajales JE, Tobón-Quintero CA
Armenise S, Stocchi F, Buenza G, Scianatico G,
(2016) Biomarkers identification in Alzheimer’s Logroscino G, Lacidogna G, Orzi F, Buttinelli
disease using effective connectivity analysis
C, Giubilei F, Del Percio C, Frisoni GB,
from electroencephalography recordings. Ing Babiloni C (2017) Classification of healthy
E Investig 36:50–57. 10.15446/ing.investig. subjects and Alzheimer’s disease patients with
v36n3.54037 dementia from cortical sources of resting state
36. Hardmeier M, Hatz F, Bousleiman H, Schindler EEG rhythms: a study using artificial neu-
C, Stam CJ, Fuhr P (2014) Reproducibility of ral networks. Front Neurosci. https://doi.
functional connectivity and graph measures based org/10.3389/fnins.2016.00604
on the phase lag index (PLI) and weighted phase 40. Amur S, LaVange L, Zineh I, Buckman-Garner
lag index (wPLI) derived from high resolution S, Woodcock J (2015) Biomarker qualifica-
EEG. PLoS One 9:e108648. https://doi. tion: toward a multiple stakeholder framework
org/10.1371/journal.pone.0108648 for biomarker development, regulatory accep-
37. Babiloni C, Triggiani AI, Lizio R, Cordone tance, and utilization. Clin Pharmacol Ther
S, Tattoli G, Bevilacqua V, Soricelli A, Ferri 98:34–46. https://doi.org/10.1002/cpt.136
R, Nobili F, Gesualdo L, Millán-Calenti JC, 41. Bian N-Y, Wang B, Cao Y, Zhang L (2006)
Buján A, Tortelli R, Cardinali V, Barulli MR, Automatic removal of artifacts from EEG data
Giannini A, Spagnolo P, Armenise S, Buenza using ICA and exponential analysis. In: Proc.
G, Scianatico G, Logroscino G, Frisoni GB, third int. conf. advnaces neural netw.—vol
del Percio C (2016) Classification of single Part II. Springer-Verlag, Berlin, Heidelberg,
normal and Alzheimer’s disease individuals from pp 719–726
Part IV
Biomarker Discovery
Chapter 11
Abstract
Mass spectrometry plays an increasingly important role in the biomarker field with the advent of targeted
proteomics. Tryptic peptides from a protein of interest can be used to create a targeted assay to interrogate
cerebrospinal fluid (CSF) for biomarkers. Since heterozygous mutations in the TREM2 gene have been
associated with an increased risk of Alzheimer’s disease, measuring this soluble protein in CSF has become
a priority. This chapter demonstrates the development, optimization, and validation of a method to measure
soluble TREM2 using a single reaction monitoring (SRM) targeted mass spectrometry assay.
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_11, © Springer Science+Business Media, LLC 2018
169
170 Amanda J. Heslegrave et al.
2 Materials
2.2 Samples CSF samples were collected from memory clinics in Sweden and
the UK according to local ethics procedures. Samples were collected
in the morning into polypropylene tubes (Sarstedt, Numbrecht,
Germany) according to standard operating procedures; samples
were then further aliquoted into polypropylene tubes, frozen, and
stored at −80 °C within 2 h.
2.3 Reagents All reagents are of mass spectrometry grade and prepared and
stored at room temperature unless otherwise stated.
2.3.1 Digestion Reagents Digestion buffer—100 mM Tris pH 7.8 containing 6 M urea, 2 M
thiourea, and 2% amidosulfobetaine-14 (ASB-14). 2 M dithioth-
reitol (DTT) is made by dissolving 30 mg of DTT in 1 mL of
100 mM Tris pH 7.8. It is important that this solution is made up
172 Amanda J. Heslegrave et al.
fresh on the day and kept on ice until use. Iodoacetamide (IAA)
was prepared by dissolving 36 mg in 1 mL of 100 mM Tris pH 7.8;
similarly this solution was made up fresh on the day and protected
from light. Sequence grade trypsin (Promega, UK) is prepared by
resuspending lyophilized 1 mg aliquots of the protease with 11 μL
of 40 mM ammonium bicarbonate buffer. All reagents are from
Sigma-Aldrich, Dorset, UK, unless otherwise stated.
2.3.2 Mass Spectrometry MS mobile phases are A, LC-MS grade water with 0.1% formic
Reagents acid (FA), and B, LC-MS grade ACN with 0.1% FA.
3 Methods
3.1 SRM Set Up To set up the SRM assay for sTREM2, we first identified a unique
peptide from the protein sequence using Skyline, https://skyline.
ms/project/home/software/Skyline/. Skyline is a freely available
and open source Windows client application for building selected
reaction monitoring (SRM)/multiple reaction monitoring (MRM)
and parallel reaction monitoring (PRM—targeted MS/MS),
although, it would be preferable to identify peptides from previous
mass spec discovery experiments. Another useful resource is the
Global Proteome Machine http://www.thegpm.org/. We checked
that the peptide was completely unique to TREM2 and that none
of the amino acids would be subject to posttranslational modifica-
tion. This peptide was synthesized by Generon (Berkshire, UK)
and then used to develop the assay.
For tuning of the peptide, standards are reconstituted in 50%
ACN containing 0.1% FA before being infused into the mass
spectrometer. The amount of peptide will need to be optimized
but a suggested starting point is 100 μg/mL. A full scan can be
performed initially to optimize tuning concentration, and the
peptide is diluted accordingly. Once the concentration has been
optimized, optimal collision and cone energies are obtained
using the automatic “Intellistart” tuning software of the Xevo
TQS mass spectrometry system. After the optimal fragmentation
and MS parameters are obtained and entered into the SRM, the
retention time of the peptide is assessed. In this case a 10 min
chromatographic run time is used, which is shown in Table 1
(Notes 1 and 2).
sTREM2 in CSF 173
Table 1
10-minute chromatographic run
3.2 Sample 100 μL of each CSF sample is aliquoted into a fresh siliconized
Preparation Eppendorf tube and freeze dried. The resulting proteins are then
resuspended in 20 μL of digestion buffer and shaken at RT for
20 min. 1.5 μL of 2 M DTT is added to reduce disulfide bridges
and the sample shaken for a further hour at RT. 3 μL of (2 M) IAA
is added in order to carboamidomethylate any cysteines contained
in the proteins, and the sample is then incubated at RT for a fur-
ther 45 min in the dark. The sample is then diluted with 165.5 μL
of LC-MS grade water and 10 μL of a solution containing 1 μg of
trypsin. Samples are then vortexed and left in a water bath at 37 °C
overnight. Before mass spectral analysis, centrifuge samples at
13,000 × g for 10 min to remove particulates before transfer into
vials suitable for mass spectral analysis.
3.3 Mass Using the chromatographic and mass spectral conditions devel-
Spectrometry oped and described above, the samples can be input into the
sample manager and the analyses completed. During any analyses
a standard curve of your peptide should also be included, and this
should cover the range that the endogenous levels of peptides are
expected to be present in your subject samples. Each standard
curve is made up in pooled CSF in order that any matrix ion sup-
pression is accounted for. It is also good practice to create quality
controls as well using pooled CSF spiked with your peptide(s) in
order that you can check the sensitivity of the assay at appropriate
intervals (should fall within ±10% of established quantities). It is
recommended to run QC samples at intervals of 10–15 subject
samples. Internal standard values should also be monitored, and
coefficients of variation (%CV) of these should also be used to
monitor assay stability.
3.4 Data Analysis Data analysis uses Waters MassLynx and TargetLynx V4.1 software
to process the raw data. Methods can be set up so that the inte-
grated peaks can be ratioed to the internal standard which can be
yeast enolase in this case or a stable isotope-labelled peptide.
Integration should be checked to confirm accuracy, and then
response values can be interpolated from your standard curve.
4 Conclusion
5 Notes and Troubleshooting
References
or spiked standards. Anal Chem 75(18): risk of Alzheimer’s disease. N Engl J Med
4818–4826 368(2):107–116. https://doi.org/10.1056/
10. Colonna M (2003) TREMs in the immune NEJMoa1211103
system and beyond. Nat Rev Immunol 3(6): 15. Heslegrave A, Heywood W, Paterson R et al
445–453. https://doi.org/10.1038/nri1106 (2016) Increased cerebrospinal fluid soluble
11. Paloneva J, Manninen T, Christman G et al TREM2 concentration in Alzheimer’s disease.
(2002) Mutations in two genes encoding dif- Mol Neurodegener 11:3. https://doi.
ferent subunits of a receptor signaling complex org/10.1186/s13024-016-0071-x
result in an identical disease phenotype. Am 16. Suarez-Calvet M, Araque Caballero MA,
J Hum Genet 71(3):656–662. https://doi. Kleinberger G et al (2016) Early changes in
org/10.1086/342259 CSF sTREM2 in dominantly inherited
12. Kondo T, Takahashi K, Kohara N et al (2002) Alzheimer’s disease occur after amyloid deposi-
Heterogeneity of presenile dementia with bone tion and neuronal injury. Sci Transl Med
cysts (Nasu-Hakola disease): three genetic 8(369):369ra178. https://doi.org/10.1126/
forms. Neurology 59(7):1105–1107 scitranslmed.aag1767
13. Guerreiro R, Wojtas A, Bras J et al (2013) 17. Suarez-Calvet M, Kleinberger G, Araque
TREM2 variants in Alzheimer’s disease. N Caballero MA et al (2016) sTREM2 cerebro-
Engl J Med 368(2):117–127. https://doi. spinal fluid levels are a potential biomarker for
org/10.1056/NEJMoa1211851 microglia activity in early-stage Alzheimer’s
14. Jonsson T, Stefansson H, Steinberg S et al disease and associate with neuronal injury
(2013) Variant of TREM2 associated with the markers. EMBO Mol Med 8(5):466–476.
10.15252/emmm.201506123
Chapter 12
Abstract
Soluble amyloid precursor proteins (sAPP) are under investigation as novel biomarkers of Alzheimer’s
disease (AD) pathophysiology, but protein levels in cerebrospinal fluid do not seem to follow a con-
sistent pattern, which limits their usefulness as diagnostic and prognostic tool. More recently, evi-
dence has been presented for sAPP blood plasma concentration differences between patients with AD
dementia and healthy control subjects. Blood can be easily accessed, which makes it an interesting
target for biomarker discovery. This chapter presents a brief overview of sAPP in blood as a new AD
biomarker candidate. Issues and challenges are discussed, both from a perspective of laboratory assess-
ment and clinical application. A relevant technique for sAPP ascertainment is also provided.
Key words Alzheimer’s disease, Dementia, Blood biomarker, Early diagnosis, Prognosis, Tau,
Amyloid, Mild cognitive impairment
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_12, © Springer Science+Business Media, LLC 2018
179
180 Robert Perneczky and Panagiotis Alexopoulos
Fig. 1 Schematic diagram representing proteolytic cleavage of amyloid precursor protein (reproduced with
permission from: R. Perneczky in [27]). The membrane-bound amyloid precursor protein (APP) is alternatively
cleaved by either α-secretase, resulting in non-amyloidogenic fragments including soluble APP (sAPP)α or by
β-secretase, resulting in sAPPβ and the harmful Aβ fragment via further cleavage by γ-secretase
3 Methods
3.1 General Pre- See [23] for proposed consensus guidelines for the standardization
analytic Guidelines of pre-analytic variables for blood-based biomarker studies in AD
research.
1. Sample withdrawal should only be undertaken by qualified
medical personnel. The identity of the patient should be
checked, and the analysis order/study documentation should
be filled out completely before withdrawal of the sample.
Written informed consent by the patient or study subject must
be available before any procedures are performed. Tubes should
be clearly labelled beforehand and immediately closed after tak-
ing the sample to avoid contamination and evaporation.
2. Samples should optimally be taken after overnight fasting at a
standard time in the morning, before any morning medica-
tions are taken. Closed blood withdrawal systems (e.g.,
Monovette, Vacuette) reduce the preanalytical influence on
the results and the infection risk for medical personnel and are
therefore preferable to syringes.
3. To avoid contamination, tubes without additive should be col-
lected before tubes with additive. When withdrawing several
tubes, the thinning tube should never be at the beginning
(release of tissue factors on puncture).
Soluble APP in Blood 183
3.2 Collection 1. Whole blood samples are collected by venepuncture into com-
of Blood Samples mercially available anticoagulant-treated tubes (e.g., citrate or
for Biomarker Testing EDTA) and put into the freezer at 4 °C within 120 min after
collection.
2. Plasma is isolated from blood cells (see Note 1) by centrifuga-
tion at 2500 × g for 15 min.
3. If the sample is not analyzed immediately, it should be appor-
tioned into aliquots (e.g., 500 μL), and the aliquots should
immediately be transferred to −20 °C and should within
2 weeks be transferred to −80 °C for long-term storage.
3.4 Enzyme-Linked Different methods have been established to quantify protein con-
Immunosorbent Assay centrations in body fluids. Here, ELISA will be discussed as an
(ELISA) example, since it is a widely used method, which has been shown
to be able to detect sAPP in human blood samples. ELISA is usu-
ally performed in duplicate and according to the commercial man-
ufacturer’s instructions (see [22] for an example of a sAPP assay).
An ELISA includes coating, blocking, incubation, and detection
184 Robert Perneczky and Panagiotis Alexopoulos
4 Conclusions
96-well assay. Having said that, there are also several shortcom-
ings, including the relatively large sample volumes needed to run
an ELISA), and restrictions in automatization due to the required
wash steps, even though manual handling of samples may be prone
to failure. A recent study across 14 clinical neurochemistry labora-
tories in Europe (Germany, Austria, and Switzerland) reported
that the coefficient of variation CV (CV) of ELISAs was in the
20–30% range when using commercially available ELISA kits for
AD diagnosis [24].
From reviewing the available literature on sAPP as a novel AD
biomarker, two main conclusions arise. First, concentration
changes of sAPPα/sAPPβ in CSF do not seem to follow a consis-
tent pattern limiting their usefulness as diagnostic markers.
Secondly, initial results relating to sAPPβ in blood are encourag-
ing, but replication and further validation are required before firm
conclusions can be drawn. Some evidence exists that a combina-
tion of the new and established biomarkers may improve the over-
all diagnostic accuracy [10], but more research in this field is
needed. There also remains a need to standardize laboratory pro-
cedures to generate reliable results across laboratories in multi-
center studies. Several quality control initiates have been launched
worldwide to establish standardized protocols for biomarker assess-
ment, including the Alzheimer’s Association Global Biomarkers
Consortium [25].
5 Notes
References
1. Jack CR Jr, Albert MS, Knopman DS et al age.2007.09.020. S1053-8119(07)00806-3
(2011) Introduction to the recommenda- [pii]
tions from the National Institute on Aging- 9. Scholl M, Lockhart SN, Schonhaut DR
Alzheimer’s Association workgroups on et al (2016) PET imaging of Tau deposi-
diagnostic guidelines for Alzheimer’s disease. tion in the aging human brain. Neuron
Alzheimers Dement 7(3):257–262. https:// 89(5):971–982. https://doi.org/10.1016/j.
doi.org/10.1016/j.jalz.2011.03.004. S1552- neuron.2016.01.028
5260(11)00100-2 [pii] 10. Perneczky R, Tsolakidou A, Arnold A et al
2. Lopez OL (2013) Mild cognitive impair- (2011) CSF soluble amyloid precursor pro-
ment. Continuum (Minneap Minn) teins in the diagnosis of incipient Alzheimer
19(2 Dementia):411–424. https://doi. disease. Neurology 77(1):35–38. https://doi.
org/10.1212/01.CON.0000429175.29601.97 org/10.1212/WNL.0b013e318221ad47
3. McKhann GM, Knopman DS, Chertkow 11. Chan K, Lucas D, Hise D et al (2004)
H et al (2011) The diagnosis of dementia Analysis of the human serum proteome. Clin
due to Alzheimer’s disease: recommenda- Proteomics 1:101–226
tions from the National Institute on Aging- 12. Wyss-Coray T (2006) Inflammation in
Alzheimer’s Association workgroups on Alzheimer disease: driving force, bystander
diagnostic guidelines for Alzheimer’s disease. or beneficial response? Nat Med 12(9):1005–
Alzheimers Dement 7(3):263–269. https:// 1015. https://doi.org/10.1038/nm1484.
doi.org/10.1016/j.jalz.2011.03.005. S1552- nm1484 [pii]
5260(11)00101-4 [pii]
13. Ray S, Britschgi M, Herbert C et al (2007)
4. McKhann GM (2011) Changing concepts Classification and prediction of clinical
of Alzheimer disease. JAMA 305(23):2458– Alzheimer’s diagnosis based on plasma signal-
2459. https://doi.org/10.1001/jama.2011. ing proteins. Nat Med 13(11):1359–1362.
810.305/23/2458 [pii] https://doi.org/10.1038/nm1653
5. Giaccone G, Arzberger T, Alafuzoff I et al 14. Zhang H, Ma Q, Zhang YW et al (2012)
(2011) New lexicon and criteria for the Proteolytic processing of Alzheimer’s beta-
diagnosis of Alzheimer’s disease. Lancet amyloid precursor protein. J Neurochem
Neurol 10(4):298–299.; author reply 120(Suppl 1):9–21. https://doi.
300–301. https://doi.org/10.1016/ org/10.1111/j.1471-4159.2011.07519.x
S1474-4422(11)70055-2
15. Wu G, Sankaranarayanan S, Wong J et al (2012)
6. Blennow K, Hampel H, Weiner M et al (2010) Characterization of plasma beta-secretase
Cerebrospinal fluid and plasma biomark- (BACE1) activity and soluble amyloid precursor
ers in Alzheimer disease. Nat Rev Neurol proteins as potential biomarkers for Alzheimer’s
6(3):131–144. https://doi.org/10.1038/ disease. J Neurosci Res 90(12):2247–2258.
nrneurol.2010.4 https://doi.org/10.1002/jnr.23122
7. Frisoni GB, Fox NC, Jack CR Jr et al (2010) 16. Lewczuk P, Popp J, Lelental N et al (2012)
The clinical use of structural MRI in Alzheimer Cerebrospinal fluid soluble amyloid-beta pro-
disease. Nat Rev Neurol 6(2):67–77. https:// tein precursor as a potential novel biomark-
doi.org/10.1038/nrneurol.2009.215 ers of Alzheimer’s disease. J Alzheimers Dis
8. Drzezga A, Grimmer T, Henriksen G et al 28(1):119–125. https://doi.org/10.3233/
(2008) Imaging of amyloid plaques and cerebral JAD-2011-110857
glucose metabolism in semantic dementia and 17. Rosen C, Andreasson U, Mattsson N et al
Alzheimer’s disease. NeuroImage 39(2):619– (2012) Cerebrospinal fluid profiles of amyloid
633. https://doi.org/10.1016/j.neuroim-
Soluble APP in Blood 187
Abstract
Aggregated tau is a major neuropathological protein implicated in the pathophysiology of common
neurodegenerative disorders, such as Alzheimer’s disease (AD), Parkinson’s disease without (PD) and
with later dementia (PDD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and cortico-
basal degeneration (CBD). Aggregated tau tangles, the result of hyperphosphorylation, is a pathological
characteristic of a group of neurodegenerative conditions known as the tauopathies. In AD, it has been
shown that the density of tau tangles closely correlates with neuronal dysfunction and cell death, which
is not the case for β-amyloid. Until now, diagnostic and pathological demonstration of tau deposition
has only been possible by invasive techniques such as brain biopsy or postmortem examination. The
recent advances in the development of selective tau positron emission tomography (PET) tracers have
allowed in vivo investigation of the presence and extent of tau pathology in patients suspected of having
tauopathies and the role of tau in the early phases of neurodegenerative diseases. In this review, the role
of aggregated tau will be discussed, as well as the challenges posed by, and the current status of, the
development of selective tau tracers as biomarkers, and the new clinical information that has been
uncovered, in addition to the opportunities for refining the diagnosis of tauopathies in the future.
Key words Tau imaging, Dementia, Neurodegenerative diseases, PET, Alzheimer’s disease
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_13, © Springer Science+Business Media, LLC 2018
189
190 Paul Edison
3.1 [18F]T807 [18F]T807 (Flortaucipar) was originally tested in vivo and demon-
(AV-1451) strated high binding affinity and selectivity [23]. [18F]T807 dem-
onstrated high specificity, with 25-fold selectivity for PHF-tau over
Aβ, with rapid brain penetration and washout, and no active
metabolites entering the brain [24]. While [18F]T807 was found
Tau Imaging in AD 193
O
18F OH OH
O 18F O
N 18F
H
N N N
H CH3 CH3
N N
CH3 CH3
(d) [18F]THK-5351
(e) [11C]PBB3
H OH N
F O
S H
HO
N N
11CH3
CH3
N N
(f) [18F]T808
(g) [18F]T807
H
N N
N N N
N
N H
N 18F
18F
Fig. 1 Chemical structures of tau tracers in current use. (a) [18F]THK-523, (b) [18F]THK-5105, (c) [18F]THK-
5117, (d) [18F]THK-5351, (e) [11C]PBB3, (f) [18F]T808, and (g) [18F]T807
Fig. 2 [18F]AV-1451 in AD, MCI, and control subjects. Top panel demonstrates transverse section of tau images
using [18F]AV1451 in Alzheimer’s subjects (a), mild cognitive impairment subjects (b), and healthy controls (c).
Bottom panel demonstrates the coronal sections for the corresponding subjects
[29, 30]. Other conditions where tau [31] is primarily driving the
disease process like FTD, chronic traumatic encephalopathy (CTE),
presents with different pattern of deposition of tau [32]. In cogni-
tively normal elderly subjects, [18F]T807 binding has been shown
to correlate with levels of CSF tau [33].
3.2 THK Compounds Okamura and colleagues reported first [18F]THK compounds,
of which [18F]THK523 was evaluated at the Tohuku University
[34]. Pharmacokinetic studies showed excellent uptake and
clearance in the brains of mice [35], and in vivo studies demon-
strated significant uptake. While the uptake correlated well with
cognitive function, there was significant uptake in white matter
[36] making the tracer less favorable for clinical use. Two closely
related compounds, [18F]THK5105 and [18F]THK5117,
were then evaluated. In vivo studies showed that there was
higher cortical retention in AD patients compared with healthy
controls, and this correlated well with cognitive test results and
brain volumes [37]. [18F]THK5117 uptake in AD and MCI
subjects correlated well with cognitive performance, and there
Tau Imaging in AD 195
3.4 Other Gobbi et al. have developed further high affinity binders,
Compounds RO6931643, RO6924963, and RO6958948. These compounds
bind with high affinity and specificity to the [3H]T808 binding
site on tau aggregates [42]. Importantly, they lack affinity for Aβ
plaques and showed low nonspecific binding to healthy brain tis-
sue. Additionally, the compounds demonstrate macro- and micro-
colocalization of radioligand binding, rapid brain entry, and
washout with well-tolerated metabolic patterns. These tracers are
currently in clinical testing.
4 Conclusions
Tau imaging will allow us to visualize and quantify the tau aggrega-
tion in neurodegenerative diseases and is able to provide us with
further information about the neuropathology of the disease pro-
cess. This could revolutionize the field, as we are now able to
understand more about the disease trajectory, and test different
196 Paul Edison
References
1. Spillantini MG, Goedert M (2013) Tau pathol- 12. Gotz J, Chen F, van Dorpe J, Nitsch RM
ogy and neurodegeneration. Lancet Neurol (2001) Formation of neurofibrillary tangles in
12(6):609–622. https://doi.org/10.1016/ P301l tau transgenic mice induced by Abeta
s1474-4422(13)70090-5 42 fibrils. Science 293(5534):1491–1495.
2. Lee HG, Perry G, Moreira PI et al (2005) https://doi.org/10.1126/science.1062097
Tau phosphorylation in Alzheimer’s disease: 13. Lewis J, Dickson DW, Lin WL et al (2001)
pathogen or protector? Trends Mol Med Enhanced neurofibrillary degeneration in
11(4):164–169. https://doi.org/10.1016/j. transgenic mice expressing mutant tau and
molmed.2005.02.008 APP. Science 293(5534):1487–1491. https://
3. Giacobini E, Gold G (2013) Alzheimer disease doi.org/10.1126/science.1058189
therapy—moving from amyloid-beta to tau. 14. Gandy S, Ikonomovic M, Mitsis E, Elder G,
Nat Rev Neurol 9(12):677–686. https://doi. Ahlers S, Barth J, Stone J, DeKosky ST (2014)
org/10.1038/nrneurol.2013.223 Chronic traumatic encephalopathy: clinical-
4. Grundke-Iqbal I, Iqbal K, Tung Y-C, Quinlan biomarkers correlations and current concepts
M, Wisniewsk H, Binder L (1986) Abnormal in pathogenesis. Mol Degener 9:37
phosphorylation of the microtubule-asso- 15. Villemagne VL, Pike KE, Chetelat G et al
ciated protein tau in Alzheimer cytoskel- (2011) Longitudinal assessment of Abeta and
etal pathology. Proc Natl Acad Sci U S A cognition in aging and Alzheimer disease.
83:4913–4917 Ann Neurol 69(1):181–192. https://doi.
5. Kosik K, Joachim C, Selkoe D (1986) org/10.1002/ana.22248
Microtubule-associated protein tau is a major 16. Villemagne VL, Burnham S, Bourgeat P et al
antigenic component of paired helical filaments (2013) Amyloid B deposition, neurodegen-
in Alzheimer disease. Proc Natl Acad Sci U S A eration, and cognitive decline in sporadic
83:4044–4048 Alzheimer’s disease: a prospective cohort
6. Braak H, Braak E (1991) Neuropathological study. Lancet Neurol 12:357–367. https://
staging of Alzheimer related changes in doi.org/10.1016/S1474-4422(13)70044-9
Alzheimer’s disease. Acta Neuropathol 17. Laruelle M, Slifstein M, Huang Y (2003)
82:239–259 Relationships between radiotracer properties
7. Braak H, Braak E (1997) Frequency of stages and image quality in molecular imaging of the
of Alzheimer-related lesions in different age brain with positron emission tomography. Mol
categories. Neurobiol Aging 18(4):351–357 Imaging Biol 5(6):363–375
8. Becker JA, Hedden T, Carmasin J et al 18. Villemagne VL, Furomoto S, Fodero-Tavoletto
(2011) Amyloid-beta associated cortical thin- M et al (2012) The challenges of tau imaging.
ning in clinically normal elderly. Ann Neurol Future Neurol 7(4):409–421
69(6):1032–1042. https://doi.org/10.1002/ 19. Villemagne VL, Okamura N (2014) In vivo tau
ana.22333 imaging: obstacles and progress. Alzheimers
9. Engler H, Forsby A, Almqvist O et al (2006) Dement 10(3 Suppl):S254–S264. https://doi.
Two year follow up of amyloid deposition org/10.1016/j.jalz.2014.04.013
in patients with Alzheimer’s disease. Brain 20. Okamura N, Harada R, Furumoto S et al (2014)
129:2856–2866 Tau PET imaging in Alzheimer’s disease. Curr
10. Gómez-Isla T, Hollister R, West H et al (1997) Neurol Neurosci Rep 14(11):500. https://
Neuronal loss correlates with but exceeds neu- doi.org/10.1007/s11910-014-0500-6
rofibrillary tangles in Alzheimer’s disease. Ann 21. Shah M, Catafau AM (2014) Molecular
Neurol 41:17–24 imaging insights into neurodegeneration:
11. Ingelsson M, Fukumoto H, Newell KL et al focus on tau PET radiotracers. J Nucl Med
(2004) Early amyloid accumulation and pro- 55(6):871–874. https://doi.org/10.2967/
gressive synaptic loss, gliosis and tangle for- jnumed.113.136069
mation in the Alzheimer’s disease brain. 22. Villemagne V, Fodero-Tavoletti M, Masters
Neurology 62:925–931 CL, Rowe C (2015) Tau imaging: early
Tau Imaging in AD 197
progress and future directions. Lancet Neurol 32. Dickerson B, McGinnis S, Gomperts S et al.
14(1):114–124 (2015) [18F]T807 of frontotemporal lobar
23. Gao M, Wang M, Zheng QH (2014) Concise degeneration. Human Amyloid Imaging
and high-yield synthesis of T808 and T808P Conference Book of Abstracts 2015 P15
for radiosynthesis of [(18)F]-T808, a PET tau Submission ID 61:37
tracer for Alzheimer’s disease. Bioorg Med 33. Chhatwal J, Schultz A, Marshall G et al. (2015)
Chem Lett 24(1):254–257. https://doi. Entorhinal, parahippocampal, and inferior tem-
org/10.1016/j.bmcl.2013.11.025 poral F18-T807 SUVR correlates with CSF
24. Xia CF, Arteaga J, Chen G et al (2013) [(18) total tau and tau T181P in cognitively normal
F]T807, a novel tau positron emission tomog- elderly. Human Amyloid Imaging Conference
raphy imaging agent for Alzheimer’s disease. Book of Abstracts 2015 Submission ID 115:88
Alzheimers Dement 9(6):666–676. https:// 34. Okamura N, Suemoto T, Furumoto S et al
doi.org/10.1016/j.jalz.2012.11.008 (2005) Quinoline and benzimidazole deriva-
25. Chien DT, Bahri S, Szardenings AK et al tives: candiate probes for in vivo imaging of tau
(2013) Early clinical PET imaging results with pathology in Alzheimer’s disease. J Neurosci
the novel PHF-tau radioligand [F-18]-T807. 25(47):10857–10862
J Alzheimers Dis 34(2):457–468. https://doi. 35. Harada R, Okamura N, Furumoto S et al
org/10.3233/JAD-122059 (2013) Comparison of the binding characteris-
26. Pontecorvo M, Devous Ml, Joshi A, Lu M, tics of [18F]THK523 and other amyloid imag-
Siderowf A, Arora A, Mintun M (2015) ing tracers to Alzheimer’s disease pathology.
Relationships between florbetapir PET amy- Eur J Nucl Med Mol Imaging 40(1):125–132
loid and 18F AV-1451 (aka 18F-T807) PET 36. Villemagne VL, Furumoto S, Fodero-Tavoletti
tau binding in cognitively normal subjects and MT et al (2014) In vivo evaluation of a novel
patients with cognitive impairments suspected tau imaging tracer for Alzheimer’s disease. Eur
of Alzheimer’s disease. Human Amyloid J Nucl Med Mol Imaging 41(5):816–826
Imaging Conference Book of Abstracts 2015 37. Okamura N, Furumoto S, Fodero-Tavoletti
ID submission 98:131 MT et al (2014) Non-invasive assessment of
27. Johnson K, Becker J, Sepulcre J et al. (2014) Alzheimer’s disease neurofibrillary pathol-
Tau PET: initial experience with F18 T807. ogy using 18F-THK5105 PET. Brain 137(Pt
Human Amyloid Imaging Conference Book of 6):1762–1771. https://doi.org/10.1093/
Abstracts 2014 ID submission 103:22 brain/awu064
28. Mintun M, Devous M, Pontecorvo M, Joshi 38. Saint-Aubert L, Lemoine L, Marutle A et al.
A, Siderowf A, Johnson K, Navitsky M, Lu (2015) Relationship between post-mortem
M (2015) Potential for PET imaging tau THK5117 binding and in-vivo PET biomarkers
tracer 18F-AV-1451 (also known as 18F- uptake in Alzheimer’s disease. Human Amyloid
T807) to detect neurodegenerative progres- Imaging Conference Book of Abstracts 2015
sion in Alzheimer’s disease. Human Amyloid ID submission 94:94
Imaging Conference Book of Abstracts 2015 39. Harada R, Okamura N, Furumoto S et al.
Submission ID 95:87 (2015) First-in-human PET study of a novel
29. Ossenkoppele R, Schonhaut D, Baker S et al. tau tracer [18F]THK5351. Human Amyloid
(2015) Distinct [18F]AV1451 retention pat- Imaging Conference Book of Abstracts 2015
terns in clinical variants of Alzheimer’s disease. Submission ID 24
Human Amyloid Imaging Conference Book of 40. Maruyama M, Shimada H, Suhara T et al
Abstracts 2015 P55 Submission 113:62 (2013) Imaging of tau pathology in a
30. Marquie M, Normandin M, Vanderburg C tauopathy mouse model and in Alzheimer
et al. (2015) Towards the validation of novel patients compared to normal controls.
PET tracer T807 on postmortem human Neuron 79(6):1094–1108. https://doi.
brain tissue samples. Abstract Online: Human org/10.1016/j.neuron.2013.07.037
Amyloid Imaging Book of Abstracts 2015 41. Shimada H, Higuchi M, Ikoma Y et al
Submission ID 89:28 (2013) In vivo visualization of tau pathol-
31. Mitsis EM, Riggio S, Kostakoglu L et al (2014) ogy. Alzheimers Dement 9(4):P101–P102.
Tauopathy PET and amyloid PET in the diag- https://doi.org/10.1016/j.jalz.2013.05.172
nosis of chronic traumatic encephalopathies: 42. Gobbi LC, Knust H, Körner M et al (2017)
studies of a retired NFL player and of a man Identification of three novel radiotracers for
with FTD and a severe head injury. Transl imaging aggregated tau in Alzheimer’s disease
Psychiatry 4:e441. https://doi.org/10.1038/ with positron emission tomography. J Med
tp.2014.91 Chem 60(17):7350–7370
Chapter 14
Abstract
Changes in the brain that lead to Alzheimer’s disease are thought to start decades before cognitive symp-
toms emerge. If biomarkers for these early stages could be identified, it would contribute to a more accu-
rate estimation of an individual’s risk of developing disease and enable the monitoring of high-risk
(presymptomatic) persons as well as providing the means for assessing the efficacy of new interventions.
The retina links to the visual processing and cognitive centers of the brain, but it is also an extension of the
brain sharing embryological origins as well as a blood supply and nerve tissue. It therefore has huge poten-
tial as a site for biomarker investigation through easy, noninvasive imaging and computational image analy-
sis to reveal valuable information about microvascular health, deposition, and neurodegenerative damage.
Capturing reliable longitudinal data pertaining to the onset of Alzheimer’s disease is a key target, but a
high degree of standardization is necessary if the potential of the retina is to be fully realized. Our goal is
to provide the reader with guidelines on how to execute robust retinal imaging and analysis for neuroreti-
nal biomarker discovery and to highlight advantages and limitations of the techniques.
Key words Alzheimer’s disease, Dementia, Retinal imaging, Non-invasive, Biomarker, Fundus,
Blood vessel, Neurodegeneration
1 Introduction
The retina is the light-sensing inner surface of the human eye and is
comprised of layers of tissue consisting of neurons and supporting
cells, interconnected by synapses (see Fig. 1). The retinal ganglion
cell (RGC) layer, the innermost cellular layer, projects its axons
across the inner retina, called the retinal nerve fiber layer (RNFL),
through the optic nerve to the visual processing and cognitive cen-
ters of the brain. Developmentally and anatomically, the retina is an
extension of the brain sharing embryonic origins as well as features,
besides nerve tissue, such as small blood vessels and a blood-tissue
barrier [1]. It also has the advantage of being easier to image than
the brain and with far superior resolution—resolvable detail in
images of the eye being at a micron level compared to millimeters
with conventional magnetic resonance (MR) imaging. The retina is
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_14, © Springer Science+Business Media, LLC 2018
199
200 Tom MacGillivray et al.
Fig. 1 Anatomy of the human eye [26]. (a) Diagram showing the main structures. (b) Illustration of the organi-
zation of the retinal layers and cells
2 Materials
2.1 Setup Prior Table 1 gives a summary of the principal retinal imaging modalities we
to Imaging use for biomarker discovery in AD. Retinal imaging is a patient-friendly
tool that is noninvasive, quick, and easy to perform. Most people are
familiar with the typical imaging procedure, as many of them will have
attended the high street optometrist for routine eye health checks. For
each instrument (see Note 1), the person being imaged is asked to sit
with his or her chin on a rest and with their forehead placed firmly
against a stabilizing bar (see Note 2). Before proceeding, the instru-
ment operator checks that the person is comfortable, adjusting the
height of the instrument table if necessary. The operator also adjusts
the headrest frame to ensure that the individual’s eye is in line with the
height adjustment mark (usually a horizontal white line). The person is
202 Tom MacGillivray et al.
Table 1
A summary of the principal retinal imaging modalities
then asked to look at a fixation light (see Note 3) while the operator
focuses and aligns the system before instigating the image acquisition,
which involves the instrument illuminating the retina with its light
source and then capturing the reflected light.
We image both eyes (see Note 4) with each of the devices and
without administering drops for dilation (see Note 5). Conducting
several imaging protocols one after the other can see an individual
become fatigued resulting in a decline of image quality. While pupil
dilation might help mitigate this effect, the ability to conduct
imaging without it amounts to a real advantage as administering
eye drops adds considerable complexity to the protocols. For
instance, individuals must have their intraocular pressure and ante-
rior chamber (the fluid-filled space inside the eye between the iris
and the cornea) assessed as a precaution for acute angle-closure
glaucoma, which is more common in older people. There is also a
restriction on driving and a sensitivity to bright light, which extends
for several hours after administering drops.
Retinal Imaging in Early Alzheimer’s Disease 203
2.2 Fundus Imaging Fundus imaging generates a 2D image of the retina directly behind
the pupil. A fundus camera achieves this with a specialized low-
power microscope, an attached digital camera, and a flash of white
light for illumination, which creates a color photograph of the ret-
ina. We use a Canon CR-DGi (Canon USA Inc., Lake Success, NY)
to image an area showing the optic disk (OD) or optic nerve head,
macula, and blood vessels (see Fig. 2a). We standardize by center-
ing the image acquisition midway between the centers of the OD
and macula to give a digital fundus photograph suitable for quanti-
tative analysis of key vascular and structural features. Repeating the
same acquisition when capturing further images within or between
individuals reduces uncertainties when comparing measurements.
A scanning laser ophthalmoscope (SLO) such as in the
Heidelberg SPECTRALIS (Heidelberg Engineering, Dossenheim,
Germany) uses an infrared (IR) laser beam and confocal micro-
scope arrangement to scan across the surface of the retina and form
a fundus picture. This particular SLO images at a slightly higher
resolution than the fundus camera but captures a smaller area (see
Table 1; Fig. 2b). The main purpose of the SLO imaging in the
SPECTRALIS is to guide the operator to the desired location for
OCT scanning and to enable accurate visit-to-visit registration for
repeat imaging sessions. Yet, the clarity and resolution of the SLO
images lend themselves to additional computational analysis of the
retinal vasculature, especially the larger vessels where small changes
or abnormalities might be more feasible to detect than with the
fundus camera. We center imaging on the optic nerve head.
To image more of the retina surface in a single capture, an ultra-
widefield SLO (UWF-SLO) employs scanning mirrors to direct laser
light to regions of the back of the eye that are inaccessible with a
fundus camera or standard SLO. We use an Optos Daytona (Optos
204 Tom MacGillivray et al.
Fig. 2 Analyzing the fundus. (a) Color fundus camera image of a right eye showing the region containing the
OD, macula, and blood vessels. (b) SLO imaging of the same eye centered on the OD and acquired with an IR
light source, which generates a grayscale image. Green circle denotes standard positioning of peripapillary
OCT acquisition around the optic nerve head. (c) UWF-SLO view centered on the macula, imaging a larger
region of the retina than the fundus camera (dashed yellow circle) and SLO (dashed yellow square). The stan-
dard positioning of OCT-A acquisitions centered on the OD and macula is also denoted (solid magenta squares).
(d–f) Retinal blood vessels automatically detected by computerized image processing techniques in each of
the three fundus imaging modalities. (g) SLO image overlaid with the standard measurement zone B (dashed
cyan circles) used in the analysis of blood vessel widths. The six largest arterioles (red) and venules (blue) and
their individual widths (yellow) are used to calculate CRAE, CRVE, and AVR. (h) Generalized dimension spec-
trum (Dq) calculated for the vasculature detected in the fundus camera image and used to quantify the com-
plexity/sparsity of the vascular branching pattern. For each value of q, the calculation of Dq from the detected
vessels is repeated a number of times to give a mean value and associated error. (i) Width measurements
(black dots) performed along the path of a vessel extracted from the UWF-SLO image with straight line fit
(dashed black line) to quantify tapering as the vessel heads out toward the periphery
Retinal Imaging in Early Alzheimer’s Disease 205
2.3 Optical OCT enables cross-sectional imaging of the internal retinal structures
Coherence (with a limited tissue depth <2 mm). An optical beam directed at the
Tomography retina and interferometry to resolve the back-scattered light signals
and Angiography and determine the depth of reflecting structures is an axial scan or
A-scan. Scanning the beam across the retina to acquire numerous
A-scans builds up the 2D image. The Heidelberg SPECTRALIS uses
spectral domain OCT (SD-OCT) in which the depth information is
computed by analyzing the interference signal based on the wave-
length of returning light. We capture a circular peripapillary OCT
scan in order to measure changes in structure around the OD, par-
ticularly in RNFL thickness (see Fig. 3a–d). In addition, we capture a
volume scan centered on the macula to measure retinal and RGCL
layer thickness. The RTVue XR Avanti (Optovue, Inc., Fremont,
CA), also a SD-OCT, maps RNFL and GCC layer thickness with an
OD-centered OCT acquisition (see Fig. 3e, f).
The RTVue XR Avanti has an angiographic mode that reveals
the very small vessels at different depths in the retina. We perform
Fig. 3 Analyzing the retinal layers. (a) SLO image focused on OD, showing the location of the peripapillary OCT
scan acquisition (green circle). (b) OCT image showing the retinal layers and delineation of the ILM (red line) and
RNFL border (cyan line), which gives RNFL thickness. (c) Evaluating RNFL thickness using the standard TSNIT
map which subdivides the measurements and color codes them compared to normative values. (d) RNFL thick-
ness profile (black line) with color coding emphasizing statistical significance in comparison to normal limits—
green indicates within normal limits; blue, borderline above; yellow, borderline below; and red, below. The person
imaged here shows mostly green, whereas we might expect more yellow and red for a person with AD progres-
sion. (e) Map of RNFL thickness around the OD and (f) GCC thickness, also color coded to emphasize the presence
of any abnormally thin areas. Again, in AD progression, thinner or more red areas might be anticipated
206 Tom MacGillivray et al.
Fig. 4 Analyzing the retina using OCT-A. (a) Nerve head segment and (b) radial peripapillary capillary (RPC)
segment and (c) superficial vascular plexus and (d) deep vascular plexus region in the macula. Measuring
retinal thickness from OCT and vessels density from OCT-A using a TSNIT map (blue zones) in the superficial
vascular plexus of (e) a normal eye and (f) an eye of individual who has suffered a stroke (and so thought to
have deteriorated vascular brain health). (g–h) Color coding of vessel density measurements performed using
a 3 × 3 grid (white dashed lines) to subdivide the superficial vascular plexus into squares. The individual
believed to have poor vascular brain health exhibits a lower vessel density
3 Image Analysis
3.1 Overview We utilize expert human grading to detect and classify retinopathy
(i.e., type, number, size, and location), including drusen, in our
retinal imaging. Computerized software tools that exploit image
processing and analysis techniques enable further quantitative and
reproducible measurements of structural features such as the vas-
culature visible in fundus images and the different retinal layers in
OCT [14, 15]. Such computer-assisted methods measure varia-
tions or abnormalities in the retina that might be imperceptible to
or missed by a human grader, in an objective and repeatable
Retinal Imaging in Early Alzheimer’s Disease 207
3.2 Fundus Imaging Our group has developed software (VAMPIRE; Vascular Assessment
(See Fig. 2) and Measurement Platform for Images of the REtina, Universities of
Edinburgh and Dundee, UK) to extract parametric information
from fundus imaging [16–18]. Retinal vessel segmentation is the
precursor to the measurement of features such as vessel width, tor-
tuosity, and complexity of the vascular network [19]. We perform
this for fundus camera, SLO, and UWF-SLO images (see Fig. 2d–f).
Additional semiautomatic classification distinguishes venules from
arterioles, removes non-vessel artifacts, and detects the center and
boundary of the OD along with center of the macula. The operator
can correct any of these labels through a user interface that allows
them to interact with an image being analyzed.
VAMPIRE software places the standard set of circular
measurement zones on a fundus camera or SLO image—Zone B is
a ring 0.5–1 OD diameters away from the center, and Zone C is a
ring extending from OD boundary to 2 OD diameters away.
Measurements of the widths of the six widest arterioles and venules
crossing Zone B (see Fig. 2g) are used to calculate the central retinal
arteriolar equivalent (CRAE) and central retinal venular e quivalent
(CRVE) and give the arteriole to venule ratio (AVR; CRAE/
CRVE) [20]. Similarly, the tortuosity (i.e., how much a vessel
twists and turns) of the six widest arterioles and venules crossing
Zone C is measured [21] and then further summarized by calculat-
ing median arteriolar and venular values along with the standard
deviations and ranges.
The complexity (or sparsity) of the vascular branching pattern
visible with fundus imaging can be quantified using fractal analysis to
yield a fractal dimension that combines contributions of individual
vessel parameters into a single global value [14]. Variations in the
fractal dimension are an indicator of deviation away from normative
branching arrangements and so a potential marker of disease [22].
We use a particular approach called multifractal analysis, which gen-
erates several fractal dimensions at different “scales” (see Fig. 2h),
and we record dimensions D0, D1, and D2 as these have previously
been reported as likely sensitive markers of small vascular changes
[23]. Application of the analysis is to vessels appearing in Zone C
and separated into arteriolar and venular measurements.
With UWF-SLO, it is not only possible to assess retinopathy and
the vasculature in the peripheral retina but to explore novel param-
eters for biomarker discovery in addition to conventional Zone B–C
measurements (see Note 7). For instance, we can analyze the main
vessels over a longer path distance than is achievable with other fun-
dus modalities and look at how the width changes as the vessel
extends out toward the periphery (see Fig. 2i). An abnormal decline
in the vessel’s width (quantified by fitting a model such as a straight
line) may indicate an unhealthy vessel.
208 Tom MacGillivray et al.
3.3 Optical The major task in analyzing OCT is accurate segmentation of reti-
Coherence nal layers as thickness changes are an important indicator of disease
Tomography status. Modern OCT instruments such as the Heidelberg
and Angiography SPECTRALIS and RTVue XR Avanti are equipped with software
(See Fig. 3) to automatically delineate the borders of the internal limiting
membrane (ILM) and the RNFL, as the distance between these
two boundaries gives the measurement of RNFL thickness (see
Fig. 3b). Thickness of the RNFL is evaluated using the standard
TSNIT (temporal, superior, nasal, inferior, temporal) mapping
approach which subdivides or regionalizes the measurement and
color codes statistical significance compared to a database of nor-
mative values (see Fig. 3c). RNFL thickness also presented as a
profile plot with color coding again underlining statistical signifi-
cance in comparison to normal limits (see Fig. 3d) or as a thickness
map (see Fig. 3e). Further delineation of boundaries in OCT
enables mapping of other layers such as the GCC (see Fig. 3f).
While these quantitative OCT mapping options present exciting
opportunities for biomarker discovery, the pathological processes
behind retinal thinning in AD are incompletely understood, and fur-
ther investigations are required to determine the use of retinal OCT
as a potential surrogate marker of neurodegeneration (Fig. 4).
OCT-A works by comparing consecutive scans at the same loca-
tion to detect flow using motion contrast and enable demarcation of
blood vessels. As a means of quantifying OCT-A, the instrument calcu-
lates vessel density parameters, i.e., the percentage area occupied by the
larger appearing vessels and the microvasculature in particular regions.
This can be the entire scan area as well as standard regions such as
TSNIT sectors or a grid-based division of the image (see Fig. 4e–g).
For each scanned region, the software calculates the vessel densities
and thicknesses in various layers of the retina and optic nerve head.
OCT-A offers potentially exciting avenues to explore in the
context of neuroretinal biomarker discovery, particularly around
small vessels and AD. However, caution is urged in the interpreta-
tion and analysis of OCT-A imaging. The modality detects flow
above a threshold level, and so not all vessels may appear in the
images such as blocked or leaky ones. In addition, it is more suscep-
tible to image artifact caused by patient motion or reflections from
internal retinal structures that may not always be obvious to the
untrained eye or computer algorithm.
Retinal Imaging in Early Alzheimer’s Disease 209
4 Conclusions
5 Notes
Acknowledgments
References
1. London A, Benhar I, Schwartz M (2013) The 2:24. https://doi.org/10.1186/
retina as a window to the brain-from eye research s40942-016-0049-4
to CNS disorders. Nat Rev Neurol 9(1):44–53. 12. Lisboa R, Paranhos A Jr, Weinreb RN et al
https://doi.org/10.1038/nrneurol.2012.227 (2013) Comparison of different spectral
2. Knopman DS (2006) Dementia and cerebro- domain OCT scanning protocols for diagnos-
vascular disease. Mayo Clin Proc 81(2):223– ing preperimetric glaucoma. Invest Ophthalmol
230. https://doi.org/10.4065/81.2.223 Vis Sci 54(5):3417–3425. https://doi.
3. Reitz C, Brayne C, Mayeux R (2011) org/10.1167/iovs.13-11676
Epidemiology of Alzheimer disease. Nat Rev 13. de Carlo TE, Romano A, Waheed NK et al (2015)
Neurol 7(3):137–152. https://doi. A review of optical coherence tomography angi-
org/10.1038/nrneurol.2011.2 ography (OCTA). Int J Retina Vitreous 1:5.
4. Patton N, Aslam T, Macgillivray T et al (2005) https://doi.org/10.1186/s40942-015-0005-8
Retinal vascular image analysis as a potential 14. Patton N, Aslam TM, MacGillivray T et al
screening tool for cerebrovascular disease: a ratio- (2006) Retinal image analysis: concepts, appli-
nale based on homology between cerebral and cations and potential. Prog Retin Eye Res
retinal microvasculatures. J Anat 206(4):319– 25(1):99–127. https://doi.org/10.1016/j.
348. https://doi. preteyeres.2005.07.001
org/10.1111/j.1469-7580.2005.00395.x 15. Abramoff MD, Garvin MK, Sonka M (2010)
5. McGrory S, Cameron JR, Pellegrini E et al Retinal imaging and image analysis. IEEE
(2017) The application of retinal fundus cam- Trans Med Imaging 3:169–208. https://doi.
era imaging in dementia: a systematic review. org/10.1109/RBME.2010.2084567
Alzheimers Dement (Amst) 6:91–107. https:// 16. MacGillivray T (2012) VAMPIRE: vessel
doi.org/10.1016/j.dadm.2016.11.001 assessment and measurement platform for
6. Ratnayaka JA, Serpell LC, Lotery AJ (2015) images of the retina. In: Tan EYKNJH, Acharya
Dementia of the eye: the role of amyloid beta in UR, Suri JS (eds) Human eye imaging and
retinal degeneration. Eye (Lond) 29(8):1013– modeling. CRC Press, Boca Raton, FL
1026. https://doi.org/10.1038/eye.2015.100 17. Cameron JR, Ballerini L, Langan C et al (2016)
7. Koronyo-Hamaoui M, Koronyo Y, Ljubimov Modulation of retinal image vasculature analysis
AV et al (2011) Identification of amyloid to extend utility and provide secondary value
plaques in retinas from Alzheimer’s patients and from optical coherence tomography imaging.
noninvasive in vivo optical imaging of retinal J Med Imaging (Bellingham) 3(2):020501.
plaques in a mouse model. Neuroimage https://doi.org/10.1117/1.JMI.3.2.020501
54(Suppl 1):S204–S217. https://doi. 18. Pellegrini E, Robertson G, Trucco E et al
org/10.1016/j.neuroimage.2010.06.020 (2014) Blood vessel segmentation and width
8. Anderson DH, Talaga KC, Rivest AJ et al estimation in ultra-wide field scanning laser
(2004) Characterization of beta amyloid ophthalmoscopy. Biomed Opt Express
assemblies in drusen: the deposits associated 5(12):4329–4337. https://doi.org/10.1364/
with aging and age-related macular degenera- BOE.5.004329
tion. Exp Eye Res 78(2):243–256 19. Trucco E, Giachetti A, Ballerini L et al. (2015)
9. Aslam A, Peto T, Barzegar-Befroei N et al Morphometric measurements of the retinal
(2014) Assessing peripheral retinal drusen pro- vasculature in fundus images with vampire. In:
gression in Alzheimer’s dementia: a pilot study Biomedical image understanding. John Wiley
using ultra-wide field imaging. Invest & Sons, Inc., pp 91–111. doi:https://doi.
Ophthalmol Vis Sci 55(13):659–659 org/10.1002/9781118715321.ch3
10. Thomson KL, Yeo JM, Waddell B et al (2015) 20. Knudtson MD, Lee KE, Hubbard LD et al
A systematic review and meta-analysis of retinal (2003) Revised formulas for summarizing retinal
nerve fiber layer change in dementia, using vessel diameters. Curr Eye Res 27(3):143–149
optical coherence tomography. Alzheimers 21. Lisowska A, Annunziata R, Loh GK et al
Dement (Amst) 1(2):136–143. https://doi. (2014) An experimental assessment of five
org/10.1016/j.dadm.2015.03.001 indices of retinal vessel tortuosity with the
11. Cunha LP, Almeida AL, Costa-Cunha LV et al RET-TORT public dataset. Conf Proc IEEE
(2016) The role of optical coherence tomogra- Eng Med Biol Soc 2014:5414–5417. https://
phy in Alzheimer’s disease. Int J Retina Vitreous doi.org/10.1109/EMBC.2014.6944850
212 Tom MacGillivray et al.
22. Doubal FN, MacGillivray TJ, Patton N et al Eye Res. https://doi.org/10.1016/j.
(2010) Fractal analysis of retinal vessels sug- preteyeres.2017.04.003
gests that a distinct vasculopathy causes 25. Croft DE, van Hemert J, Wykoff CC et al
lacunar stroke. Neurology 74(14):1102– (2014) Precise montaging and metric quantifi-
1107. https://doi.org/10.1212/ cation of retinal surface area from ultra-wide-
WNL.0b013e3181d7d8b4 field fundus photography and fluorescein
23. Stosic T, Stosic BD (2006) Multifractal analysis angiography. Ophthal Surg Lasers Imaging
of human retinal vessels. IEEE Trans Med Retina 45(4):312–317. https://doi.
Imaging 25(8):1101–1107 org/10.3928/23258160-20140709-07
24. Cameron JR, Megaw RD, Tatham AJ et al 26. Kolb H (1995) Simple anatomy of the retina. In:
(2017) Lateral thinking – interocular sym- Kolb H, Fernandez E, Nelson R (eds) Webvision:
metry and asymmetry in neurovascular pat- the organization of the retina and visual system.
terning, in health and disease. Prog Retin University of Utah Health, Salt Lake City, UT
Chapter 15
Abstract
Degeneration of cortically projecting acetylcholine-containing neuronal populations within the basal forebrain
cholinergic system (BFCS) is a central pathogenetic aspect of Alzheimer’s disease (AD) and forms the rationale
for the use of cholinomimetics as antidementive treatment. The role of the cholinergic deficit in AD patho-
physiology has mainly been studied in experimental animal models and in neuropathologic examinations of
human autopsy data of advanced disease stages. Interactions between cholinergic deficits and accumulation of
cortical amyloid pathology point to a relevant role of BFCS degeneration for the preclinical stage of AD. The
advent of novel computational morphometry techniques for the automated analysis of high-resolution MRI
data allows studying AD-related atrophy in the living human brain with ever-increasing temporal and regional
detail. Combining these morphometry techniques with recently developed stereotactic mappings of the BFCS
provides a method for automated MRI-based volumetry that can sensitively assess degenerative changes of the
BFCS in vivo. Here, we outline the general methodological approach for MRI-based BFCS volumetry and
describe the specifics of different image processing choices and analysis strategies. We further discuss possibili-
ties and limitations of this method for studying BFCS degeneration in the course of AD, with a special empha-
sis on using MRI-based BFCS volumetry as an imaging biomarker for defining the preclinical disease stage.
Key words Cholinergic basal forebrain, Nucleus basalis Meynert, Substantia innominata, MRI,
Morphometry, Amyloid pathology
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_15, © Springer Science+Business Media, LLC 2018
213
214 Michel J. Grothe et al.
Fig. 1 Overview of substantia innominata and cholinergic basal forebrain. (a) A coronal proton-weighted MRI scan at
the level of the anterior commissure illustrating the anatomic location of the substantia innominata and neighboring
structures (Adapted from [21] with permission to reprint granted by Oxford University Press). (b) Computer-assisted
3D reconstruction of the basal forebrain cholinergic nuclei based on histology of a postmortem brain. Cholinergic
nuclei stretch in anterior-posterior direction from the medial septum (Ch1) over the vertical and horizontal limb of the
diagonal band of Broca (Ch2 and Ch3) to the nucleus basalis (Ch4). ac anterior commissure, fo fornix
Fig. 2 In vivo association between basal forebrain volume and cortical amyloid load. Scatter plots with linear
regression lines illustrate the association between cortical amyloid load measured in vivo by AV45-PET standard
uptake value ratio (SUVR) (x-axis) and cholinergic basal forebrain volume extracted using MRI-based volumetry
(y-axis) in cognitively normal (CN+; left) and early mild cognitive impairment (EMCI+; right) individuals with
detectable amyloid pathology. Figures in the bottom row show voxel-wise effects of a linear regression of corti-
cal AV45-PET SUVR on regional gray matter volume in the combined CN(+) and EMCI(+) subgroups. Effects are
superposed on coronal sections of the reference template and magnified to better depict the cholinergic basal
forebrain. Blue numbers indicate MNI space coordinates. Note the relative regional specificity of the amyloid
effect for the cholinergic basal forebrain nuclei. AC anterior commissure; AMG amygdala; DB diagonal band of
Broca; CA caudate; GP globus pallidus; (a, i, p) NBM, anterior, intermediate, posterior nucleus basalis Meynert;
PU putamen; TH thalamus (Figure adapted from [36] with permission to reprint granted by Elsevier)
2 Materials
2.1 Stereotactic Generation of MRI-based stereotactic atlases of the BFCS has been
Maps of the BFCS hindered by the lack of imaging contrast for the distinct cholinergic
cell clusters on common high-resolution structural MRI scans.
However, their localization can be mapped indirectly to stereotactic
In Vivo Volumetry of the Cholinergic Basal Forebrain 217
Fig. 3 Stereotactic mapping based on histology and postmortem MRI. The upper part of the figure shows the
postmortem processing with MRI scans of the brain in cranio (i.e., in situ) and after formalin fixation. Afterward
the subregions were identified by histological staining and manually transferred into the MRI of the dehydrated
brain in the alcohol space (color-coded mask at the lowest row of the figure). MRI scans were transferred first
into the MRI space of the in cranio scan and then into MNI standard space. The combined transformation
matrices were used to transfer the BFCS mask from alcohol space to MNI space via MRI in cranio space
(Reprinted from [34], with permission from IOS Press)
2.1.1 Acquisition Stereotactic mapping of the BFCS nuclei was based on an autopsy
of Postmortem MRI brain specimen of a non-demented 56-year-old man who died from
and Histologic Preparation a myocardial infarction. Results from a standardized and validated
questionnaire interview with an informant of the brain donor were
reviewed by a behavioral neurologist and two geriatricians and con-
sidered to be without any evidence for cognitive decline or psychiat-
ric illnesses prior to death [45]. A cerebral MRI scan was performed
in situ 15 h after death. Afterward the brain was removed from the
skull and placed into formalin deposit for 3 months and subsequently
dehydrated by upscaling ethanol storage. Before and after the brain
dehydration procedure, further MRI scans were performed.
After dehydration, the brain was prepared for histological stain-
ing [46, 47]. BFCS subregions were identified and delineated on
digital pictures of the stained slices (400 μm) following the Mesulam
nomenclature [22, 48]. The basal forebrain refers to a brain region
whose central parts are formed by the anterior perforate substance in
humans. This region is included in the “unnamed medullary sub-
stance” (substantia innominata) described by Reil [49], which
extends parallel to the optic nerve on the basis of the forebrain. In
their comprehensive studies, Heimer et al. [50, 51] summarized that
a high number of fiber systems and nuclei, including the ventral
striatum with ncl. accumbens, ventral pallidum, and extended amyg-
dala, are crowded in the substantia innominata. The NBM is distin-
guished by predominantly big hyperchromic neurons. Individual
neurons can already be identified at low magnification as small blue
dots after staining with gallocyanin, a classical Nissl stain (Fig. 4).
The cholinergic nature of many of these big hyperchromic neurons
Fig. 4 (continued) level of the anterior compartment of the ncl. accumbens [52] and central parts of the parater-
minal gyrus. Bar 1 mm, valid for all figures of the plate. Left hemisphere, 58-year-old male, myocardial infarct. (b)
Plane of section 2 mm caudal to (a) passing ventral pallidum extending into caudal compartment of ncl. accum-
bens. (c) Plane of section 4 mm caudal to (a) comprising the caudal parts of the anterior perforate substance and
the bed nucleus of the stria terminalis. (d) Plane of section 11 mm caudal to (a) comprising the optic tract with
supraoptic nucleus. (e) Plane of sections 16 mm caudal to (a) comprising crura cerebri and central nucleus of the
amygdaloid complex. (f) Horizontal gallocyanin-stained section through the left hemisphere of a 60-year-old
female, myocardial infarct. Arrowheads = branches of the anterolateral central arteries (Aa. centrales anterolate-
rales, perforant arteries); white arrowhead in figure (c) = perforating branch of central artery subdividing Ch4am
from Ch4al; asterisks = artifacts caused by either vacuum embedding of tissue, leakage of the blood-brain bar-
rier, or condensed galactolipids during alcohol dehydration of brain tissue. a c anterior commissure, acc ncl.
accumbens, acj juxtacommissural cells of the anterior commissure [53, 54], a l ansa lenticularis, a p ansa pedun-
cularis, Ayala pp periputaminal part of Ayala’s nucleus, Ayala sp subputimale part of Ayala’s nucleus, B st bed
nucleus of the stria terminalis, caud caudate nucleus, Ce central nucleus of the amygdala, Ch1 medial septum,
Ch2 vertical limb of the diagonal band of Broca, Ch3 horizontal limb of the diagonal band of Broca, Ch4al antero-
lateral part of Ch4, Ch4am anteromedial part of Ch4, Ch4p posterior part of Ch4, cr crus cerebri, c t claustrum
temporale or endopiriform nucleus, d B diagonal band of Broca, g p globus pallidus, gpe external pallidum, gpi
internal pallidum, g pt paraterminal (subcallosal) gyrus, I C insula magna of Calleja, i ol olfactory islands, put puta-
men, s o supraoptic nucleus, s t stria terminalis, s th subthalamic nucleus, tr o optic tract, v p ventral pallidum
In Vivo Volumetry of the Cholinergic Basal Forebrain 219
Fig. 4 Histology of the cholinergic basal forebrain nuclei. (a) Coronal gallocyanin (Nissl)-stained section at the
220 Michel J. Grothe et al.
2.1.2 Mapping The BFCS subregions identified on the histological slices were
from Histologic Slices manually transferred from the digital pictures into the correspond-
to Stereotactic Standard ing MR slices of the dehydrated brain. Due to the small size of
Space Via Postmortem MRI some of the delineated cell clusters, clusters corresponding to Ch1
and Ch2 were merged into one subdivision (Ch1-2), and within the
NBM we only considered a distinction between an anterior-inter-
In Vivo Volumetry of the Cholinergic Basal Forebrain 221
Fig. 5 Stereotactic maps of the cholinergic basal forebrain. Illustrations of the stereotactic maps of the cholin-
ergic basal forebrain in MNI space as published by [21] (a), [34] (b), and [18] (c). Panel (d) shows the stereo-
tactic maps from [18] that have finally been made publicly available through the SPM Anatomy toolbox [59].
Panel (c) is taken from the original publication in [18] (with permission to reprint granted by Elsevier), and
panels (a), (b), and (d) have been produced by projecting the stereotactic maps onto coronal sections with
identical MNI space coordinates as in (c) (running from y = 8 in the upper left corner to y = −7 in the lower
right corner). The probabilistic maps in (c) and (d) are represented as discrete maximum probability maps.
Note that these maps are designed to match the single-subject MNI space template, whereas maps in (a) and
(b) correspond to standard MNI space (MNI152 average template)
In Vivo Volumetry of the Cholinergic Basal Forebrain 223
3 Methods
3.1 Automated The availability of detailed stereotactic maps of the BFCS renders
Volumetry Approaches this brain region accessible to “atlas-based” automated volumetry
approaches. The overarching principle of these volumetry approaches
is to spatially transform (“normalize”) the individual native space
MRI scan into the standardized stereotactic space where the BFCS
atlas is defined in (typically MNI space) and use this transform to
calculate individual BFCS volumes. After matching the native space
scan to a stereotactic standard space template, the regional informa-
tion of an individual’s anatomy is encoded in the transformation
parameters that describe the deformations necessary for the nonlin-
ear matching (represented by vector fields, also called “deforma-
tion” fields). The Jacobian determinant map of these vector fields
specifically reflects the regional volumetric changes, i.e., the amount
of dilation or shrinkage that occurred at each voxel during matching
to the stereotactic standard space template.
In the most widely used “voxel-based morphometry” approach
to automated volumetry, the native space MRI scan is first seg-
mented into broad classes of tissue types (gray matter, white matte,
cerebrospinal fluid), and then the gray matter tissue of interest is
spatially transformed into stereotactic standard space, and voxel
values of the spatially transformed gray matter maps are finally
scaled (“modulated”) by the Jacobian determinant to reflect the
amount of regional gray matter tissue present before warping. The
sum of modulated gray matter voxel values within a given region
of interest (ROI) defined in template space (here the BFCS atlas)
represents the gray matter volume of this region in the individual
MRI scan (Fig. 6) [60].
In an alternative approach, sometimes referred to as “Jacobian
integration” or “tensor-based morphometry” [60, 61], the voxel
values are directly summed up on the Jacobian determinant maps
instead of the modulated gray matter maps. This approach produces
individual native space volumes of a given ROI independent of tissue
type and is equivalent to the volume calculation of a ROI mask that
has been transformed from template space to the individual’s native
space based on the inverse spatial transformation parameters [62].
224 Michel J. Grothe et al.
Fig. 6 Automated analysis of cholinergic basal forebrain volume. Flowchart illustrating the main preprocessing and
analysis steps for automated volumetry of the cholinergic basal forebrain. For details, see descriptions in the text
4 Conclusions
References
1. Whitehouse PJ, Price DL, Struble RG et al senile plaques and mental test scores in senile
(1982) Alzheimer's disease and senile demen- dementia. Br Med J 2(6150):1457–1459
tia: loss of neurons in the basal forebrain. 8. Ikonomovic MD, Klunk WE, Abrahamson EE
Science 215(4537):1237–1239 et al (2011) Precuneus amyloid burden is associ-
2. McGeer PL, McGeer EG, Suzuki J et al (1984) ated with reduced cholinergic activity in Alzheimer
Aging, Alzheimer's disease, and the choliner- disease. Neurology 77(1):39–47. https://doi.
gic system of the basal forebrain. Neurology org/10.1212/WNL.0b013e3182231419
34(6):741–745 9. Beach TG, McGeer EG (1992) Senile plaques,
3. Vogels OJ, Broere CA, ter Laak HJ et al (1990) amyloid beta-protein, and acetylcholinesterase
Cell loss and shrinkage in the nucleus basa- fibres: laminar distributions in Alzheimer's disease
lis Meynert complex in Alzheimer's disease. striate cortex. Acta Neuropathol 83(3):292–299
Neurobiol Aging 11(1):3–13 10. Arendt T, Bigl V, Tennstedt A et al (1985)
4. Perry EK, Blessed G, Tomlinson BE et al Neuronal loss in different parts of the nucleus
(1981) Neurochemical activities in human basalis is related to neuritic plaque formation
temporal lobe related to aging and Alzheimer- in cortical target areas in Alzheimer's disease.
type changes. Neurobiol Aging 2(4):251–256 Neuroscience 14(1):1–14
5. Bartus RT, Dean RL 3rd, Beer B et al (1982) 11. Beach TG, Honer WG, Hughes LH (1997)
The cholinergic hypothesis of geriatric mem- Cholinergic fibre loss associated with diffuse
ory dysfunction. Science 217(4558):408–414 plaques in the non-demented elderly: the
6. Birks J (2006) Cholinesterase inhibitors for preclinical stage of Alzheimer's disease? Acta
Alzheimer's disease. Cochrane Database Neuropathol 93(2):146–153
Syst Rev 1:CD005593. https://doi. 12. Potter PE, Rauschkolb PK, Pandya Y et al (2011)
org/10.1002/14651858.cd005593 Pre- and post-synaptic cortical cholinergic defi-
7. Perry EK, Tomlinson BE, Blessed G et al (1978) cits are proportional to amyloid plaque presence
Correlation of cholinergic abnormalities with and density at preclinical stages of Alzheimer's
In Vivo Volumetry of the Cholinergic Basal Forebrain 229
disease. Acta Neuropathol 122(1):49–60. 24. Muir JL (1997) Acetylcholine, aging, and
https://doi.org/10.1007/s00401-011-0831-1 Alzheimer’s disease. Pharmacol Biochem
13. Beach TG, Kuo YM, Spiegel K et al (2000) The Behav 56(4):687–696
cholinergic deficit coincides with Abeta depo- 25. Pappas BA, Bayley PJ, Bui BK et al (2000)
sition at the earliest histopathologic stages of Choline acetyltransferase activity and cogni-
Alzheimer disease. J Neuropathol Exp Neurol tive domain scores of Alzheimer's patients.
59(4):308–313 Neurobiol Aging 21(1):11–17
14. Young AL, Oxtoby NP, Daga P et al (2014) A 26. George S, Mufson EJ, Leurgans S et al
data-driven model of biomarker changes in spo- (2011) MRI-based volumetric measure-
radic Alzheimer's disease. Brain 137(Pt 9):2564– ment of the substantia innominata in amnes-
2577. https://doi.org/10.1093/brain/awu176 tic MCI and mild AD. Neurobiol Aging
15. Jack CR Jr, Knopman DS, Jagust WJ et al 32(10):1756–1764. https://doi.org/10.1016/
(2013) Tracking pathophysiological processes j.neurobiolaging.2009.11.006
in Alzheimer's disease: an updated hypothetical 27. Hanyu H, Tanaka Y, Shimizu S et al (2005)
model of dynamic biomarkers. Lancet Neurol Differences in MR features of the substan-
12(2):207–216. https://doi.org/10.1016/ tia innominata between dementia with Lewy
s1474-4422(12)70291-0 bodies and Alzheimer's disease. J Neurol
16. Huckman MS (1995) Where's the chicken? 252(4):482–484. https://doi.org/10.1007/
AJNR Am J Neuroradiol 16(10):2008–2009 s00415-005-0611-8
17. Hanyu H, Asano T, Sakurai H et al (2002) MR 28. Gao FQ, Pettersen JA, Bocti C et al (2013) Is
analysis of the substantia innominata in normal encroachment of the carotid termination into the
aging, Alzheimer disease, and other types of substantia innominata associated with its atrophy
dementia. AJNR Am J Neuroradiol 23(1):27–32 and cognition in Alzheimer's disease? Neurobiol
18. Zaborszky L, Hoemke L, Mohlberg H et al Aging 34(7):1807–1814. https://doi.
(2008) Stereotaxic probabilistic maps of the org/10.1016/j.neurobiolaging.2013.01.009
magnocellular cell groups in human basal fore- 29. Muth K, Schonmeyer R, Matura S et al (2010)
brain. NeuroImage 42(3):1127–1141. https:// Mild cognitive impairment in the elderly is
doi.org/10.1016/j.neuroimage.2008.05.055 associated with volume loss of the cholin-
19. Halliday GM, Cullen K, Cairns MJ (1993) ergic basal forebrain region. Biol Psychiatry
Quantitation and three-dimensional recon- 67(6):588–591. https://doi.org/10.1016/j.
struction of Ch4 nucleus in the human basal biopsych.2009.02.026
forebrain. Synapse 15(1):1–16. https://doi. 30. Moon WJ, Kim HJ, Roh HG et al (2008)
org/10.1002/syn.890150102 Atrophy measurement of the anterior com-
20. Teipel SJ, Flatz W, Ackl N et al (2014) Brain atro- missure and substantia innominata with 3T
phy in primary progressive aphasia involves the high-resolution MR imaging: does the mea-
cholinergic basal forebrain and Ayala's nucleus. surement differ for patients with fronto-
Psychiatry Res 221(3):187–194. https://doi. temporal lobar degeneration and Alzheimer
org/10.1016/j.pscychresns.2013.10.003 disease and for healthy subjects? AJNR Am
J Neuroradiol 29(7):1308–1313. https://doi.
21. Teipel SJ, Flatz WH, Heinsen H et al (2005) org/10.3174/ajnr.A1103
Measurement of basal forebrain atrophy in
Alzheimer's disease using MRI. Brain 128(Pt 31. Teipel SJ, Meindl T, Grinberg L et al (2011)
11):2626–2644. https://doi.org/10.1093/ The cholinergic system in mild cogni-
brain/awh589 tive impairment and Alzheimer's disease:
an in vivo MRI and DTI study. Hum Brain
22. Mesulam MM, Mufson EJ, Levey AI et al Mapp 32(9):1349–1362. https://doi.
(1983) Cholinergic innervation of cortex by org/10.1002/hbm.21111
the basal forebrain: cytochemistry and cortical
connections of the septal area, diagonal band 32. Grothe M, Zaborszky L, Atienza M et al (2010)
nuclei, nucleus basalis (substantia innomi- Reduction of basal forebrain cholinergic sys-
nata), and hypothalamus in the rhesus monkey. tem parallels cognitive impairment in patients
J Comp Neurol 214(2):170–197. https://doi. at high risk of developing Alzheimer's disease.
org/10.1002/cne.902140206 Cereb Cortex 20(7):1685–1695. https://doi.
org/10.1093/cercor/bhp232
23. Zaborszky L, Csordas A, Mosca K et al (2013)
Neurons in the basal forebrain project to the cor- 33. Grothe M, Heinsen H, Teipel SJ (2012)
tex in a complex topographic organization that Atrophy of the cholinergic basal forebrain
reflects Corticocortical connectivity patterns: an over the adult age range and in early stages
experimental study based on retrograde tracing of Alzheimer’s disease. Biol Psychiatry
and 3D reconstruction. Cereb Cortex. https:// 71(9):805–813. https://doi.org/10.1016/j.
doi.org/10.1093/cercor/bht210 biopsych.2011.06.019
230 Michel J. Grothe et al.
34. Kilimann I, Grothe M, Heinsen H et al 45. Grinberg LT, Ferretti RE, Farfel JM et al (2007)
(2014) Subregional basal forebrain atrophy Brain bank of the Brazilian aging brain study
in Alzheimer's disease: a multicenter study. group - a milestone reached and more than 1,600
J Alzheimers Dis 40(3):687–700. https://doi. collected brains. Cell Tissue Bank 8(2):151–162.
org/10.3233/jad-132345 https://doi.org/10.1007/s10561-006-9022-z
35. Teipel S, Heinsen H, Amaro E Jr et al (2014) 46. Grinberg LT, Amaro E Jr, Teipel S et al (2008)
Cholinergic basal forebrain atrophy pre- Assessment of factors that confound MRI and
dicts amyloid burden in Alzheimer's disease. neuropathological correlation of human postmor-
Neurobiol Aging 35(3):482–491. https://doi. tem brain tissue. Cell Tissue Bank 9(3):195–203.
org/10.1016/j.neurobiolaging.2013.09.029 https://doi.org/10.1007/s10561-008-9080-5
36. Grothe MJ, Ewers M, Krause B et al (2014) 47. Grinberg LT, Amaro Junior E, da Silva AV et al
Basal forebrain atrophy and cortical amyloid (2009) Improved detection of incipient vascular
deposition in nondemented elderly subjects. changes by a biotechnological platform combin-
Alzheimers Dement 10(5 Suppl):S344–S353. ing post mortem MRI in situ with neuropathol-
https://doi.org/10.1016/j.jalz.2013.09.011 ogy. J Neurol Sci 283(1–2):2–8. https://doi.
37. Kerbler GM, Fripp J, Rowe CC et al (2015) org/10.1016/j.jns.2009.02.327
Basal forebrain atrophy correlates with amy- 48. Mesulam MM, Mufson EJ, Wainer BH et al
loid beta burden in Alzheimer's disease. (1983) Central cholinergic pathways in the rat: an
Neuroimage Clin 7:105–113. https://doi. overview based on an alternative nomenclature
org/10.1016/j.nicl.2014.11.015 (Ch1-Ch6). Neuroscience 10(4):1185–1201
38. Schmitz TW, Nathan Spreng R (2016) Basal 49. Reil JC (1809) Untersuchungen über den Bau
forebrain degeneration precedes and predicts des grossen Gehirns im Mensch. Arch Physiol
the cortical spread of Alzheimer's pathol- (Halle) 9:136–146
ogy. Nat Commun 7:13249. https://doi. 50. Alheid GF, Heimer L (1988) New perspectives in
org/10.1038/ncomms13249 basal forebrain organization of special relevance
39. Grothe M, Heinsen H, Teipel S (2013) for neuropsychiatric disorders: the striatopallidal,
Longitudinal measures of cholinergic fore- amygdaloid, and corticopetal components of
brain atrophy in the transition from healthy substantia innominata. Neuroscience 27:1–39
aging to Alzheimer's disease. Neurobiol Aging 51. Heimer L, De Olmos J, Alheid GF et al (1991)
34(4):1210–1220. https://doi.org/10.1016/j. “perestroika” in the basal forebrain: opening
neurobiolaging.2012.10.018 the border between neurology and psychiatry.
40. Grothe MJ, Heinsen H, Amaro E Jr et al (2016) Prog Brain Res 87:109–165
Cognitive correlates of basal forebrain atro- 52. Lauer M, Heinsen H (1996) Cytoarchitectonics
phy and associated cortical Hypometabolism of the human nucleus accumbens. Journal of.
in mild cognitive impairment. Cereb Cortex Brain Res 37:243–254
26(6):2411–2426. https://doi.org/10.1093/ 53. Grinberg LT, Heinsen H (2007) Computer-
cercor/bhv062 assisted 3D reconstruction of the human basal
41. Ray NJ, Metzler-Baddeley C, Khondoker MR forebrain. Dementia & Neuropsychologia
et al (2015) Cholinergic basal forebrain struc- 2:140–146
ture influences the reconfiguration of white 54. Heinsen H, Hampel H, Teipel SJ (2006)
matter connections to support residual mem- Nucleus subputaminalis: neglected part of the
ory in mild cognitive impairment. J Neurosci basal nucleus of Meynert - response to Boban
35(2):739–747. https://doi.org/10.1523/ et al.: computer-assisted 3D reconstruction
jneurosci.3617-14.2015 of the nucleus basalis complex, including the
42. Cantero JL, Zaborszky L, Atienza M (2016) nucleus subputaminalis (Ayala's nucleus). Brain
Volume loss of the nucleus Basalis of Meynert 129(4):U1–U4
is associated with atrophy of innervated regions 55. Ayala G (1915) A hitherto undifferentiated
in mild cognitive impairment. Cereb Cortex. nucleus in the basal forebrain (nucleus subpu-
https://doi.org/10.1093/cercor/bhw195 taminalis). Brain 37:433–438
43. Grothe MJ, Scheef L, Bauml J et al (2016) 56. Simic G, Mrzljak L, Fucic A et al (1999)
Reduced cholinergic basal forebrain integrity links Nucleus subputaminalis (Ayala): the still disre-
neonatal complications and adult cognitive deficits garded magnocellular component of the basal
after premature birth. Biol Psychiatry. https:// forebrain may be human specific and connected
doi.org/10.1016/j.biopsych.2016.12.008 with the cortical speech area. Neuroscience
44. Kilimann I, Hausner L, Fellgiebel A et al 89(1):73–89
(2017) Parallel atrophy of cortex and basal 57. Ashburner J (2007) A fast diffeomorphic
forebrain cholinergic system in mild cognitive image registration algorithm. NeuroImage
impairment. Cereb Cortex 27(3):1841–1848. 38(1):95–113. https://doi.org/10.1016/j.
https://doi.org/10.1093/cercor/bhw019 neuroimage.2007.07.007
In Vivo Volumetry of the Cholinergic Basal Forebrain 231
58. Klein A, Andersson J, Ardekani BA et al (2009) disease. Front Aging Neurosci 6:264. https://
Evaluation of 14 nonlinear deformation algo- doi.org/10.3389/fnagi.2014.00264
rithms applied to human brain MRI registra- 69. Boban M, Kostovic I, Simic G (2006) Nucleus
tion. NeuroImage 46(3):786–802. https:// subputaminalis: neglected part of the basal nucleus
doi.org/10.1016/j.neuroimage.2008.12.037 of Meynert. Brain 129(Pt 4):E42.; author reply
59. Eickhoff SB, Stephan KE, Mohlberg H E43. https://doi.org/10.1093/brain/awl025
et al (2005) A new SPM toolbox for 70. Schliebs R, Arendt T (2006) The significance
combining probabilistic cytoarchitec- of the cholinergic system in the brain during
tonic maps and functional imaging data. aging and in Alzheimer’s disease. J Neural
NeuroImage 25(4):1325–1335. https://doi. Transm 113(11):1625–1644. https://doi.
org/10.1016/j.neuroimage.2004.12.034 org/10.1007/s00702-006-0579-2
60. Ashburner J (2009) Computational anatomy 71. Rogers JD, Brogan D, Mirra SS (1985) The
with the SPM software. Magn Reson Imaging nucleus basalis of Meynert in neurological disease:
27(8):1163–1174. https://doi.org/10.1016/j. a quantitative morphological study. Ann Neurol
mri.2009.01.006 17(2):163–170. https://doi.org/10.1002/
61. Boyes RG, Rueckert D, Aljabar P et al (2006) ana.410170210
Cerebral atrophy measurements using Jacobian 72. Samuel W, Terry RD, DeTeresa R et al (1994)
integration: comparison with the boundary shift Clinical correlates of cortical and nucleus basa-
integral. NeuroImage 32(1):159–169. https:// lis pathology in Alzheimer dementia. Arch
doi.org/10.1016/j.neuroimage.2006.02.052 Neurol 51(8):772–778
62. Malone IB, Leung KK, Clegg S et al (2015) 73. Iraizoz I, Guijarro JL, Gonzalo LM et al
Accurate automatic estimation of total intra- (1999) Neuropathological changes in the
cranial volume: a nuisance variable with less nucleus basalis correlate with clinical measures
nuisance. NeuroImage 104:366–372. https:// of dementia. Acta Neuropathol 98(2):186–196
doi.org/10.1016/j.neuroimage.2014.09.034 74. Grothe MJ, Schuster C, Bauer F et al (2014)
63. Avants BB, Epstein CL, Grossman M et al Atrophy of the cholinergic basal forebrain in
(2008) Symmetric diffeomorphic image regis- dementia with Lewy bodies and Alzheimer's dis-
tration with cross-correlation: evaluating auto- ease dementia. J Neurol 261(10):1939–1948.
mated labeling of elderly and neurodegenerative https://doi.org/10.1007/s00415-014-7439-z
brain. Med Image Anal 12(1):26–41. https:// 75. Arendt T, Bigl V, Arendt A et al (1983) Loss
doi.org/10.1016/j.media.2007.06.004 of neurons in the nucleus basalis of Meynert
64. Callaert DV, Ribbens A, Maes F et al (2014) in Alzheimer's disease, paralysis agitans
Assessing age-related gray matter decline with and Korsakoff's disease. Acta Neuropathol
voxel-based morphometry depends signifi- 61(2):101–108
cantly on segmentation and normalization pro- 76. Teipel S, Raiser T, Riedl L et al (2016)
cedures. Front Aging Neurosci 6:124. https:// Atrophy and structural covariance of the cho-
doi.org/10.3389/fnagi.2014.00124 linergic basal forebrain in primary progressive
65. Shen Q, Zhao W, Loewenstein DA et al (2012) aphasia. Cortex 83:124–135. https://doi.
Comparing new templates and atlas-based org/10.1016/j.cortex.2016.07.004
segmentations in the volumetric analysis of 77. Wolf D, Grothe M, Fischer FU et al (2014)
brain magnetic resonance images for diagnos- Association of basal forebrain volumes and
ing Alzheimer's disease. Alzheimers Dement cognition in normal aging. Neuropsychologia
8(5):399–406. https://doi.org/10.1016/ 53:54–63. https://doi.org/10.1016/j.
j.jalz.2011.07.002 neuropsychologia.2013.11.002
66. Eggert LD, Sommer J, Jansen A et al (2012) 78. Olesen OF, Dago L, Mikkelsen JD (1998)
Accuracy and reliability of automated gray mat- Amyloid beta neurotoxicity in the cholinergic
ter segmentation pathways on real and simulated but not in the serotonergic phenotype of RN46A
structural magnetic resonance images of the cells. Brain Res Mol Brain Res 57(2):266–274
human brain. PLoS One 7(9):e45081. https://
doi.org/10.1371/journal.pone.0045081 79. Zheng WH, Bastianetto S, Mennicken F et al
(2002) Amyloid beta peptide induces tau phos-
67. Gaser C (2009) Partial volume segmentation phorylation and loss of cholinergic neurons
with adaptive maximum a posteriori (MAP) in rat primary septal cultures. Neuroscience
approach. NeuroImage 47:S121. https://doi. 115(1):201–211
org/10.1016/s1053-8119(09)71151-6
80. Boncristiano S, Calhoun ME, Kelly PH et al
68. Voevodskaya O, Simmons A, Nordenskjold R (2002) Cholinergic changes in the APP23 trans-
et al (2014) The effects of intracranial volume genic mouse model of cerebral amyloidosis.
adjustment approaches on multiple regional J Neurosci 22(8):3234–3243. doi:20026314
MRI volumes in healthy aging and Alzheimer's
232 Michel J. Grothe et al.
Abstract
Advancement in biomarker research has enabled the in vivo detection of Alzheimer’s disease (AD) patho-
physiology, including amyloid plaques, neurofibrillary tangles, and neuronal degeneration. AD biomarkers
play an important role in characterizing the trajectory of AD and have been incorporated in the research
criteria for AD diagnosis. The presence of abnormal biomarkers for AD pathology in cognitively normal
individuals has further led to the proposal of a preclinical stage in the AD spectrum. With the emerging
conceptual framework that intervention in the early stages of the disease offers the greatest chance of suc-
cess in preventing or delaying AD progression, recent clinical trials are now focusing on individuals with
preclinical AD. While there are clear benefits from the use of biomarkers in research settings such as the
enrichment of clinical trial population to confirm the presence of target brain pathology and target engage-
ment by the intervention, the role of biomarkers in the clinical setting is less clear, especially in asymptom-
atic individuals. Potential ethical issues also arise with the use of biomarkers due to the conflict between
the principles of benefits and not doing harm. In fact, a unique set of ethical issues arises in asymptomatic
individuals, such as the disclosure of genetic mutation status, and abnormal biomarker results when their
diagnostic validity is uncertain. In this chapter, we will discuss the issues and clinical meaningfulness of
biomarkers in AD research. Specifically, we will focus on the potential benefits and ethical considerations
when genetics and biomarkers for amyloid, tau, and neurodegeneration are used in the early stages of AD.
Key words Alzheimer’s disease, Dementia, Mild cognitive impairment, Biomarker, Cerebrospinal
fluid, Imaging, Tau, Amyloid-beta, Ethics, Genetics, Early diagnosis, Prognosis
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_16, © Springer Science+Business Media, LLC 2018
235
236 Kok Pin Ng et al.
2 Definition of Preclinical AD
3 Ethical Principles
4 Genetic Biomarkers
4.1 Autosomal- Familial AD represents <1% of AD, and patients often present
Dominant AD Genes with an early-onset (before 65 years old) of cognitive symptoms.
(APP, PS1, PS2) In familial AD, there is overproduction of Aβ, either due to APP
gene mutation or mutations in genes encoding presenilin 1
(PSEN1) or presenilin 2 (PSEN2), which are key components of
the γ-secretase complexes that cleave and release Aβ. Given the
autosomal-dominant inheritance of these genes, individuals with a
positive family history of first-degree relatives with these genetic
mutations have a 50% chance of having the mutation. Also, with a
penetrance of almost 100%, the chance of progression to AD in
these mutation carriers is almost 100%.
4.1.2 Ethical Issues The presymptomatic ADAD mutation carriers represent a vulner-
able group of individuals who has full autonomy and competency.
Hence, genetic counseling plays a key role in protecting their inter-
est, and ample time must be allowed for making a decision to
undergo genetic testing. In fact, a joint practice guideline for
genetic counseling and testing has been proposed to guide clini-
cians in the effective counseling on hereditary risk and genetic test-
ing [11]. In addition, a structured three-phase framework has also
been proposed for the disclosure of AD diagnosis, which is espe-
cially important in this group of asymptomatic individuals with full
insight [20]. In the “before phase,” the key objective is to deter-
mine whether one wants to know the diagnosis, their coping strat-
egies and their psychological profiles, the time and place where
disclosure will take place, and the information that will be con-
veyed. In the “during phase,” the individual’s understanding of the
disease should be established, and confidentiality has to be main-
tained unless consent is obtained to disclose the diagnosis to any
family member. The subsequent phase focuses on ensuring that the
individual understands the information that is delivered and fol-
low-up meeting and support are provided.
The association of MCI and AD diagnosis with social stigmati-
zation and discrimination is well established [21]. This social stig-
matization and discrimination will likely be faced by ADAD
mutation carriers, who are destined to develop AD. Given a recent
shift of research from clinical stages to presymptomatic disease
stages, the knowledge of the mutation status and its impact on
social stigmatization in the cognitively normal population are still
elusive. Hence, it is important that this sensitive information be
kept confidential as they may have serious implications on employ-
ment and insurance. This is especially important when a mutation
carrier has intact cognition and mental capacity and is perfectly
well to work and contribute to the society.
While the potential benefits of ADAD genetic testing in gain-
ing access to clinical trials have been discussed, the therapeutic risk-
benefit ratio of the clinical trial in presymptomatic ADAD carriers
has to be considered. For example, the use of a novel anti-amyloid
treatment, although desirable in individuals with higher risk of
AD, may be less acceptable in healthy asymptomatic individuals,
due to potential side effects such as amyloid-related imaging
abnormalities [22]. In this regard, it is important to ensure that
the interventions being studied must pass through rigorous clinical
trials that evaluate drug safety and effectiveness [9].
4.2 APOE Gene There are three polymorphic alleles in the APOE gene, ε2, ε3, and
ε4, with a frequency of 8.4%, 77.9%, and 13.7%, worldwide.
However, the frequency of ε4 allele is increased in AD at an esti-
mate of 40% [23]. The ε4 allele is the strongest genetic risk factor
for sporadic AD, leading to an earlier age of onset in a gene dose-
240 Kok Pin Ng et al.
4.2.2 Ethical Issues However, as APOE ε4 is neither necessary nor sufficient for
developing AD, genetic testing for APOE ε4 is controversial, and
genetic counseling for the individual is important to discuss the
benefits and risks prior to testing. Although there is no effective
prevention or treatment when one is tested positive for APOE ε4,
these individuals may benefit from participating in preventive tri-
als. It is also important to emphasize that the confidentiality of
APOE ε4 status should be maintained, given that APOE ε4 is
increasingly tested in the research setting. This will protect the
interest of the individual as APOE ε4 carriers may face social stig-
matization and discrimination.
There have been concerns that disclosure of the genetic
screening results may lead to negative emotions among the APOE
ε4 carriers. In the Risk Evaluation and Education for AD
(REVEAL) study which examines the effects of APOE genotype
status disclosure to asymptomatic individuals with a first-degree
relative with AD, no difference was found in terms of anxiety and
Patient Benefit and Ethical Considerations of Biomarkers 241
5 Amyloid Biomarkers
Low CSF Aβ42 and high uptake of amyloid PET tracers are bio-
markers of amyloid fibrillary deposition [2]. There is a close cor-
relation between in vivo amyloid biomarkers and brain amyloid
plaques assessed in postmortem studies [29–31]. Longitudinal
studies have suggested that AD biomarkers, beginning with
amyloid deposition, become abnormal sequentially in a pro-
longed preclinical period [19]. In fact, CSF Aβ42 levels decline
up to 20 years before symptom onset [32]. There is high con-
cordance between CSF Aβ42 and amyloid PET in MCI and AD
patients [33]. In addition, amyloid PET provides information
regarding regional distribution and longitudinal changes of amy-
loid deposition. However, 10–30% of cognitively normal people
have a positive amyloid PET scan [34], and pathological amyloid
deposition in the brain is also shown in non-demented individu-
als [35]. Hence, amyloid pathology is proposed to be necessary,
but not sufficient to cause progression to a symptomatic stage of
the disease [2].
5.2 Ethical Issues While it is expected that AD biomarkers, including amyloid, will be
able to identify preclinical individuals who will progress to AD,
current biomarker follow-up studies provide elusive information
regarding biomarker signatures of those who will progress to
dementia [40, 41]. The uncertainty of AD diagnosis based on bio-
markers is further demonstrated in MCI individuals, where 50%
have amyloid pathology, but only 40% of them develop dementia
in 3 years [38]. Hence, the clinical use of amyloid biomarkers is
currently not recommended in asymptomatic individuals [37].
The disclosure of amyloid status to study participants is also a
key ethical issue. Among investigators, there are debates as to
whether the results of amyloid biomarkers should be revealed to
the participants. The Alzheimer’s Disease Neuroimaging Initiative
(ADNI) study has maintained a position where biomarker results,
including amyloid, are not disclosed to study participants, due to
uncertainty of the prognostic value of the biomarker [42].
However, this stance is likely to change once clinical utility associ-
ated with biomarkers is better established. In fact, recent clinical
trials, such as the A4 which studies disease-modifying agents in
amyloid-positive individuals, will, by study design, result in
asymptomatic participants being informed of their amyloid imag-
ing results [10]. Given that the implications of amyloid status on
employment, health insurance, and social stigmatization are not
fully understood, there is a need to accelerate the development of
a best practice framework for disclosure of amyloid status and to
identify individuals at risk of potential devastating reactions.
While amyloid PET has a clear benefit in research, its clinical
cost-effectiveness is less clear [43]. There remains an uncertainty
regarding the validity of amyloid PET, such as the presence of false
positives and false negatives, the adjustment of threshold for amyloid
positivity due to age and genetic factors, and the optimal threshold
for determining scan positivity using a quantitative approach [44].
Patient Benefit and Ethical Considerations of Biomarkers 243
6 Tau Biomarkers
6.1 Benefits Tau biomarkers have been incorporated in the research diagnostic
criteria which enables the detection of AD pathophysiology at the
early stages of the disease [4, 36]. Increased CSF p-tau is more
sensitive and specific than CSF t-tau and Aβ42 in discriminating AD
from normal aging and non-AD dementia [55], and a high CSF
p-tau/Aβ42 ratio also provides high diagnostic accuracy in differen-
tiating AD from normal controls and from subjects with non-AD
dementia [56]. High uptake of tau tracers in the temporal lobes is
able to distinguish AD from healthy individuals [57], and in
amyloid-positive individuals, increasing levels of tau binding are
associated with higher cognitive impairment [58].
Studies and clinical trials have tested interventions that target
pathological tau aggregation [59, 60], and similar to amyloid bio-
markers, it is likely that tau biomarkers will play a key role in future
clinical trials and observational studies. Although tau PET imaging
is still in the early stages of development, it is anticipated that tau
imaging agents will enable the staging of the disease, selection of
patients appropriate for a given tau therapy, confirmation of target
engagement, and monitoring treatment efficacy for AD [54].
244 Kok Pin Ng et al.
6.2 Ethical Issues Given that tau PET imaging is still in the early stages of develop-
ment, there has been limited clinical and research data for its cur-
rent use compared to amyloid PET imaging. In fact, off target
binding of tau tracers has been reported [61, 62], and its implica-
tions on the interpretation of scan findings will need to be carefully
studied, given the potential use of tau PET imaging in future clini-
cal trials as a marker of target engagement. In this regard, the dis-
closure of tau PET imaging results is also a potential ethical issue,
given the uncertainty of the validity of this biomarker.
7 Neurodegeneration Biomarkers
7.2 Ethical Issues While neurodegeneration biomarkers support the diagnosis of AD,
it is important to note that these biomarkers are least specific for
AD compared to biomarkers for amyloid and tau pathologies. Brain
atrophy in MRI and hypometabolism in FDG PET scan involving
regions associated with AD can also occur in a variety of other dis-
orders [71]. For example, atrophy in the anterior, medial, and basal
temporal lobes can occur in non-AD tauopathies such as progres-
sive supranuclear palsy, as well as in cerebrovascular disease, epi-
lepsy, hippocampal sclerosis, primary age-related tauopathy, and
argyrophilic grain disease. Hypometabolism in the temporoparietal
region can also be found in corticobasal degeneration and cerebro-
vascular disease [50]. The non-specificity of neurodegeneration
biomarkers has to be taken into account when explaining the
biomarker results to patients as this can lead to inaccurate diagnosis,
wrong treatment, and unnecessary anxiety.
8 Summary
Acknowledgments
References
1. Serrano-Pozo A, Frosch MP, Masliah E, 11. Goldman JS, Hahn SE, Williamson Catania
Hyman BT (2011) Neuropathological altera- J et al (2011) Genetic counseling and testing
tions in Alzheimer disease. Cold Spring Harb for Alzheimer disease: joint practice guidelines
Perspect Med 1:a006189 of the American college of medical genetics
2. Jack CR, Knopman DS, Jagust WJ et al (2013) and the national society of genetic counselors.
Tracking pathophysiological processes in Genet Med 13:597–605
Alzheimer’s disease: an updated hypothetical 12. Morris E, Chalkidou A, Hammers A et al
model of dynamic biomarkers. Lancet Neurol (2016) Diagnostic accuracy of (18)F amyloid
12:207–216 PET tracers for the diagnosis of Alzheimer’s
3. Dubois B, Feldman HH, Jacova C et al disease: a systematic review and meta-analysis.
(2007) Research criteria for the diagnosis of Eur J Nucl Med Mol Imaging 43:374–385
Alzheimer’s disease: revising the NINCDS- 13. Sevigny J, Chiao P, Bussière T et al (2016) The
ADRDA criteria. Lancet Neurol 6:734–746 antibody aducanumab reduces Aβ plaques in
4. McKhann GM, Knopman DS, Chertkow H Alzheimer’s disease. Nature 537:50–56
et al (2011) The diagnosis of dementia due to 14. Gauthier S, Leuzy A, Racine E, Rosa-Neto
Alzheimer’s disease: recommendations from P (2013) Diagnosis and management of
the national institute on aging-Alzheimer’s Alzheimer’s disease: past, present and future
association workgroups on diagnostic guide- ethical issues. Prog Neurobiol 110:102–113
lines for Alzheimer’s disease. Alzheimers 15. Liu C, Kanekiyo T, Xu H, Bu G (2013)
Dement 7:263–269 Apolipoprotein E and Alzheimer disease: risk,
5. Sperling RA, Aisen PS, Beckett LA et al mechanisms and therapy. Nat Rev Neurol
(2011) Toward defining the preclinical stages 9:106–118
of Alzheimer’s disease: recommendations 16. Sperling RA, Aisen PS, Beckett LA et al (2011)
from the national institute on aging and the Toward defining the preclinical stages of
Alzheimer’s association workgroup. Alzheimers Alzheimer’s disease: recommendations from
Dement 7:1–13 the national institute on aging- Alzheimer’s
6. Dubois B, Feldman HH, Jacova C et al (2014) association workgroups on diagnostic guide-
Advancing research diagnostic criteria for lines for Alzheimer’s disease. Alzheimers
Alzheimer’s disease: the IWG-2 criteria. Lancet Dement 7:280–292
Neurol 13:614–629 17. Steinbart EJ, S D MZ et al (2001) Impact of
7. Dubois B, Hampel H, Feldman HH et al DNA testing for early-onset familial Alzheimer
(2016) Preclinical Alzheimer’s disease: defini- disease and frontotemporal dementia. Arch
tion, natural history, and diagnostic criteria. Neurol 58:1828
Alzheimers Dement 12:292–323 18. Bateman RJ, Xiong C, Benzinger TLS,
8. Folch J, Petrov D, Ettcheto M et al (2016) Fagan AM, Goate A, Fox NC, Marcus DS,
Current research therapeutic strategies for Cairns NJ, Xie X, Nlazey TM, Holtman DM,
Alzheimer’s disease treatment. Neural Plast Santacruz A, Buckles V, Oliver A, Moulder
2016:1–15 K, Aisen PS, Ghetti B, Klunk WE, McDade
9. Bateman RJ, Benzinger TL, Berry S et al E, Martins RN, Masters CL, Mayeux R,
(2017) The DIAN-TU next generation Ringman JM, Rossor MN, Schofield PR,
Alzheimer’s prevention trial: adaptive design Sperling RA, Salloway S, Morris JC (2013)
and disease progression model. Alzheimers Clinical and biomarker changes in dominantly
Dement 13:8–19 inherited Alzheimer’s disease. N Engl J Med
10. Sperling RA, Aisen PS (2016) Anti-amyloid 367:795–804.
treatment of asymptomatic AD: A4 and 19. Bateman RJ, Xiong C, Benzinger TLS et al
beyond. Alzheimers Dement 12:P326–P327 (2012) Clinical and biomarker changes in
Patient Benefit and Ethical Considerations of Biomarkers 247
in the diagnosis of early Alzheimer’s disease. clinical diagnosis, age and cognition. Brain
Alzheimers Dement 11:58–69 140:748–763
46. Caselli R, Woodruff B (2016) Clinical impact 59. Gauthier S, Feldman HH, Schneider LS et al
of amyloid positron emission tomography—is it (2016) Efficacy and safety of tau-aggregation
worth the cost? JAMA Neurol 73:1396–1398 inhibitor therapy in patients with mild or
47. Sabbagh MN, Cooper K, DeLange J et al moderate Alzheimer’s disease: a randomised,
(2010) Functional, global and cognitive decline controlled, double-blind, parallel-arm, phase
correlates to accumulation of Alzheimer’s 3 trial. Lancet 388:2873–2884
pathology in MCI and AD. Curr Alzheimer 60. Yanamandra K, Jiang H, Mahan TE et al
Res 7:280–286 (2015) Anti-tau antibody reduces insoluble tau
48. Nelson PT, Alafuzoff I, Bigio EH et al (2012) and decreases brain atrophy. Ann Clin Transl
Correlation of Alzheimer disease neuropatho- Neurol 2:278–288
logic changes with cognitive status: a review 61. Ng KP, Pascoal TA, Mathotaarachchi S et al
of the literature. J Neuropathol Exp Neurol (2017) Monoamine oxidase B inhibitor, sele-
71:362–381 giline, reduces 18 F-THK5351 uptake in the
49. Blennow K, Hampel H (2003) CSF markers for human brain. Alzheimers Res Ther 9:25
incipient Alzheimer’s disease. Lancet Neurol 62. Vermeiren C, Mercier J, Viot D et al (2015)
2:605–613 T807, a reported selective tau tracer, binds
50. Jack CR, Hampel HJ, Universities S et al with nanomolar affinity to monoamine oxidase
(2016) A/T/N: an unbiased descriptive classi- a. Alzheimers Dement 11:P283
fication scheme for Alzheimer disease biomark- 63. Mosconi L (2013) Glucose metabolism in normal
ers. Neurology 87:539–547 aging and Alzheimer’s disease: m ethodological
51. Tapiola T, Alafuzoff I, Herukka S-K et al and physiological considerations for PET stud-
(2009) Cerebrospinal fluid β-amyloid 42 and ies. Clin Transl Imaging 1:217–233
tau proteins as biomarkers of Alzheimer-type 64. Ost M, Nylén K, Csajbok L et al (2006) Initial
pathologic changes in the brain. Arch Neurol CSF total tau correlates with 1-year outcome in
66:734–746 patients with traumatic brain injury. Neurology
52. Villemagne VL, Fodero-Tavoletti MT, Masters 67:1600–1604
CL, Rowe CC (2015) Tau imaging: early 65. Frisoni GB, Fox NC, Jack CR et al (2010) The
progress and future directions. Lancet Neurol clinical use of structural MRI in Alzheimer
14:114–124 disease. Nat Rev Neurol 6:67–77
53. Braak H, Braak E (1995) Staging of 66. Hansson O, Zetterberg H, Buchhave P et al
Alzheimer’s disease-related neurofibrillary (2006) Association between CSF biomarkers
changes. Neurobiol Aging 16:271–278. discus- and incipient Alzheimer’s disease in patients
sion 278–284 with mild cognitive impairment: a follow-up
54. Johnson KA, Schultz A, Betensky RA et al study. Lancet Neurol 5:228–234
(2016) Tau positron emission tomographic 67. Skillbäck T, Farahmand BY, Rosén C et al (2015)
imaging in aging and early Alzheimer disease. Cerebrospinal fluid tau and amyloid-β 1-42 in
Ann Neurol 79:110–119 patients with dementia. Brain 138:2716–2731
55. Koopman K, Le Bastard N, Martin J, Nagels 68. Mosconi L, Berti V, Glodzik L et al (2010)
G (2009) Improved discrimination of Pre-clinical detection of Alzheimer’s disease
autopsy-confirmed Alzheimer’s disease (AD) using FDG-PET, with or without amyloid
from non-AD dementias using CSF P-tau imaging. J Alzheimers Dis 20:843–854
181P. Neurochem Int 55:214–218 69. Foster NL, Heidebrink JL, Clark CM et al
56. Maddalena A, Papassotiropoulos A (2003) (2007) FDG-PET improves accuracy in dis-
Biochemical diagnosis of Alzheimer disease by tinguishing frontotemporal dementia and
measuring the cerebrospinal fluid ratio of phos- Alzheimer’s disease. Brain 130:2616–2635
phorylated tau protein to β-amyloid peptide42. 70. Scheltens P, Leys D, Barkhof F et al (1992)
Arch Neurol 60:1202–1206 Atrophy of medial temporal lobes on MRI in
57. Harada R, Okamura N, Furumoto S et al “probable” Alzheimer’s disease and normal
(2016) 18F-THK5351: a novel PET radio- ageing: diagnostic value and neuropsychologi-
tracer for imaging neurofibrillary pathology in cal correlates. J Neurol Neurosurg Psychiatry
Alzheimer’s disease. J Nucl Med 57:208–214 55:967–972
58. Pontecorvo MJ, Devous MD Sr, Navitsky 71. Fotuhi M, Do D, Jack C (2012) Modifiable
M et al (2017) Relationships between flor- factors that alter the size of the hippocampus
taucipir PET tau binding and amyloid burden, with ageing. Nat Rev Neurol 8:189–202
Chapter 17
Abstract
In older adults who are developing Alzheimer-typical pathology but do not experience any symptoms—a
condition termed “preclinical” Alzheimer’s disease—the individual prognostic value of current biomarkers
is low. Only a minority of those who have positive findings on biomarkers in the cerebrospinal fluid or
upon brain imaging experience cognitive decline within 3–5 years. On the other hand, the majority of
people who have negative biomarker results remain cognitively intact. This may be expected in research
settings where alternative causes of cognitive impairment are infrequent. The main reasons for the poor
predictive performance of biomarkers in asymptomatic individuals are the dynamic nature of the assess-
ments with abnormality evolving over time and the large interindividual variability of the threshold for
clinical manifestation. We do not recommend biomarker assessment and disclosure of biomarker results to
asymptomatic individuals outside of research protocols or clinical trials at this point in time. More needs
to be learned about the interplay between biomarkers, risk factors, and protective factors. Also, studies are
required with extended follow-up intervals on less selected participant groups to determine the long-term
predictive potential of biomarkers in presymptomatic Alzheimer’s disease.
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_17, © Springer Science+Business Media, LLC 2018
249
250 Alexander F. Kurz and Nicola T. Lautenschlager
2 Methods
3 Results
3.1 What Kind Two types of biomarkers can be distinguished: one category indi-
of Information Do cating β-amyloid pathology (low β-amyloid concentration in the
Biomarkers Provide? CSF and evidence of β-amyloid deposits on imaging) and another
category demonstrating neuronal injury and neurodegeneration
(high total tau or phosphorylated tau in the CSF, glucose hypome-
tabolism in typical locations, accumulation of tau, and hippocam-
pal atrophy on imaging). When using and interpreting biomarkers,
particularly in asymptomatic people, their specific nature and asso-
ciated problems need to be observed.
–– Biomarkers of both categories capture different single ele-
ments of AD neurodegeneration [5, 6]. These features are not
specific of AD but also occur in other brain diseases such as
Ethics of Biomarker-Based Preclinical Diagnosis 251
3.2 How Well Do Among cognitively healthy older adults, 25–45% have abnormal CSF
Biomarkers Predict or imaging biomarker values [33]. Amyloid positivity on imaging
Cognitive Decline? increases with age from 5% in those 50–60 years to 46% in people
aged 81 years and older [34]. Abnormal CSF biomarker results are
associated with an increased likelihood of cognitive decline within a
follow-up interval of 3 years. However, even in a research setting,
using the best biomarker combination and optimal cutoff values,
individual prediction only achieved 65% accuracy, with sensitivity and
specificity values being 74% and 57%, respectively. These figures indi-
cate that one third of cognitive decliners were not identified, while
almost half of the predictions were false positive [35]. In a longitudi-
nal study at Washington University, cognitively healthy adults over
the age of 65 years who were positive for CSF ß-amyloid showed
clinical deterioration over 5 years in only 13%. If they also had a posi-
tive biomarker for neuronal injury, the progression rate increased to
25% [1]. Thus, a considerable number of healthy older individuals
with abnormal CSF biomarker findings will not experience cognitive
deterioration within the next couple of years. Similarly, abnormal
findings on ß-amyloid imaging are generally associated with greater
decline in memory and other domains and indicate an increased risk
of dementia on a group level [36]. However, such results do not
predict with certainty if and when someone will develop clinical
symptoms [37–40]. In the Australian Imaging, Biomarkers and
Lifestyle (AIBL) study, only 26% of cognitively healthy individuals
who had an AD-like result on ß-amyloid imaging progressed to MCI
or dementia within 3 years. If a positive ß-amyloid scan was com-
bined with reduced hippocampal volume on MRI, the percentage of
cognitive decliners increased to 47%. Among participants with nor-
mal imaging findings in both modalities, 92% remained cognitively
intact [41]. These findings show that within a follow-up period of
several years, less than half of cognitively healthy older adults who
have abnormal biomarker findings will experience cognitive decline.
Most but not all of those who have normal biomarker results are
Ethics of Biomarker-Based Preclinical Diagnosis 253
3.4 What Are There are two kinds of risks associated with biomarker assessment.
the Risks Associated The first kind are minor adverse effects that may be associated with
with Biomarker the assessment procedure, i.e., the invasiveness of lumbar puncture
Information? for CSF measurements, and the radiation exposure associated with
PET scanning. The second kind of risk is related to the informa-
tion provided by the biomarker results. In the absence of preven-
tive or treatment options, this information is a potentially harmful
knowledge. It may cause psychological distress including anxiety,
depression or even suicidal ideation, disruption of family dynamics,
and worries about loss of or failure to obtain insurance or other
social or economical benefits such as employment [45, 46] or lead
254 Alexander F. Kurz and Nicola T. Lautenschlager
3.6 What Will Novel treatments are currently being evaluated which address the
Change amyloid and tau components of AD. Examples are passive or active
with the Advent immunization which inhibits the aggregation of the two proteins
of Disease-Modifying and/or removes the proteins from the brain or enzyme blockers
Treatment? that reduce the production of the proteins. Expected efficacy is a
slowing of neurodegeneration, and the hope is that early use of
these treatments will delay the onset of cognitive impairment and
disability in daily living. Biomarkers will then be the gatekeepers to
treatment and determine the access to treatment. However, these
novel treatments are not without side effects and most probably
will not be tolerated by everyone. In addition, the indication (when
to start treatment, when to stop) and reimbursement by health
insurances is unknown to date. Furthermore, a proportion of peo-
ple who might be eligible for treatment may not wish to be treated,
balancing the benefit of postponing the onset of symptoms in an
uncertain future with the procedural burden and cost of treatment
[58]. It is therefore likely that only a minority of people who are at
risk, i.e., who have no symptoms but ongoing neurodegeneration,
will access these novel treatments. The majority of those tested
positively for the presence of AD pathology will not gain access to
disease-slowing treatment but will have the information of a fatal
brain disease. For them and their families, practical coping strate-
gies and concrete problem solutions need to be delineated [47].
4 Discussion
References
1. Vos SJB, Xiong C, Visser PJ et al (2013) 6. Witte MM, Foster NL, Fleisher AS et al
Preclinical Alzheimer’s disease and its out- (2015) Clinical use of amyloid-positron emis-
come: a longitudinal cohort study. Lancet sion tomography neuroimaging: practical and
Neurol 12:957–965 bioethical considerations. Alzheimers Dement
2. Dubois B, Hampel H, Feldman HH et al 1:358–367
(2016) Preclinical Alzheimer’s disease: defini- 7. Jack CR, Albert M, Knopman DS et al (2011)
tion, natural history, and diagnostic criteria. Introduction to revised criteria for the diag-
Alzheimers Dement 12:292–323 nosis of Alzheimer’s disease: national institute
3. Petersen RC (2011) Mild cognitive impair- on aging and the Alzheimer association work-
ment. N Engl J Med 364:2227–2234 groups. Alzheimers Dement 7:257–262
4. Ritchie K, Carrière I, Berr C et al (2016) The 8. Arlt S (2013) Non-Alzheimer’s disease–related
clinical picture of Alzheimer’s disease in the memory impairment and dementia. Dialogues
decade before diagnosis: clinical and biomarker Clin Neurosci 15:465–473
trajectories. J Clin Psychiatry 77:e305–e311 9. Villemagne VL, Burnham S, Bourgeat P et al
5. Roberts JS, Dunn LB, Rabinovici GD (2013) Amyloid beta deposition, and cogni-
(2013) Amyloid imaging, risk disclosure and tive decline in sporadic Alzheimer’s disease:
Alzheimer’s disease: ethical and practical issues. a prospective cohort study. Lancet Neurol
Neurodegener Dis Manag 3:219–229 12:357–367
Ethics of Biomarker-Based Preclinical Diagnosis 257
10. Han SD, Gruhl J, Beckett L et al (2012) 23. Murray AD, Staff RT, McNeil CJ et al (2011)
Beta amyloid, tau, neuroimaging, and cogni- The balance between cognitive reserve and
tion: sequence modeling of biomarkers for brain imaging biomarkers of cerebrovascular and
Alzheimer’s disease. Brain Imaging Behav Alzheimer’s diseases. Brain 134:3687–3696
6:610–620 24. Almeida RP, Schultz SA, Austin BP et al (2015)
11. Jack CR, Knopman DS, Jagust WJ et al (2013) Cognitive reserve modifies age-related altera-
Tracking pathophysiological processes in tions in CSF biomarkers of Alzheimer’s disease.
Alzheimer’s disease: an updated hypothetical JAMA Neurol 72:699–706
model of dynamic biomarkers. Lancet Neurol 25. Doraiswamy PM, Sperling RA, Johnson K
12:207–216 et al (2014) Florbetapir F 18 amyloid PET
12. Sutphen CL, Jasielec MS, Shah AR et al (2015) and 36-month cognitive decline: a pro-
Longitudinal cerebrospinal fluid biomarker spective multicenter study. Mol Psychiatry
changes in preclinical Alzheimer disease during 19:1044–1051
middle age. JAMA Neurol 72:1029–1042 26. McKhann GM, Knopman DS, Chertkow
13. Wirth M, Villeneuve S, Haase CM et al (2013) H et al (2011) The diagnosis of dementia
Association between Alzheimer disease bio- due to Alzheimer’s disease: recommenda-
markers, neurodegeneration, and cognition in tions from the national institute on aging
cognitively normal older people. JAMA Neurol and the Alzheimer’s association workgroup.
70:1512–1519 Alzheimers Dement 7(3):263–269
14. Knopman DS, Jack CR, Lundt ES et al (2015) 27. Janelidze S, Zetterberg H, Mattsson N
Role of beta-amyloidosis and neurodegenera- et al (2016) CSF Abeta 42/Abeta 40 and
tion in subsequent changes in mild cognitive Abeta42/−abeta38 ratios: better diagnos-
impairment. JAMA Neurol 72:1475–1483 tic markers of Alzheimer’s disease. Ann Clin
15. Alexopoulos P, Werle L, Roesler J et al (2016) Transl Neurol 3:154–165
Conflicting cerebrospinal fluid biomarkers and 28. Counts SE, Ikonomovic MD, Mercao N
progression to dementia due to Alzheimer’s et al (2017) Biomarkers for the early detec-
disease. Alzheimers Res Ther 8:51 tion and progression of Alzheimer’s disease.
16. Porteri C, Frisoni GB (2014) Biomarker-based Neurotherapeutics 14:35–53
diagnosis of mild cognitive impairment due to 29. Chételat G, La Joie R, Villain N et al (2013)
Alzheimer’s disease: how and what to tell. A Amyloid imaging in cognitively normal indi-
kickstart to an ethical discussion. Front Aging viduals, at-risk populations and preclini-
Neurosci 6:41 cal Alzheimer’s disease. NeuroImage Clin
17. Landau SM, Lu M, Joshi AD et al (2013) 2:356–365
Comparing PET imaging and CSF measure- 30. Soldan A, Pettigrew C, Cai Q et al (2016)
ments of A beta. Ann Neurol 74:826–836 Hypothetical preclinical Alzheimer disease
18. Toledo JB, Weiner MW, Wolk DA et al (2014) groups and longitudinal cognitive change.
Neuronal injury biomarkers and prognosis in JAMA Neurol 73:698–705
ADNI subjects with normal cognition. Acta 31. Goldman JS (2012) New approaches to genetic
Neuropathol Commun 2:26 counseling and testing for Alzheimer’s disease
19. Leuzy A, Carter SF, Chiotis K et al (2015) and frontotemporal degeneration. Curr Neurol
Concordance and diagnostic accuracy of 11C Neurosci Rep 12:502–510
PIB PET and cerebrospinal fluid biomarkers in 32. Hauser PS, Ryan RO (2013) Impact of apo-
a sample of patients with mild cognitive impair- lipoprotein E on Alzheimer’s disease. Curr
ment and Alzheimer’s disease. J Alzheimers Alzheimer Res 10:809–817
Dis 45:1077–1088 33. Randall C, Mosconi L, de Leon M et al (2014)
20. Leuzy A, Chiotis K, Hasselbalch SG et al Cerebrospinal fluid biomarkers of Alzheimer’s
(2016) Pittsburgh compound B imaging and disease in cognitively healthy elderly. Front
cerebrospinal fluid amyloid-beta in a multi- Biosci 18:1150–1173
centre European memory clinic study. Brain 34. Fleisher AS, Chen K, Liu X et al (2013)
139:2540–2553 Apolipoprotein E e4 effects on florbetapir
21. Vos SJB, Gordon BA, Su Y et al (2016) positron emission tomography in healthy
NIAA-AA staging of preclinical Alzheimer dis- aging and Alzheimer disease. Neurobiol Aging
ease: discordance and concordance of CSF and 34:1–12
imaging biomarkers. Neurobiol Aging 44:1–8 35. Steenland K, Zhao L, Goldstein F et al (2014)
22. Valenzuela MJ (2008) Brain reserve and the Biomarkers for predicting cognitive decline in
prevention of dementia. Curr Opin Psychiatry those with normal cognition. J Alzheimers Dis
21:296–302 40:587–594
258 Alexander F. Kurz and Nicola T. Lautenschlager
36. Johnson KA, Minoshima S, Bohnen NI et al 49. Villemagne VL, Fodero-Tavoletti MT, Masters
(2013) Appropriate use criteria for amy- CL et al (2015) Tau imaging: early progress and
loid PET: a report of the amyloid imaging future directions. Lancet Neurol 14:114–124
task force, the society of nuclear medicine 50. Tanzi RE (2012) The genetics of Alzheimer
and molecular imaging, and the Alzheimer’s disease. Cold Spring Harb Perspect Med
association. Alzheimers Dement 9: 2:a006296
e-1–e-15 51. Bemelmans SASA, Tromp K, Bunnik EM et al
37. Morris JC, Roe CM, Grant EA et al (2009) PIB (2016) Psychological, behavioral and social
imaging predicts progression from cognitively effects of disclosing Alzheimer’s disease bio-
normal to symptomatic Alzheimer’s disease. markers to research participants: a systematic
Arch Neurol 66:1469–1475 review. Alzheimers Res Ther 8:46
38. Villemagne VL, Pike KE, Chételat G et al 52. Lim YY, Maruff P, Getter C et al (2016)
(2011) Longitudinal assessment of Abeta and Disclosure of positron emission tomography
cognition in aging and Alzheimer disease. Ann amyloid imaging results: a preliminary study
Neurol 69:181–192 of safety and tolerability. Alzheimers Dement
39. Harkins K, Sankar P, Sperling R et al (2015) 12:454–458
Development of a process to disclose amyloid 53. Paulsen JS, Nance M, Kim JI et al (2013) A
imaging results to cognitively normal older review of quality of life after predictive testing
adult research participants. Alzheimers Res for and earlier identification of neurodegenera-
Ther 7:26 tive diseases. Prog Neurobiol 110:2–28
40. Rabinovici GD, Karlawish J, Knopman D et al 54. Hooper M, Grill JD, Rodriguez-Agudelo Y
(2016) Testing and disclosure related to amy- et al (2013) The impact of the availability of
loid imaging and Alzheimer’s disease: common prevention studies on the desire to undergo
questions and fact sheet summary. Alzheimers predictive testing in persons at-risk for autoso-
Dement 12:510–515 mal dominant Alzheimer’s disease. Contemp
41. Rowe CC, Bourgeat P, Ellis KA et al (2013) Clin Trials 36:256–262
Predicting Alzheimer disease with beta-amyloid 55. Steinbart EJ, Smith CO, Poorkay P et al (2001)
imaging: results from the Australian imaging, Impact of DNA testing for early-onset familial
biomarkers, and lifestyle study of ageing. Ann Alzheimer disease and frontotemporal demen-
Neurol 74:905–913 tia. Arch Neurol 58:1828–1831
42. Howe E (2013) Clinical implications of the 56. Cassidy MR, Roberts JS, Bird TD et al (2008)
new diagnostic guidelines for dementia. Innov Comparing test-specific distress of suscep-
Clin Neurosci 10:32–38 tibility versus deterministic genetic testing
43. Baum ML (2016) Patient requests for off-label for Alzheimer’s disease. Alzheimers Dement
bioprediction of dementia. Camb Q Healthc 4:406–413
Ethics 25:686–690 57. Goldman JS, Rademakers R, Huey ED et al
44. Gordon M (2013) Identification of potential (2011) An algorithm for genetic testing of
or preclinical cognitive impairment and the frontotemporal lobar degeneration. Neurology
implications of sophisticated screening with 76:476–483
biomarkers and cognitive testing: does it really 58. Molinuevo JL, Cami J, Carné S et al (2016)
matter? Biomed Res Int 2013:976130 Ethical challenges in preclinical Alzheimer’s
45. Karlawish J (2011) Addressing the ethi-
disease observational studies and trials: results
cal, policy, and social challenges of pre- of the Barcelona summit. Alzheimers Dement
clinical Alzheimer disease. Neurology 77: 12:614–622
1487–1493 59. Soldan A, Pettigrew C, Li S et al (2013)
46. Kim SYH, Karlawish J, Berkman BE (2015) Relationship of cognitive reserve and CSF bio-
Ethics of genetic and biomarker test disclosures markers to emergence of clinical symptoms
in neurodegenerative disease prevention trials. in preclinical Alzheimer’s disease. Neurobiol
Neurology 84:1488–1494 Aging 34:2827–2834
47. Schicktanz S, Schweda M, Ballenger JF et al 60. Donovan NJ, Hsu DC, Dagley AS et al (2015)
(2014) Before it is too late: professional respon- Depressive symptoms and biomarkers of
sibilities in late-onset Alzheimer’s research Alzheimer’s disease in cognitively normal older
and pre-symptomatic prediction. Front Hum adults. J Alzheimers Dis 46:63–73
Neurosci 8:921 61. Garrett MD, Valle R (2016) A methodologi-
48. Caselli RJ, Langbaum J, Marchant GE et al cal critique of the national institute of aging
(2014) Public perceptions of presymptomatic and Alzheimer’s association guidelines for
testing for Alzheimer’s disease. Mayo Clin Proc Alzheimer’s disease, dementia, and mild cogni-
89:1389–1396 tive impairments. Dementia 15:239–254
Chapter 18
Abstract
Shared decision-making is a model of medical decision-making, which aims at reducing the informa-
tional and decisional power asymmetry between physician and patient to obtain jointly reached
agreements on medical treatment. SDM can also be applied in the case of early stage of Alzheimer’s
disease, as long as individuals’ with dementia decisional capacity still exists. Because of the loss of
cognitive functions in the course of the disease, upcoming important issues like medical treatment
and legal and social issues should be made as soon as possible after diagnosis, so that persons with
dementia can be involved in the process of decision-making.
Key words Alzheimer’s disease, Social aspects, Early diagnosis, Dementia, Shared decision-making
1 Introduction
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4_18, © Springer Science+Business Media, LLC 2018
259
260 Katharina Bronner and Johannes Hamann
2.1 Four Models In their seminal paper, Emanuel and Emanuel [10] describe four
of Physician-Patient models of the doctor-patient relationship. In the paternalistic model
Relationship patients get selected information and instructions from the physi-
cian, which they are expected to follow. The physician makes deci-
sions about medical procedures like clinical diagnostics and therapy
all alone. And there is no explicit patient involvement in decision-
making. In the informative model the physician provides the patient
with all relevant specialized information without giving an advice.
The patient then comes to a decision all alone, which the physician
is expected to implement. In the interpretative model, the physician
provides the relevant information, inquires moral values and objec-
tives, and proposes appropriate measures of therapy. The patient
decides alone; however, the physician is involved in decision-
making. The deliberative model assumes that the physician discusses
the best treatment option with the patient. He knows about the
living conditions and also the moral values and objectives of the
patient. He tries to convince the patient to accept the best option,
but if the patient disagrees, the physician doesn’t try to overrule
him. Decisions are made by the patient and physician together.
Shared Decision-Making and Important Medical and Social Decisions 261
2.2 Shared Shared decision-making (SDM) has been located right in the middle
Decision-Making of the paternalistic and the informative model [11]. Already in 1997
four key features were postulated: (1) At least two persons are partici-
pating, physician and patient; (2) both sides share information with
each other; (3) both sides take steps to actively engage in a process of
decision-making; (4) both sides accept the decision and actively con-
tribute to its implementation [9]. Since the 1990s enormous political
efforts have been undertaken to foster research on and implementa-
tion of SDM in various healthcare systems [12–14].
Research has shown that SDM has the potential to improve
patients’ treatment satisfaction as well as their adherence with
therapies [15, 16].
3.1 Decision-Making In mental health, patient participation and SDM have not yet been
in Mental Illness fully implemented, although the majority of individuals with men-
tal illnesses express a clear wish for information and participation in
medical decisions and although there is growing evidence that
SDM may positively influence treatment outcomes [17]. Meanwhile
several clinical guidelines explicitly recommend patient participa-
tion in all medical decisions for the majority of mental illnesses,
such as depression or schizophrenia (e.g., National Institute for
Health and Care Excellence (NICE) [18, 19]).
However, in mental health it may be a special challenge to
engage individuals into medical decision-making, especially due to
the cognitive limitations, lack of insight, and reduced decisional
capacity that all may be a result of the mental illness [20].
3.3 Decision-Making Individuals in early stages of dementia are in most cases still capable
in Early-Stage of making decisions [26] and actually want to have a say in decisions
Dementia concerning their own treatment [27]. Thereby, participation pref-
erences of persons with dementia were especially marked in deci-
sions about social issues, for example, future housing or restitution
of the driver’s license. On the contrary, individuals with dementia
prefer to hand over decisions about medical issues to their physi-
cians. From the persons’ with dementia point of view, relatives
should play a minor part in medical as well as in social decisions
[28]. It can be assumed, however, that individuals’ participation in
decision-making does not take place, because psychiatrists tend to
exclude cognitively impaired individuals in decision-making [29],
i.e., when they are worried about the decrease of decisional capacity
in the progression of dementia [26, 30]. Additionally many persons
with AD are prevented from the process of decision-making,
because their relatives tend to take over control [27].
In most studies on decision-making in early-stage dementia,
more emphasis was placed on the relationship between individuals
with dementia and relatives and relatives and physicians than on
the issue of shared decision between individuals with dementia and
physicians [21, 31, 32].
3.4 Decision-Making For individuals at the stage of MCI and even more for individuals
at the Stage of MCI in a preclinical stage of AD, decisional capacity is in most cases not
and at the Preclinical a problematic issue in decision-making. Here it is rather a compli-
Stage of AD cated matter regarding the often debatable decisions (e.g., for or
against excessive diagnostic testing) that can or should be made at
these stages of the disease.
Thus, before dementia can be diagnosed, many individuals
pass through a period of mild cognitive impairment (MCI) [33].
This diagnosis has actually no therapeutic consequences, because
there is no medical indication for the use of the licensed antide-
mentives for prevention at the stage of MCI and at the preclinical
stage of AD, respectively [34, 35]. Moreover, there is an ongoing
debate about whether persons without any symptom of dementia
should participate in a predictive examination of AD by use of
diagnostic methods of early detection [36]. Although studies have
finally identified relevant cardiovascular risk factors for develop-
ment of a dementia (smoking, hypertension, obesity, diabetes mel-
litus, and physical inactivity) [37, 38] and there are hints for
protecting dementia by special nutrition [39] and sport activity
[40], there are no specific recommendations for risk reduction of
AD but only generally effective measures, which have a preventive
effect on other diseases, too (“what is good for the heart, is good
for the brain”) [41]. Predictive diagnostics can also be adverse for
individuals. Experiences with cancer screening have shown that
early detection is in no way without side effects [42]. Using cur-
rent methods there is, except the rare heritable form of AD [43],
Shared Decision-Making and Important Medical and Social Decisions 263
4.1 Medical Although there isn’t an effective therapy yet, which can cure or at least
Treatment stop the disease process, there are drugs with disease-delaying proper-
ties and drugs for accompanying symptoms such as depression, anxiety,
and aggression [47]. It is also possible for persons with AD to partici-
pate in clinical trials. In addition, psychosocial interventions are avail-
able, which can increase individuals’ with dementia and relatives’
well-being and quality of life.
4.1.1 Drug Treatment Currently there is no causal medical therapy for AD. There are
hints that early drug treatment may have positive effects on the
disease process [48]. Licensed antidementives are able to delay
dementia symptoms for all stages of AD [49] and also have an
effect on mental and behavioral symptoms [50] in some cases.
However, over the course of time, all individuals with AD will
264 Katharina Bronner and Johannes Hamann
develop severe dementia with the need for care, and, like all drugs,
antidementives may have side effects, especially on the gastrointes-
tinal system. On the part of the physicians, comprehensive infor-
mation should take place about the potential benefits and side
effects using a shared decision-making approach. In addition, the
benefit-risk profile should be observed carefully, especially when
prescribed for early stages of AD [7].
In addition to treatment with licensed drugs, persons with
dementia may participate in clinical trials, which are performed
at specialized and certificated centers. The European parent
organization Alzheimer Europe has published information and a
register of current clinical trials [51].
4.1.2 Psychosocial Besides drug treatment psychosocial interventions are part of persons’
Interventions with dementia and relatives’ support in dementia treatment. This
includes, for example, cognitive training, occupational therapy, physi-
cal activity, and art and sensory therapy. When making the decision
whether or not to accept support with psychosocial therapies, provid-
ers and individuals with dementia are faced with conflicting evidence
on these therapies because of the heterogeneous studies and partially
poor study designs [52]. The effects measured are often small and not
constant over several studies. Nevertheless, various psychosocial thera-
pies were recommended, because they have shown beneficial effects
[7, 23], for example, cognitive training for depression, cognition, and
quality of life [53]; occupational therapy for apathy, quality of life,
cognition [54], and functional decline [55]; and physical activity for
basic life skills [56]. In addition, support for relatives with the aim of
improving the individuals’ with dementia situation yields effects
regarding behavioral symptoms and depression [51]. Since mental
and physical health of the caregivers is often affected [57], structured
offers can help to prevent illnesses and reduce burden [58]. Persons
with dementia and relatives, which are integrated in dementia net-
works, are more frequently in contact with physicians and get more
frequently drug-free therapy and psychosocial interventions [59].
4.2 Legal Issues Over the course of the disease, individuals with dementia lose their
capacity to conclude legal transactions or to provide a declaration of
will. Therefore, all legal issues that could be of importance in the
early or later stages of AD should be discussed with persons with
dementia as long as they are definitely capable to do so. Written
powers and advance directives help persons of authority to make
arrangements in lieu of the individual with dementia in later stages
of the disease and avoid lengthy legal disputes. However, it is advis-
able to prove and certify the individual’s with dementia ability to
make a will even in early stages of the disease by a medical specialist,
mostly a psychiatrist.
Shared Decision-Making and Important Medical and Social Decisions 265
4.2.1 Healthcare Proxy, After diagnosis of AD, it is the right time, according to experts, to
Last Will, Advance settle legal issues [8], for example, to arrange the legacy; the
Healthcare Directive, healthcare proxy, which designates a person for arranging personal
and Advance Care Planning and economic affairs, if AD is progressing; and the advance health-
care directive, which among others defines the extent of medical
procedures acceptable to the person with dementia. Because of the
increased accident risk of dementia, it may be advisable to contract
homeowner’s insurance or to request a disabled person’s pass,
which in some countries includes financial relief [60] and advanced
worker protection [61].
Again, last will, healthcare proxy, and advance healthcare directives
can only be done by persons who are still capable to consent and who
can sign personally.
In recent years the concept of advance care planning (ACP)
[62] has gained more and more attention. It addresses persons in
critical stages of illness and may remedy shortcomings of advance
healthcare directives, which persons often feel overburdened to
complete. Moreover, an advance healthcare directive is often
vaguely formulated; it may not be available at the critical moment
and therefore will be disregarded. ACP provides personal assis-
tance and support in filling in individual forms. In addition an
emergency guide is linked, which specifies how to act in a case of
emergency [63].
4.2.2 Car Driving Another important “legal” issue is car driving. Here the persons’
with dementia wish for autonomy may conflict with his or her abil-
ity to keep driving motor vehicles. Although AD at early stages is
not necessarily associated with the loss of capacity to drive [64],
reaction capacity and speed estimation can be affected. In some
European countries [65] there are obligatory regular medical
examinations at a certain age, which confirm or reject the ability to
drive. In other countries the responsibility is with persons con-
cerned, their relatives, or their physicians. Here individuals with
dementia should be informed by their physicians at the time of
diagnosis that AD leads to loss of the driving ability during the
course of the disease. In case of doubt, a medical-psychological
assessment can give some guidance in the decision-making process.
If AD progresses and individuals’ ability to drive is impaired, it may
be required that another person informs authorities to revoke the
persons’ with dementia driving license.
4.3 Social Issues and The progression of AD symptoms raises a lot of social issues.
Coping with Illness After diagnosis the question arises, how individuals with AD
and their relatives will deal with the illness. Many persons have
4.3.1 Coping with Illness
the desire to “carry on as always” [66] and not to further look
into AD. This also touches the issue of informing the social
environment about the disease, which individuals with demen-
tia want to impart or not. Many individuals with dementia and
266 Katharina Bronner and Johannes Hamann
4.3.2 Assistance During the course of the disease, individuals with dementia lose
by Family Members more and more abilities and become dependent on support. In
and External Help most cases, relatives take over the additional assistance needed,
which is both expected and claimed from persons with dementia
and is taken for granted from relatives, who adapt their living con-
ditions for that. Both sides rather disapprove the possibility of
external help. In the course of AD, many caring relatives become
overburdened with this situation [8], have a worse quality of life,
risk their own health, and cannot ensure adequate care with the
best will in the world. Therefore professionals in dementia recom-
mend to plan external help and to extend the social network as
early as possible [69].
4.3.3 Living Conditions The issue of keeping up living conditions (especially housing condi-
tions) or changing them is also a difficult one, which from the point
of view of professionals in dementia is often repressed and ignored.
Most individuals with AD express the wish to stay in their familiar
environment as long as possible. Their relatives confirm this and
often promise that individuals shall stay at home even in the later
stages of AD [8]. During the course of AD, living at home may,
however, become more and more difficult, especially for persons
with dementia living alone without families or lack of assistance by
family. Therefore, persons with dementia and their relatives should
timely be informed about alternative living conditions such as
assisted community living or care homes for dementia.
6 Conclusions
References
tives of people with Alzheimer’s disease, spouses 22. Okonkwo OC et al (2008) Medical decision-
and professionals? BMC Res Notes 9:149 making capacity in mild cognitive impair-
9. Charles C et al (1997) Shared decision-making ment: a 3-year longitudinal study. Neurology
in the medical encounter: what does it mean? 71:1474–1480
(or it takes at least two to tango). Soc Sci 23. NICE (2006) Dementia: supporting people with
44:681–692 dementia and their carers in health and social
10. Emanuel EJ, Emanuel LL (1992) Four mod- care: NICE Clinical Guideline [CG42]. https://
els of the physician-patient relationship. JAMA www.nice.org.uk/guidance/cg42/resources/
267:2221–2226 dementia-supporting-people-with-dementia-
11. Charles C et al (1999) What do we mean by and-their-carers-in-health-and-social-care-
partnership in making decisions about treat- pdf-975443665093. Accessed 27 April 2017
ment? BMJ 319:780–782 24. Alzheimer’s Association Early-Stage Initiative
12. Coulter A et al (2017) Shared decision making (2006) History of the Alzheimer’s associa-
in the UK: moving towards wider uptake. Z Evid tion early-stage initiative. http://www.alz.
Fortbild Qual Gesundhwes 123- 124:99–103. org/documents/national/EDInitiative.pdf.
https://doi.org/10.1016/j.zefq.2017.05.010 Accessed 27 April 2017
13. Spatz ES et al (2017) Shared decision making 25. Alzheimer Europe (2006) Paris declaration
as part of value based care: new U.S. policies 2006. http://www.alzheimer-europe.org/
challenge our readiness. Z Evid Fortbild Qual Policy-in-Practice2/Paris-Declaration-2006.
Gesundhwes 123-124:104–108. https://doi. Accessed 27 April 2017
org/10.1016/j.zefq.2017.05.012 26. Moye J et al (2006) Neuropsychological pre-
14. Martin H et al (2017) The long way of imple- dictors of decision-making capacity over 9
menting patient-centered care and shared months in mild-to-moderate dementia. J Gen
decision-making in Germany. Z Evid Fortbild Intern Med 21:78–83
Qual Gesundhwes 123-124:46–51. https:// 27. Hirschman KB et al (2005) Do Alzheimer’s
doi.org/10.1016/j.zefq.2017.05.006 disease patients want to participate in a treat-
15. Stacey D et al (2017) Decision aids for people ment decision, and would their caregivers let
facing health treatment or screening decisions. them? Gerontologist 45:381–388
Cochrane Database Syst Rev 4:CD001431 28. Hamann J et al (2011) Patient participation in
16. Hamann J et al (2006) Partizipative medical and social decisions in Alzheimer’s dis-
Entscheidungsfindung. Implikationen des ease. J Am Geriatr Soc 59:2045–2052
Modells “Shared Decision Making” für 29. Hamann J et al (2009) Psychiatrists’ use of
Psychiatrie und Neurologie. Nervenarzt 9: shared decision making in the treatment of
1071–1078 schizophrenia: patient characteristics and deci-
17. Reichhart T et al (2008) Patientenbeteiligung sion topics. Psychiatr Serv 60:1107–1112
in der Psychiatrie–eine kritische 30. Vollmann J et al (2004) Mental compe-
Bestandsaufnahme. Psychiatr Prax 35:111–121 tence and neuropsychologic impairments in
18. NICE (2009) Depression in adults: rec- demented patients. Nervenarzt 75:29–35
ognition and management: NICE Clinical 31. Karlawish JHT et al (2000) Caregivers’ pref-
Guideline [CG90]. https://www.nice.org. erences for the treatment of patients with
uk/guidance/cg90/resources/depression- Alzheimer’s disease. Neurology 55:1008–1014
in-a dults-r ecognition-and-management- 32. Karlawish JHT et al (2002) Relationship
pdf-975742636741. Accessed 27 April 2017 between Alzheimer’s disease severity and patient
19. NICE (2014) Psychosis and schizophrenia in participation in decision about their medical
adults: prevention and management: NICE care. J Geriatr Psychiatry Neurol 15:68–72
Clinical Gudeline [CG178]. https://www.nice. 33. Peterson RC et al (2001) Current concepts
org.uk/guidance/cg178/resources/psychosis- in mild cognitive impairment. Arch Neurol
and-schizophrenia-in-adults-prevention-and- 58:1985–1992
management-pdf-35109758952133. Accessed 34. Raschetti R et al (2007) Cholinesterase inhibi-
27 April 2017 tors in mild cognitive impairment: a systematic
20. Hamann J, Heres S (2014) Adapting shared review of randomised trials. PLoS Med 4:e338
decision making for individuals with severe 35. DeKosky ST (2008) Ginkgo Biloba for pre-
mental illness. Psychiatr Serv 65:1483–1486 vention of dementia: a randomized controlled
21. Hirschman KB et al (2004) How does an trial. JAMA 300:2253–2262
Alzheimer’s disease patients’ role in medical 36. Rosen C et al (2013) Fluid biomarkers in
decision making change over time? J Geriatr Alzheimer’s disease–current concepts. Mol
Psychiatry Neurol 17:55–60 Neurodegener 8:20
Shared Decision-Making and Important Medical and Social Decisions 269
37. Kivipelto M, Soloman A (2008) Alzheimer’s 51. Alzheimer Europe (2016) Clinical trials watch.
disease–the ways of prevention. J Nutr Health http://www.alzheimer-europe.org/Research/
Aging 12:89S–94S Clinical-Trials-Watch. Accessed 3 May 2017
38. Alonso A et al (2009) Cardiovascular risk fac- 52. Institut für Qualität und Wirtschaftlichkeit
tors and dementia mortality: 40 years of fol- im Gesundheitswesen (IQWiG) (2009)
low-up in the seven countries study. J Neurol Nichtmedikamentöse Behandlung der
Sci 280:79–83 Alzheimer Demenz. Abschlussbericht A05-
39. Feart C et al (2009) Adherence to a 19D(Version 1.0 Stand: 13.01.2009), Köln
Mediterranean diet, cognitive decline, and risk 53. Pinquart M, Forstmeier S (2012) Effects of
of dementia. JAMA 302:638–648 reminiscence interventions on psychosocial
40. Scarmeas N et al (2009) Physical activity, outcomes: a meta-analysis. Aging Ment Health
diet, and risk of Alzheimer disease. JAMA 16:541–558
302:627–637 54. Korczak D, Habermann C, Braz S (2013) The
41. Sindi S, Mangialasche F, Kivipelto M (2015) effectiveness of occupational therapy for per-
Advances in the prevention of Alzheimer’s dis- sons with moderate and severe dementia. GMS
ease. F1000Prime Rep 7:50. 10.12703/P7-50 Health Technol Assess 9:1–7
42. Marckmann G, in der Schmitten J (2014) 55. McLaren AN, Lamantia MA, Callahan CM
Krebsfrüherkennung aus Sicht der Public- (2013) Systematic review of non-pharmacologic
Health-Ethik. Bundesgesundheitsbl interventions to delay functional decline in
57:327. https://doi.org/10.1007/ community-dwelling patients with dementia.
s00103-013-1913-0 Aging Ment Health 17:655–666
43. Williamson J, Goldman J, Marder KS 56. Forbes D et al (2013) Exercise programs for
(2009) Genetic aspects of Alzheimer disease. people with dementia. Cochrane Database Syst
Neurologist 15:80–86 Rev 12:CD006489
44. Jessen F, Dodel R (2014) Prädiktion 57. Schölzel-Dorenbos CJ et al (2009) Quality of
der Alzheimer Demenz. Nervenarzt 85: life and burden of spouses of Alzheimer dis-
1233–1237 ease patients. Alzheimer Dis Assoc Disord
45. Quian W et al (2016) Misdiagnosis of 23:171–177
Alzheimer’s disease: inconsistencies between 58. Chien LY et al (2011) Caregiver support groups
clinical diagnosis. Alzheimers Dement 12:293 in patients with dementia: a meta-analysis. Int
46. Deutsche Alzheimer Gesellschaft (2013) J Geriatr Psychiatry 26:1089–1098
Empfehlungen zum Umgang mit 59. Wolf-Ostermann K et al (2017) Users
Frühdiagnostik bei Demenz. https://www. of regional dementia care networks in
deutsche-alzheimer.de/fileadmin/alz/pdf/ Germany: first results of the evaluation study
empfehlungen/empfehlungen_fruehdiagnos- DemNet-D. Z Gerontol Geriatr 50:21–27
tik_demenz_dalzg.pdf. Accessed 2 May 2017 60. Sozialgesetzbuch (SGB) Neuntes Buch
47. Robert PH et al (2005) Grouping for behav- (IX) - Rehabilitation und Teilhabe behinder-
ioral and psychological symptoms in dementia: ter Menschen - (Artikel 1 des Gesetzes
clinical and biological aspects. Consensus paper v. 19.6.2001, BGBl. I S. 1046) § 145
of the European Alzheimer disease consortium. Unentgeltliche Beförderung, Anspruch auf
Eur Psychiatry 20:490–496 Erstattung der Fahrgeldausfälle
48. Winblad B et al (2006) 3-year study of done- 61. Sozialgesetzbuch (SGB) Neuntes Buch
pezil therapy in Alzheimer’s disease: effects of (IX) - Rehabilitation und Teilhabe behinder-
early and continuous therapy. Dement Geriatr ter Menschen - (Artikel 1 des Gesetzes v.
Cogn Disord 21:353–363 19.6.2001, BGBl. I S. 1046) § 85 Erfordernis
49. NICE (2011) Donepezil, galantamine, riv- der Zustimmung
astigmine and memantine for the treat- 62. Emanuel LL, von Gunten CF, Ferris FD
ment of Alzheimer’s disease. Technology (2000) Advance care planning. Arch Fam Med
appraisal guidance [TA217]. https://www. 9:1181–1187
nice.org.uk/guidance/ta217/resources/ 63. Coors M, Jox R, in der Schmitten J (Hrsg)
donepezil-galantamine-rivastigmine-and- (2015) Advance care planning: Von der
memantine-for-the-treatment-of-alzheimers- Patientenverfügung zur gesundheitlichen
disease-pdf-82600254699973. Accessed 2 Vorausplanung. Kohlhammer, Stuttgart
May 2017 64. Wolter DK (2014) Beginnende Demenz und
50. McShane R, Areosa Sastre A, Minakaran N Fahreignung. Teil 2: Das Assessment und seine
(2006) Memantine for dementia. Cochrane praktischen Konsequenzen. Z Gerontol Geriat
Database Syst Rev 19:CD003154 47:345–355
270 Katharina Bronner and Johannes Hamann
65. Fastenmeier W, Gstalter H (2014) Fahreignung dementia patients: a focus group study on gen-
älterer Kraftfahrer im internationalen Vergleich. eral practitioners’ views and experiences. PLoS
Literaturrecherche, Analyse und Bewertung. One 9:e84905
Forschungsbericht Nr. 25. Unfallforschung der 69. Callahan CM et al (2009) Integrating care for
Versicherer, Berlin older adults with cognitive impairment. Curr
66. Von Kutzleben M et al (2012) Community- Alzheimer Res 6:368–374
dwelling persons with dementia: what do they 70. Morant N, Kaminskiy E, Ramon S (2016)
need? What do they demand? What do they Shared decision making for psychiatric medi-
do? A systematic review on the subjective expe- cation management: beyond the micro-social.
riences of persons with dementia. Aging Ment Health Expect 19:1002–1014
Health 16:378–390 71. Towle A, Godolphin W (1999) Framework for
67. Hirschman KB, Kapo JM, Karlawish JH (2008) teaching and learning informed shared decision
Identifying the factors that facilitate or hinder making. BMJ 319:766–771
advance planning by persons with dementia. 72. O'Connor AM et al (2009) Decision aids
Alzheimer Dis Assoc Disord 22:293–298 for people facing health treatment or screen-
68. DeVleminck A et al (2014) Barriers to advance ing decisions. Cochrane Database Syst Rev
care planning in cancer, heart failure and 8:CD001431
Index
A D
Aging���������������������������������15, 16, 18–21, 23–26, 31–35, 45–47, Default mode network (DMN)���������������� 124, 129, 131, 132,
64, 77, 100, 119–121, 127, 129–133, 135, 138, 141, 180, 134–136, 139, 140
214, 226, 237, 243 Dementia����������������������������3, 15, 45, 55, 75, 93, 95, 107, 112,
Alpha linolenic acid (ALA)������������������������������������������ 18, 19 120, 149, 150, 152, 175, 179–181, 189, 193, 213, 214,
Alzheimer’s disease (AD)������������������������� 3–5, 15, 71, 74–76, 236, 249, 250, 254, 259, 260
93, 107, 119, 151, 154, 157–164, 169, 179, 189–196, Dementia subtypes����������������������������������������������������� 95, 115
199–210, 213, 214, 235–245, 249, 259 Diagnosis�������������������� 5, 16, 18, 55, 58, 72, 76, 79, 80, 87, 88,
Amyloid-beta��������������������������������72–75, 82, 86, 87, 93, 149, 93, 100, 101, 115, 141, 150–152, 155, 179, 181, 185,
155, 180, 189, 190, 250–252, 254 235–239, 241, 242, 245, 249, 259
Amyloid precursor protein (APP)��������������������������73, 74, 76, Diet���������������������������������������������������������������������� 15, 17, 240
88, 179–186, 238–239 Diffusion tensor imaging (DTI)������������������������� 27, 114, 115
Apolipoprotein E (APOE)������������������������� 16, 19–21, 24, 26, Disease classification����������������������������������������������������71–88
27, 120, 126, 127, 133–137, 139, 140, 152, 155, 236, Disease staging��������������������������������������������������������� 190, 243
239–241, 252, 254 DNA����������������������������������������������������������������������������������22
Drug development������������������������������������������������������������264
B
E
Biomarkers����������������������������������18, 26, 27, 31–33, 35, 46, 93, 95,
99–104, 114–115, 120–122, 128, 132, 134, 137, 138, 141, Early diagnosis����������������������������������������������4, 151, 250, 259
154–155, 157, 169, 180–183, 185, 191, 200, 201, 207, Economic burden��������������������������������������������������������� 4, 5, 9
208, 214, 216, 226–228, 236, 237, 242–245, 249, 251 Eicosapentaenoic acid (EPA)�������������������������������������� 18, 19,
Blood plasma���������������������������������������������������� 181, 183, 192 27, 31, 34
Blood serum��������������������������������������������������������������� 20, 185 Environment����������������������������������� 16, 67, 99, 100, 265, 266
Blood vessel�����������������������������������������������199, 202–204, 208 Enzyme-linked immunosorbent assay (ELISA)���������� 96, 97,
Brain������������������������������3, 45, 63–65, 67, 68, 71, 75, 77–80, 83, 99, 102, 169, 183–185
85, 86, 93, 100, 107–115, 119–141, 149–155, 160, 171, Epidemiology���������������������������������������������������������������15–36
180, 190, 199, 200, 214, 218, 236, 241, 250, 262 Ethics��������������������������������������������������������171, 237, 249–256
Brain reserve������������������������������������������������������� 17, 254, 255
F
C
F-fluorodeoxyglucose������������������������������������������������� 27, 29
18
Robert Perneczky (ed.), Biomarkers for Preclinical Alzheimer’s Disease, Neuromethods, vol. 137,
https://doi.org/10.1007/978-1-4939-7674-4, © Springer Science+Business Media, LLC 2018
271
Biomarkers for Preclinical Alzheimer’s Disease
272 Index
L Postmortem issue���������������������������������������191, 217, 241, 243
Predictive modeling���������������������������������������������������� 26, 110
Lifestyle����������������������������������� 16, 17, 25, 33, 54, 58, 68, 141, Predictor�������������������������������������������� 107, 115, 128, 180, 216
240, 252, 253, 256 Prevention����������������������������������� 9, 16, 17, 22, 23, 26, 34, 35,
Luminex xMAP�����������������������������������������������������������������98 114, 236, 240, 253, 262
Prognosis���������������������������������������������������100, 101, 254, 255
M
Protective factors������������������������������������������������������ 127, 255
Magnetic resonance imaging (MRI)������������������20, 27, 31–33, Proteomics����������������������������������������������������������������169–171
107–110, 112, 113, 115, 119, 121–125, 130, 132–140,
153, 154, 162, 180, 214–228, 237, 244, 245, 250, 252 Q
Mild cognitive impairment (MCI)�������������������������24, 45, 94, Quantitative electroencephalography�������������������������������157
120, 136, 151, 179, 194, 201, 216, 238, 249, 262
Morphometry���������������������107, 110, 114, 214, 221–223, 225 R
Multivariate models���������������������������������������������������� 26, 110
Retinal imaging����������������������������������������������������������������200
N Risk classification�������������������������������������������������������������114
Risk factors�������������������������������� 9, 15, 57, 120, 126, 130, 133,
Network analysis��������������������������������������������������������������163 134, 138, 141, 155, 171, 239, 240, 245, 255, 262
Neuritic plaques (NP)�����������������������72, 75, 77, 81, 83, 85, 86
Neurodegeneration��������������������������16, 19, 27, 33, 34, 71–77, S
101, 119, 121, 122, 125, 126, 129, 130, 136–138, 179,
Shared decision-making�������������������������������������������259–267
190–192, 208, 209, 214, 215, 226, 236, 237, 244, 245,
Single nucleotide polymorphism����������������������������������������16
249–251, 254–256
Social networks�����������������������������������������������������������������266
Neuroinflammation���������������������������������������������������� 19, 191
Societal burden���������������������������������������������������������������3–11
Neuropathology������������������������������������ 19, 20, 33, 35, 72–76,
Standardization��������������������������������������������������97, 102, 104,
121, 131, 193, 195
182, 228
Neuropsychology������������������������������������ 46, 51–57, 100, 109,
Subjective cognitive impairment����������������������������������� 46, 47
110, 115, 141, 255, 260
Substantia innominata�������������������������������������� 214, 215, 218
Non-invasive biomarker������������������������������������������� 125, 140
Support vector machine (SVM)���������������������������������������110
Nucleus basalis Meynert (NBM)����������������������213, 216, 218,
Systems biology����������������������������������������������������������������171
220, 226
Nutrition�������������������������������������������������������������� 17–36, 262 T
P Tau�������������������������������������� 16, 72, 83–85, 94, 113, 120, 180,
189, 236, 243, 250
Pattern recognition������������������������������������������� 108, 111–115
TREM2�������������������������������������������������������������������� 171, 172
Physical activity������������������������������������� 17, 54, 240, 253, 264
Polyunsuturated omega-3 fats��������������������������������������18–20 V
Positron-emission-tomography (PET)����������� 27, 33, 63, 115,
120–122, 125–133, 138–141, 149–154, 180, 190, 191, Validation����������������������������������������32, 53, 97, 104, 108, 111,
195, 216, 227, 228, 236, 237, 241–245, 250, 253, 254 113, 158, 169, 185, 238