PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Similar documents
PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Guidance for Industry Alzheimer s Disease: Developing Drugs for the Treatment of Early Stage Disease

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Biomarkers for Alzheimer's Disease in Down Syndrome

CRITERIA FOR AD DEMENTIA June 11, 2010

Cholinesterase inhibitors and memantine use for Alzheimer s disease TOPIC REVIEW

Victims Compensation Claim Status of All Pending Claims and Claims Decided Within the Last Three Years

The diagnosis of dementia due to Alzheimer s disease: Recommendations from the National Institute on Aging and the Alzheimer s Association workgroup

Primary Endpoints in Alzheimer s Dementia

MCDB 4777/5777 Molecular Neurobiology Lecture 38 Alzheimer s Disease

THEORIES OF NEUROLOGICAL AGING AND DEMENTIA

CEREBROSPINAL FLUID MARKERS FOR THE EARLY AND DIFFERENTIAL DIAGNOSIS OF ALZHEIMER S DISEASE

Alzheimer s & Dementia 7 (2011)

NEW CRITERIA AND GUIDELINES FOR THE DIAGNOSIS OF ALZHEIMER S DISEASE PUBLISHED FOR FIRST TIME IN 27 YEARS

Clinical Features of Mild Cognitive Impairment and Dementia in a Community: An update of the Osaki-Tajiri Project

Primary Care Update January 28 & 29, 2016 Alzheimer s Disease and Mild Cognitive Impairment

2012 Medical School for Actuaries Nov. 6-7, 2012 Session #1: Alzheimer s Disease

Graphical tasks to aid in diagnosing and monitoring Parkinson s disease

Subject Review. p.17 Alzheimer s Disease: An Update. p.21 Diabetes Team and Glycemic Control DANIEL A. LLANO, MD, PHD

Parkinson s Disease: Deep Brain Stimulation and FDOPA-PET

Social Security Disability Insurance and young onset dementia: A guide for employers and employees

Dementia Causes and Neuropsychological Evaluation of the Older Adult

RESEARCHERS REPORT NEW WAYS TO PREDICT THE DEVELOPMENT OF ALZHEIMER S DISEASE

Steps to getting a diagnosis: Finding out if it s Alzheimer s Disease.

Copywrite - Eric Freitag, Psy.D., 2012

Opening the psychological black box in genetic counseling

Chapter 10. Summary & Future perspectives

Local Clinical Trials

M.A.Lips. Copyright 2014, M.A.Lips, Amsterdam, the Netherlands

Running Head: THE DETECTION OF ALZHEIMER S DISEASE: A LITERATURE 1. The Detection of Alzheimer s Disease: A Literature Review and Case Study

Sandro Sorbi DIPARTIMENTO DI SCIENZE NEUROLOGICHE E PSICHIATRICHE

Diseases of the Nervous System. Neal G. Simon, Ph.D. Professor, Dept of Biological Sciences Lehigh University

Diagnostik der Zukunft: Wissen wir mit Proteomik und Genomik wirklich mehr?

The Effect of Age and Education Transformations on Neuropsychological Test Scores of Persons With Diffuse or Bilateral Brain Damage 1

In conjunction with clinical history, structural

Childhood onset MS and MS during Pregnancy

Alzheimer s disease. What is Alzheimer s disease?

Stefano F. Cappa Department of Neuroclinical Sciences Vita Salute University and San Raffaele Scientific Institute Milan, Italy

Implantable Cardioverter Defibrillator Treatment: Benefits and Pitfalls in the Currently Indicated Population. Carel Jan Willem Borleffs

2016 Programs & Information

Stephen L. Benson, Psy.D. November 17, 2015

Early Changes in the Brain Proteome Associated with Alzheimer's Disease Risk

Update on Treatment of the Dementias

Cutaneous B-cell lymphoma: classification, prognostic factors and management recommendations. Nancy J. Senff

2014 Alzheimer s Disease Facts and Figures

Is there a Distinct Phenotype to Memory Loss in Alzheimer's Disease?

The Role of Neuropsychological Testing in Guiding Decision- Making Related to Dementia

Small artery tone under control of the endothelium

Objectives. Aging and Forgetfulness Define Dementia Types of Dementia Treatment

Normal and Abnormal Aging and the Brain. Joel Kramer, PsyD Saul Villeda, PhD Kristine Yaffe, MD

Dementia Episodes of Care

Clinical Applications of Imaging to Drug Development

Functional neuroinflammatoryand serotonergic imaging in Alzheimer s disease: validation and preliminary clinical findings

Linguistic Ability in Early Life and the Neuropathology of Alzheimer s Disease and Cerebrovascular Disease

PREDICTION AND PREVENTION OF DEMENTIA: NEW HOPE SECOND INTERNATIONAL MEETING OF THE MILAN CENTER (NEUROMI) MILAN, JULY 6-8, 2016 FOR NEUROSCIENCE

The Concept of Cognitive Reserve: A Catalyst for Research

Title: Apolipoprotein E 4 accelerates dementia and increases cerebrospinal fluid tau levels in Alzheimer's disease

Early detection of dementia and the Initial-phase intensive support team for preventing BPSD.

J/601/2874. This unit must be assessed in accordance with Skills for Care and Development s QCF Assessment Principles.

Changes affecting concentration,

Use of anti-dementia drugs and delayed care home placement: an observational study

1 st December Cardiff Crown Court. Dear. Claimant: Maurice Kirk Date of Birth: 12 th March 1945

Preventing Dementia: The Depression-Diabetes Nexus

Summary chapter 2 chapter 2

Objectives. Evaluation of Memory Loss. Cognitive Impairment. Clinical Questions. Medicare Wellness Visit

Cognitive Testing for Underwriting Life Insurance

Placement of dementia sufferers in residential and nursing home care

What is PD? Dr Catherine Dotchin MD MRCP Consultant Geriatrician

1 in 3 seniors dies with Alzheimer s or another dementia.

GOING BEYOND RISK REDUCTION: PHYSICAL EXERCISE MAY BE AN EFFECTIVE TREATMENT FOR ALZHEIMER S DISEASE AND VASCULAR DEMENTIA

Stem cells for brain cures

2.1 Who first described NMO?

Treating patients with compounds or biological agents that pass the safety-test

PRACTICE PARAMETER: DIAGNOSIS OF DEMENTIA (AN EVIDENCE-BASED REVIEW)

Measures of diagnostic accuracy: basic definitions

UNDERSTANDING MIND AND BRAIN RESEARCH MASTER S PROGRAMME IN COGNITIVE NEUROPSYCHOLOGY

Polyphenols in the Prevention of Alzheimer s Disease:

2015 Alzheimer s Disease Facts and Figures

Electrohypersensitivity and multiple chemical sensitivity: two clinicbiological. disorder?

Clinically isolated syndrome (CIS)

Mental health issues in the elderly. January 28th 2008 Presented by Éric R. Thériault

The Need for a PARP in vivo Pharmacodynamic Assay

Montreal Cognitive Assessment (MoCA) Debbie Froese, B.M.R.-O.T., B.A. Christine Knight, Ph.D.,R.Psych.

A Support System for Diagnosis of Dementia, Alzheimer or Mild Cognitive Impairment

ß-interferon and. ABN Guidelines for 2007 Treatment of Multiple Sclerosis with. Glatiramer Acetate

Bedside cognitive examination beyond the MMSE. Dr Richard Perry Dept of Neurosciences Imperial College

Regional Brain Atrophy Rate Predicts Future Cognitive Decline: 6-year Longitudinal MR Imaging Study of Normal Aging 1

KIDNEY FUNCTION RELATION TO SIZE OF THE TUMOR IN RENAL CELL CANCINOMA

PARTNERING WITH YOUR DOCTOR:

Age-Related Incidence and Family History in Frontotemporal Dementia: Data from the Swedish Dementia Registry

The Genetic Basis of Neurological Disorders

Emergency Room Treatment of Psychosis

Dr V. J. Brown. Neuroscience (see Biomedical Sciences) History, Philosophy, Social Anthropology, Theological Studies.

Diagnosis and Initial Management of Cognitive Disorders

Glioblastoma (cancer affecting the brain) A guide for journalists on glioblastoma and its treatment

Neurobiology of Depression in Relation to ECT. PJ Cowen Department of Psychiatry, University of Oxford

Neuropsychological Testing

Abnormal Psychology PSY-350-TE

Mild cognitive impairment beyond controversies, towards a consensus: report of the International Working Group on Mild Cognitive Impairment

SOCIO-DEMOGRAPHIC RISK FACTORS IN PATIENTS WITH MILD COGNITIVE IMPAIRMENT CONVERTED TO DEMENTIA

Transcription:

PDF hosted at the Radboud Repository of the Radboud University Nijmegen The following full text is a publisher's version. For additional information about this publication click this link. http://hdl.handle.net/2066/92727 Please be advised that this information was generated on 2015-10-21 and may be subject to change.

The ref lection of Alzheimer disease in CSF Petra Elise Spies

Part of the research presented in this thesis was performed within the framework of the Center for Translational Molecular Medicine (www.ctmm.com). Financial support for printing of this thesis was kindly provided by Zambon Nederland B.V., Nutricia Advanced Medical Nutrition, Danone Research Centre for Specialised Nutrition, Novartis Pharma B.V., Lundbeck B.V., Luminex B.V., Janssen B.V., Internationale Stichting Alzheimer Onderzoek, Crossbeta Biosciences (Utrecht) Developing Therapeutics for Alzheimer s Disease, Alzheimer Nederland (Bunnik). ISBN 978-94-91027-23-9 Design cover and lay out by In Zicht Grafisch Ontwerp, Arnhem Printed and bound by PrintPartners Ipskamp, Enschede 2012 P.E. Spies All rights reserved. No parts of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopy, recording or otherwise, without prior written permission of the author.

The reflection of Alzheimer disease in CSF Een wetenschappelijke proeve op het gebied van de Medische Wetenschappen Proefschrift ter verkrijging van de graad van doctor aan de Radboud Universiteit Nijmegen op gezag van de rector magnificus prof. mr. S.C.J.J. Kortmann, volgens besluit van het college van decanen in het openbaar te verdedigen op donderdag 15 maart 2012 om 10.30 uur precies door Petra Elise Spies geboren op 14 januari 1983 te Tiel

Promotor Prof. dr. M.G.M. Olde Rikkert Copromotoren Dr. ir. M.M. Verbeek Dr. J.A.H.R. Claassen Manuscriptcommissie Prof. dr. P. Wesseling Prof. dr. G.J.M. Martens Prof. dr. P.M.M. Bossuyt (Academic Medical Center - University of Amsterdam)

The reflection of Alzheimer disease in CSF An academic essay in Medical Sciences Doctoral thesis to obtain the degree of doctor from Radboud University Nijmegen on the authority of the Rector Magnificus prof. dr. S.C.J.J. Kortmann, according to the decision of the Council of Deans to be defended in public on Thursday March 15, 2012 at 10.30 hours by Petra Elise Spies born on January 14, 1983 in Tiel

Supervisor Prof. dr. M.G.M. Olde Rikkert Co-supervisors Dr. ir. M.M. Verbeek Dr. J.A.H.R. Claassen Doctoral Thesis Committee Prof. dr. P. Wesseling Prof. dr. G.J.M. Martens Prof. dr. P.M.M. Bossuyt (Academic Medical Center - University of Amsterdam)

Contents Prologue 9 Chapter One Introduction and Outline 15 Part One CSF biomarkers in the differential diagnosis of dementia Chapter Two The CSF Aβ 42 /Aβ 40 ratio 39 Chapter Three CSF α-synuclein 55 Chapter Four The correlation between CSF α-synuclein and CSF Aβ 69 Chapter Five Experiences with CSF analysis in memory clinics 77 Chapter Six A prediction model to calculate the probability of AD 91 Part Two CSF biomarkers in pathophysiological hypotheses Chapter Seven Dynamic hypotheses 113 Chapter Eight The correlation between CSF biomarkers 119 and cognitive functioning Chapter Nine AD and cerebrovascular disease 131 Chapter Ten Why is CSF Aβ 42 decreased in AD? 145 Chapter Eleven Summary and general discussion 165 Chapter Twelve Nederlandse samenvatting 185 Dankwoord 195 Curriculum Vitae 202 List of publications 203 Donders Graduate School for Cognitive Neuroscience Series 204

Prologue On elderly chaps and ferocious Orcs This chapter has been published as: Elderly: a term to avoid or to embrace? Petra E. Spies, Jurgen A.H.R. Claassen, J Am Geriatr Soc 2011;59:1986-7 Prologue Prologue

On elderly chaps and ferocious Orcs With the surge in articles that report research on aging, the question has risen whether the commonly used elderly is an appropriate term to describe old people. 1,2 A strong plea was even launched to ban this term from medical journals, and replace it by older persons. 3 Three main arguments were put forward: 1) old people reject being called elderly ; 2) elderly may be confused with frail ; 3) the term elderly is pejorative, and its use thus reflects negatively on older patients as well as on health care for the old. 2,3 We question the validity of these arguments. Old people reject being called elderly This argument was based on a European survey on preferential terminology for older citizens, conducted in 12 countries. 4 The multi-linguistic setting was a surprising choice for an investigation of semantic preferences. Noteworthy, only preference, and not rejection, was measured. It remains unclear how linguistic and cultural differences were taken into account. Were participants addressed in their own language? Were the intuitive subtleties of the English language correctly brought across? The Danish for example quite liked being called elderly maybe their translation carries with it a different meaning than the English elderly. In the English-speaking countries, elderly was indeed not a preferred term, but remarkably, neither was older people. Senior (citizen) received most votes. In contrast, the American media guide issued by The International Longevity Center advised against the use of senior. Apparently, semantic preferences cannot easily be generalized. Elderly is easily confused with frail We find this hard to believe. For example, in the 2001 cinema hit Lord of the Rings, the character Gandalf was described as an elderly chap. Gandalf, who in this film subsequently slays a dragon-like creature as well as several ferocious Orcs, can hardly be considered frail. If the large audience can distinguish elderly from frail, so can and should people who work in the field of aging research. And they do: even though the concept of frailty remains ill-defined and disputed, old age in itself is not considered a criterion by most researchers. Underlining this is the fact that those who work in the field do not shy from the term. When a number of medical journals were scanned for the use of elderly people and older people, it was found that most authors and editors, even in geriatrics, had continued to use elderly in spite of the proposed ban. 1 11

Prologue One may argue that the term elderly has insufficient categorical validity. Obviously, the inherent heterogeneity in the older age group is tremendous, making the use of a single term as a group denominator an easy target for criticism. However, we argue that elderly is as unambiguous a term as child or adult, terms that also cover a wide spectrum and that must be further specified by descriptives such as prepubertal or healthy. Elderly is a pejorative term Is it the term elderly that is responsible for the negative image of older people and of the health services and professionals devoted to their care? Will frenetic avoidance of this term remove this stigma? Naturally, it can be defended that people do not want to be seen as old. Most adults, if told that they look much younger than their date of birth suggests, glow with happiness. When geriatric patients are asked how they get along at day care, they complain that it is full of old people. The unpleasantness of being confronted with one s advanced age may transpire to how any term describing old age is perceived, including elderly. But does that make these terms inappropriate? Calling an 80-year-old elderly, senior or in later life will not change his actual age. Even if one preferred word were found, it would be a matter of time before this new term would carry the stigma of the old one. It would be better to change the stigma and not the word. We should remember that elderly is not the same as frail, that old is not the same as worthless. This message is far more important than the discussion whether elderly or older people feel insulted by either term, or whether health care for the elderly will rise in status if it changes its name to health care for later life. Clinicians and researchers could therefore embrace elderly as the preferred term to describe the old people they care for and investigate. Stemming from the Old English aeld it means somewhat older, a clear and gentle way to describe an old person. Let s stop debating semantics, and start changing a stigma. 12

On elderly chaps and ferocious Orcs References 1. Quinlan N, O Neill D. Older or elderly --are medical journals sensitive to the wishes of older people? J Am Geriatr Soc 2008;56:1983-4. 2. Francis RM. Editor s view. Age Ageing 2010;39:220. 3. Falconer M, O Neill D. Out with the old, elderly, and aged. BMJ 2007;334:316. 4. Walker A. Age and attitudes: main results from a Eurobarometer survey. Brussels: Commission of the European Communities, 1993. 13

Introduction and Outline The case report in this chapter has been published as: Cerebrospinal Fluid Biomarkers in Diagnosing Alzheimer s Disease in Clinical Practice: An Illustration with 3 Case Reports. Diane Slats, Petra E. Spies, Magnus J.C. Sjögren, Frans R. Verhey, Marcel M. Verbeek, Marcel G.M. Olde Rikkert, Case Rep Neurol. 2010;2:5-11 Chapter One Chapter One

Introduction and Outline Dementia Dementia is a cognitive disorder that, by definition, interferes with activities of daily living. 1,2 Different types of dementia exist. Alzheimer disease (AD) is the most prevalent and therefore the best known cause of dementia. This disease will be described in more detail below. Both vascular dementia (VaD) and dementia with Lewy bodies (DLB) are reported to be the second most prevalent types of dementia. 3,4 Patients with vascular dementia present with impairments in multiple cognitive domains, slowness in executive functioning, bradyphrenia, and sometimes focal neurological deficits. Neuro imaging shows vascular lesions such as cerebral infarcts or extensive white matter disease. 5 DLB patients have fluctuating functioning and attention, together with visuospatial dysfunction on neuropsychological examination. They often suffer from visual hallucinations and parkinsonism. Neuropathologically, this disorder is characterized by intraneuronal inclusions composed of α-synuclein, the so-called Lewy bodies. 6 A less common cause of dementia is frontotemporal dementia (FTD). Features are early decline in social interpersonal conduct, emotional blunting, loss of insight, behavioural disorders and impairments in speech and language. 7 The neuropathological substrate of FTD is heterogeneous. Most cases display cellular inclusions of tau or TAR DNA-binding protein-43 (TDP-43), but in some cases, the pathologic protein is unknown. 7-11 Alzheimer disease AD is a progressive disorder that generally affects memory first, but other cognitive domains such as language, perception and praxis are usually affected as well. 2,12 Aloïs Alzheimer was the first to describe the amyloid β (Aβ) plaques and neurofibrillary tangles that are seen upon neuropathological examination. 13 In addition, severe loss of neurons is found, and often at least a mild degree of cerebral amyloid angiopathy (CAA): Aβ deposits in the walls of blood vessels. The Aβ content of amyloid plaques and of these vascular deposits was discovered in the 1980s, the same decade that tau protein and its hyperphosphorylated form were identified in neurofibrillary tangles. 14-18 Aβ is a small protein derived from sequential cleaving of the Aβ precursor protein (APP) by β- and γ-secretase. Several isoforms of Aβ have been identified, but Aβ 42 and Aβ 40 have received most attention. Aβ 40, 40 amino acids long, is the most abundant Aβ peptide, and makes up about 60% of the cerebrospinal fluid (CSF) Aβ concentration. Aβ 42 accounts for approximately 10% of the CSF Aβ concentration in healthy people. 19-21 17

Chapter ONE Two types of Aβ plaques are found in AD patients: diffuse plaques without an amyloid core that contain only Aβ 42, and classic plaques that have an amyloid core and consist of both Aβ 42 and Aβ 40. 22,23 In contrast, the vascular deposits in the walls of leptomeningeal and cortical arteries and less frequently, in capillaries 24 contain mostly Aβ 40. 25 Tau is a protein that stabilizes the microtubules of the neuronal cytoskeleton. In AD, abnormal phosphorylation of tau leads to destabilizing of microtubules and is suggested to obstruct axonal transport, resulting in synaptic dysfunction. 26 Hyperphosphorylated tau accumulates intracellularly and forms neurofibrillary tangles. After deterioration of the neuron, the tangles remain visible as extraneuronal ghost tangles. 26,27 Diagnosing dementia Several criteria sets have been developed that summarize the core features of AD, VaD, FTD and DLB. 2,5-7,12 The Dutch dementia guideline 28 states that a diagnosis of dementia can be made clinically using these criteria. History taking with the patient and a proxy should focus on gaining the information that is confined in the criteria sets. Neuropsychological assessment can aid in determining whether or not a cognitive disorder is present that may indicate dementia. When more diagnostic certainty is desired, for example to differentiate between the different types of dementia, additional examinations are advised such as structural magnetic resonance imaging (MRI) of the brain and CSF analysis. One may wonder whether differentiating between the dementia types (in other words, to establish an etiological diagnosis) really adds important information that extends beyond the knowledge that a dementia is present. As there are no cures available for any of these causes of dementia, what is the point of knowing the underlying cause? There are several reasons that make it important to clarify the cause of dementia. First, AD, VaD, DLB and FTD differ importantly in the types of deficits they induce in cognition and behaviour. Therefore, an adequate etiological diagnosis allows appropriate and specific education, guidance and care for patients and their informal caregivers. 29,30 Second, knowledge on the type of dementia can provide information on a patient s prognosis. For example, DLB patients deteriorate faster than AD patients, with shorter time between onset of cognitive symptoms and death, 31,32 and in FTD patients, behavioural problems are far more dominant than in AD patients. Third, accurately diagnosing the cause of dementia is important for pharmacological management. Cholinesterase inhibitors may not cure the disease, but they can offer 18

Introduction and Outline functional improvement in AD. DLB patients also respond well to cholinesterase inhibitors, but these patients are extremely sensitive to the side effects of neuroleptic drugs. 4,33 Knowing the cause of the dementia therefore helps in deciding which drugs may be beneficial, and which ones should be avoided. A fourth reason to clarify the cause of the dementia is to advance research in this area. For example, an accurate diagnosis is essential to select patients for trials. If a drug is aimed at treating AD, but the patient group is contaminated by patients with other types of dementia, the drug under study may show no overall effect, despite an effect in patients with true AD. Recognizing AD in the very early stages of the disease may help in selecting patients for trials aimed at prevention of the disease. Underlined by the ongoing efforts to redefine them, 2,34,35 the criteria sets that were laid out so many years ago are insufficient to reliably differentiate between dementia types. A comparison of the clinical diagnoses made with these criteria to the neuropathological diagnoses made post-mortem showed that their sensitivity or their specificity is low, depending on the criteria set used and the comparison made. 36-38 Especially mixed pathologies are often not recognized clinically. 36,37,39 One strategy to improve the accuracy of clinical diagnosis is by introducing biomarkers in addition to the clinical criteria. A biomarker, as defined by the National Institute of Health, is a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. 40 The Dutch dementia guideline already suggested the use of biomarkers such as atrophy on MRI or decreased CSF Aβ 42 and increased CSF total tau (t-tau) to gain more certainty about the cause of dementia. This reasoning was explicated in the research criteria for AD as published in 2007, in which positive biomarkers are considered a supportive feature of Alzheimer disease, regardless of whether or not dementia is clinically present. 41,42 Biomarkers for AD The ideal biomarker for AD should have the following properties: it should be able to detect a fundamental feature of AD s neuropathology, it should be validated against neuropathologically confirmed AD cases, it should be precise, reliable, non-invasive, simple to perform and inexpensive. 43 AD biomarkers discovered thus far can be divided into biomarkers of Aβ plaque deposition (CSF Aβ 42, Aβ imaging with positron emission tomography (PET)) and biomarkers of neurodegeneration (structural MRI of the 19

Chapter ONE hippocampus, CSF t-tau and hyperphosphorylated tau (p-tau), fluorodeoxyglucose (FDG)-PET). 44 Hippocampal atrophy, seen on computed tomography (CT), was the first AD biomarker to be established. 45 The involvement of the hippocampus in AD had been recognized a few years earlier, when severe neuronal loss in this region in AD cases was described in a neuropathology study. 46 A visual rating scale was developed for use with CT or MRI that is still in use today. 47 Hippocampal volume on MRI correlates well with AD pathology at neuropathological examination, although hippocampal atrophy can also be found with increasing age and in other diseases such as FTD and hippocampal sclerosis. 48,49 FGD-PET gives a measure of synaptic activity based on glucose metabolism in the brain. In AD, bilateral temporo-parietal hypometabolism is observed that cannot only be explained by brain atrophy. 50 A positive scan correlates with the presence of AD pathology on post-mortem examination. 51,52 CSF biomarkers are derived from the fluid that circulates around the brain and spinal cord and thus can provide information from close to the brain. Measuring Aβ 42, t-tau and p-tau in the CSF succeeded a few years after their discovery 53-57 and it was shown that CSF Aβ 42 is decreased and CSF t-tau and p-tau are increased in AD. 56,58-62 These three biomarkers provide excellent discrimination between AD patients and healthy controls. 63,64 The specificity for discrimination with other dementias is lower, 65,66 but the combination of biomarkers may still help in the discrimination between AD and VaD, 67 DLB 68 and FTD. 69 As with all biochemical analyses, the concentrations of CSF biomarkers vary between different laboratories. 70 This variation makes it difficult to compare the results between laboratories and makes it impossible to provide generally applicable reference values for use in clinical practice. The variation is probably a result of variations in analytical procedures and batch-to-batch variation in the biomarker assays. Laboratories therefore establish their own reference values. 64,71 The Alzheimer s Association is currently executing a quality control programme that aims to standardize CSF biomarker measurements in both research and clinical laboratories. 72 Aβ imaging is thus far only advocated for research purposes. 2 11 C-Pittsburgh compound B (PiB) binds to fibrillar Aβ in the brain and can be visualized using nuclear imaging. Because this technique is relatively new, cases with post-mortem confirmation of the PiB-PET findings are still limited. Some cases have shown a positive correlation between PiB-binding and fibrillar Aβ deposition, others showed clear Aβ deposition upon neuropathological examination but a negative PiB-PET scan during life. 73-77 20

Introduction and Outline Furthermore, it has been shown that 33% of control subjects had positive PiB-PET scans and that PiB-retention increased in elderly without dementia that were followed longitudinally. 78,79 The clinical relevance of these findings is as yet unclear and longer follow up is needed to determine whether these are preclinical cases of AD or whether these cases should be considered as false positive scans in truly healthy controls. Despite the availability of this variety of biomarkers, differentiating between dementia disorders in clinical practice can still be a challenge, as is shown in the following case report. Case report A 63-year-old woman was referred to our memory clinic for a second opinion. The past two years she had become increasingly anxious in new situations. She had difficulty finding words and engaging in conversations. In 2007, a neurologist identified no cognitive disorder. At that time, she was diagnosed by a psychiatrist as having dysthymic disorder and post-traumatic stress disorder. She was treated with venlafaxine 75 mg once a day. When she presented to our memory clinic nine months later, her complaints had not disappeared. The difficulty in engaging in conversations was still present and she did not manage to finish daily chores because she kept forgetting what she was doing. The anxiety had made her give up driving, internet banking and using the phone and video recorder. There was severe loss of initiative. Her Mini Mental State Examination (MMSE) score was 21 out of 30. 80 The score on the Revised Cambridge Cognitive Examination (CAMCOG-R), a multiple-domain cognitive assessment, was 68 out of 104 (cut-off adjusted for age and education 83). 81,82 She scored 2 out of 15 on the geriatric depression scale (GDS) (a score above 7 indicates depression). 83 These results suggested cognitive decline, possibly AD, since no other underlying disease was apparent, and history taking and GDS score indicated that her depressive mood was in remission. An MRI, made in 2007, was re-evaluated and showed asymmetrical frontotemporal atrophy without medial temporal lobe atrophy (Figure 1). A lumbar puncture was performed, because the screening neuropsychological assessment and the MRI were not fully conclusive and left the possibility of a diagnosis of AD as well as FTD. CSF analysis showed normal concentrations of all biomarkers [reference values] 64 : Aβ 42 560 pg/ml [>500 pg/ml]; t-tau 335 pg/ml [<350 pg/ml]; p-tau 77 pg/ml [<85 pg/ml]. These results made AD less likely. Specific neuropsychological testing was performed, which showed below-average memory learning curves with unimpaired delayed recall and recognition, and a decline in attention and executive functions with reduced information processing 21

Chapter ONE Figure 1 Transversal T2-FLAIR showing frontal cortical and asymmetrical left temporal lobe atrophy speed and apathy. Apart from below-average word fluency, there were no language disorders. The combination of MRI, CSF biomarker results and neuropsychological examination at that moment resulted in the diagnosis FTD. Fifteen months later the patient had deteriorated mildly. Her MMSE was 18 out of 30. No behavioural problems had occurred and her mood was stable. This clinical follow up is atypical for FTD and AD cannot be ruled out. CSF biomarkers in clinical practice As illustrated by this case report, additional examinations to collect biomarkers do not always result in a clear-cut diagnosis. CSF analysis showed that biomarker concentrations were all within the pre-determined normal range, but clinical follow up still left room for AD. It is worth noticing that all CSF biomarker concentrations were close to the cut-off values. It has been suggested that in patients with a naturally occurring high Aβ load, a decrease in Aβ 42 with AD may not be as clear. 84 Relating Aβ 42 to Aβ 40, the latter as a measure of the total Aβ load, was suggested to provide a more precise reflection of the decrease in Aβ 42. This ratio could be a means to improve the accuracy of the existing biomarker CSF Aβ 42. Questions like these fuelled us to start this clinical diagnostic 22

Introduction and Outline research. Whether this Aβ 42 /Aβ 40 ratio can help us to differentiate between dementia disorders, alone and in combination with p-tau and t-tau, was addressed in Chapter Two. Besides improving the accuracy of existing biomarkers, differentiating between dementia disorders may also be improved if new, specific biomarkers are found. Most research has focused on biomarkers for AD, however, if hallmarks of other dementias could be measured, these could serve as biomarkers for that type of dementia. An example of such a hallmark is α-synuclein, the protein found to accumulate in DLB. It was isolated from brain tissue in 1993 85 and was identified in CSF ten years later. 86 Chapter Three and Four describe the properties of CSF α-synuclein as a possible biomarker for DLB. However, finding new CSF biomarkers is only useful if clinicians are willing to use lumbar puncture as a diagnostic tool. Therefore, the current use of CSF analysis in the diagnostic work up of memory clinic patients, both in the Netherlands and in Europe, was investigated in chapter Five. Finally, in order to facilitate the interpretation of CSF biomarkers in clinical practice, we developed a prediction model based on CSF biomarkers which estimates the probability that dementia in a memory clinic patient is due to AD (Chapter Six). Hypotheses for the pathophysiological cascade of AD Although the involvement of plaques and tangles in AD has been known for over a century, many questions around the pathophysiological cascade of AD remain unresolved. Several hypotheses have been formulated, the amyloid cascade hypothesis being the oldest one that still finds support (Figure 2). 87-89 It postulated that the critical event in AD is the deposition of Aβ as a result of APP mismetabolism. Aβ, or APP cleavage products that contain Aβ, were suggested to be neurotoxic and to lead to phosphorylation of tau, possible by increasing the intraneuronal calcium concentration. Hyperphosphorylated tau polymerized into neurofibrillary tangles which further led to neuronal death. The temporal sequence of the hypothesis (Aβ plaques precede tau tangles) was based on cases of familial AD who displayed both Aβ plaques and neurofibrillary tangles and in whom a mutation in APP was discovered. Further support was found in reports of persons with Down syndrome, in whom it was shown that diffuse plaques were the first detectable lesion, followed by senile plaques, neurofibrillary tangles, and eventually severe loss of neurons and atrophy. A critical reappraisal almost 20 years later concluded that research findings thus far are compatible with this theory, even though the pathway between Aβ and tau is still 23

Chapter ONE Figure 2 The amyloid cascade hypothesis as defined in 1991 Reprinted from Trends Pharmacol Sci, Volume 12, Hardy J, Allsop D, Amyloid deposition as the central event in the aetiology of Alzheimer's disease, Page 387, Copyright (1991), with permission from Elsevier. Figure 3 Hypothetical model of the AD pathological cascade Abnormal Biomarker magnitude Aβ Tau-meditated neuronal injury and dysfunction Brain structure Memory Clinical function Normal Cognitively normal MCI Dementia Clinical disease stage Reprinted from Lancet Neurology, Volume 9, Jack CR, Jr., Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ, Hypothetical model of dynamic biomarkers of the Alzheimer's pathological cascade, Page 122, Copyright (2010), with permission from Elsevier. 24

Introduction and Outline Figure 4 The vascular hypothesis Vascular Dementia Alzheimer s Dementia Vascular risk factors Abeta clearance Amyloid precursor protein Vascular Insufficiency Secretase activity Vascular dysregulation Beta-Amyloid Brain dysfunction and damage Dementia With kind permission from Springer Science+Business Media: Acta Neuropathologica, The overlap between neurodegenerative and vascular factors in the pathogenesis of dementia, Volume 120, 2010, Page 292, Iadecola C, Figure 3. unclear, and Aβ toxicity has not sufficiently been demonstrated in vivo to explain the massive cell loss as seen in AD. 90 Correlations between Aβ plaque load and cognitive impairment or dementia severity have both been described and disputed 91-95 and it has been suggested that not the Aβ plaques, but the still soluble oligomeric Aβ species are in fact the culprit. 96 Recently, a hypothetical model of the AD pathological cascade was described that incorporated information from studies on several AD biomarkers. 44 In line with the amyloid cascade hypothesis, it was suggested that the initiating event in AD is related to abnormal processing of Aβ, resulting in Aβ deposition. Aβ biomarkers (decreased CSF Aβ 42 concentration and increased uptake of PiB) were hypothesized to reach a plateau before clinical symptoms are apparent (Figure 3). Biomarkers of neuronal injury and neurodegeneration, such as increased CSF t-tau and p-tau and cerebral atrophy on MRI, were suggested to become abnormal in a later stage of the disease and to correlate with clinical symptom severity. 25

Chapter ONE The vascular hypothesis of AD can be regarded as an extension of the amyloid hypothesis (Figure 4). It proposes a bidirectional relationship between cerebrovascular disease and AD pathology. 97 Cerebrovascular disease, through chronic or intermittent hypoperfusion and oxidative stress, is suggested to lead to APP upregulation and perhaps to Aβ deposition, thus initiating or aggravating AD. 98-100 Vice versa, AD may lead to vascular disease since Aβ exerts detrimental effects on cerebrovascular function. 101 The influence of vascular factors on tau pathology is less well understood, although ischemia might induce hyperphosphorylation of tau. 97 Hypotheses serve no purpose if they are not tested and challenged. They often incorporate information from a multitude of sources, such as animal models, post-mortem research and explorative studies in selected patients groups with strict in- and exclusion criteria. It cannot simply be assumed that these data can be extrapolated to clinical practice. For example, previous studies showed that results from animal models cannot always be replicated in humans 102,103 and neuropathological studies generally suffer from selection bias. For example, one of the largest and most influential series of neuropathological studies on AD (the Nun study) consisted fully of members of a religious order. 104 These participants may not be representative for the general population. Even for other neuropathological studies it is important to realize that the subset of persons who agree to autopsy may not be representative. Testing hypotheses in the population of interest is therefore an essential part of research. In Chapter Seven the proposed separation between the timing of Aβ and tau pathology was investigated in an AD patient with several years of follow up. Chapter Eight takes a closer look at the suggestion that Aβ biomarkers reach a plateau before clinical symptoms are apparent, while tau biomarkers supposedly continue to increase during the clinical stages and thus correlate with clinical symptom severity. The vascular hypothesis is subject of Chapter Nine, in which the relation between cerebrovascular disease and AD pathology was investigated in a sample of the memory clinic population. Once it is established how pathological factors such as Aβ, tau and vascular factors interact and how this is reflected in their biomarkers, these biomarkers may be used as intermediate outcomes in trials that aim to influence the pathology of the disease. Since AD is a disease that develops over several years or even decades, an intervention that slows or halts this development cannot be expected to yield obvious clinical results in a short period of time. A major advantage of biomarkers is that they may provide early evidence that an intervention is successful in changing a pathological process. With the search for disease-modifying treatments, CSF biomarkers, and especially Aβ 42, have been advocated as markers to detect and monitor the biochemical effects of newly 26

Introduction and Outline developed drugs for AD. 105,106 To be able to interpret changes in biomarker concentrations that occur during a pharmacological intervention, it is important to understand the reason why these biomarker concentrations were abnormal in the first place. In Chapter Ten, (theoretical) reasons why the CSF Aβ 42 concentration is decreased in AD have been systematically examined. Aims of this thesis This thesis has two aims: One, to explore the potential use of CSF biomarkers as diagnostic tools in the differential diagnosis of dementia. Two, to investigate whether CSF biomarkers, derived from a sample of memory clinic patients, support the current hypotheses on the pathophysiological cascade of AD. Outline of this thesis This thesis is divided in two parts that address the aims outlined above by means of several investigations. This is followed by a summary and general discussion of the findings. Part One CSF biomarkers in the differential diagnosis of dementia Chapter Two investigates the diagnostic value of the CSF Aβ 42 /Aβ 40 ratio in differentiating patients with AD from controls and from patients with either VaD, DLB or FTD. It is hypothesized that this ratio provides a more valid measure for reflecting changes in Aβ levels in dementia patients than CSF Aβ 42 alone. The Aβ 42 /Aβ 40 ratio combined with p-tau and t-tau is also compared to the combination of Aβ 42, p-tau and t-tau. Chapter Three examines the value of CSF α-synuclein as biomarker for DLB, by comparing its concentration between groups of DLB, AD, VaD and FTD patients. In addition, a comparison to the CSF α-synuclein concentration in controls is made. Chapter Four further explores the properties of the CSF α-synuclein concentration in two ways: the presence of a circadian rhythm in CSF α-synuclein concentrations is 27

Chapter ONE investigated, and the correlation between CSF α-synuclein and Aβ concentrations in healthy elderly, AD and DLB patients is investigated. Chapter Five assesses the current use of CSF analysis in memory clinics in Europe and in the Netherlands. Chapter Six describes the development and validation of a prediction model for AD based on CSF biomarkers, meant to facilitate the use of CSF biomarkers in clinical practice. A few models have been described previously, but they do not offer a formula that can be applied in clinical practice for the differentiation between dementia due to AD and dementia due to another cause. 107,108 The prediction model in this chapter translates the results of CSF analysis into the probability that dementia in a memory clinic patient is due to AD. Part Two CSF biomarkers in pathophysiological hypotheses Chapter Seven challenges the hypothetical model of the AD pathophysiological cascade (Figure 3) with empirical data. Especially the separation in the moments at which Aβ and tau become abnormal is questioned. This will be illustrated by a case of early-onset AD with the longest follow-up of CSF biomarkers reported in the literature. Chapter Eight explores the correlation between CSF biomarkers and cognitive performance in a heterogeneous group of memory clinic patients. It is hypothesized that changes in the CSF biomarkers reflect neuronal damage and directly underlie cognitive impairment. According to the suggested pathophysiological cascade of AD, the strongest correlation should be expected with tau, and only a weak or absent correlation with Aβ. Chapter Nine investigates the hypothesis that AD patients have an increased susceptibility to develop cerebrovascular disease. In addition, possible mechanisms for this relationship are investigated, using CSF Aβ 42, CSF Aβ 40 and APOE-ε4 status. Chapter Ten reviews the evidence for several proposed reasons why the CSF Aβ 42 concentration is decreased in AD, in order to create a better understanding of the underlying mechanisms that lead to its recognition as an AD biomarker. Chapter Eleven provides a summary and discussion of the findings presented in this thesis. 28

Introduction and Outline References 1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Text Revision. Washington DC, American Psychiatric Association 2000. 2. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Jr., Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. The diagnosis of dementia due to Alzheimer s disease: Recommendations from the National Institute on Aging- Alzheimer s Association workgroups on diagnostic guidelines for Alzheimer s disease. Alzheimers Dement 2011;7:263-9. 3. Kalaria R. Similarities between Alzheimer s disease and vascular dementia. J Neurol Sci 2002;203-204:29-34. 4. McKeith I, Mintzer J, Aarsland D, Burn D, Chiu H, Cohen-Mansfield J, Dickson D, Dubois B, Duda JE, Feldman H, Gauthier S, Halliday G, Lawlor B, Lippa C, Lopez OL, Carlos MJ, O Brien J, Playfer J, Reid W. Dementia with Lewy bodies. Lancet Neurol 2004;3:19-28. 5. Roman GC, Tatemichi TK, Erkinjuntti T, Cummings JL, Masdeu JC, Garcia JH, Amaducci L, Orgogozo JM, Brun A, Hofman A. Vascular dementia: diagnostic criteria for research studies. Report of the NINDS-AIREN International Workshop. Neurology 1993;43:250-60. 6. McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen LA, Salmon DP, Lowe J, Mirra SS, Byrne EJ, Lennox G, Quinn NP, Edwardson JA, Ince PG, Bergeron C, Burns A, Miller BL, Lovestone S, Collerton D, Jansen EN, Ballard C, de Vos RA, Wilcock GK, Jellinger KA, Perry RH. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology 1996;47:1113-24. 7. Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, Freedman M, Kertesz A, Robert PH, Albert M, Boone K, Miller BL, Cummings J, Benson DF. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology 1998;51:1546-54. 8. Cairns NJ, Bigio EH, Mackenzie IR, Neumann M, Lee VM, Hatanpaa KJ, White CL, III, Schneider JA, Grinberg LT, Halliday G, Duyckaerts C, Lowe JS, Holm IE, Tolnay M, Okamoto K, Yokoo H, Murayama S, Woulfe J, Munoz DG, Dickson DW, Ince PG, Trojanowski JQ, Mann DM. Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol 2007;114:5-22. 9. Hodges JR, Davies RR, Xuereb JH, Casey B, Broe M, Bak TH, Kril JJ, Halliday GM. Clinicopathological correlates in frontotemporal dementia. Ann Neurol 2004;56:399-406. 10. Mackenzie IR, Neumann M, Bigio EH, Cairns NJ, Alafuzoff I, Kril J, Kovacs GG, Ghetti B, Halliday G, Holm IE, Ince PG, Kamphorst W, Revesz T, Rozemuller AJ, Kumar-Singh S, Akiyama H, Baborie A, Spina S, Dickson DW, Trojanowski JQ, Mann DM. Nomenclature and nosology for neuropathologic subtypes of frontotemporal lobar degeneration: an update. Acta Neuropathol 2010;119:1-4. 11. Seilhean D, Le B, I, Sarazin M, Lacomblez L, Millecamps S, Salachas F, Pradat PF, Le FN, LeGuern E, Dubois B, Meininger V, Brice A, Hauw JJ, Duyckaerts C. Fronto-temporal lobar degeneration: neuropathology in 60 cases. J Neural Transm 2011;118:753-64. 12. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer s Disease. Neurology 1984;34:939-44. 13. Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer s 1907 paper, Uber eine eigenartige Erkankung der Hirnrinde. Clin Anat 1995;8:429-31. 14. Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A 1986;83:4913-7. 15. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, Wisniewski HM. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem 1986;261:6084-9. 16. Grundke-Iqbal I, Vorbrodt AW, Iqbal K, Tung YC, Wang GP, Wisniewski HM. Microtubule-associated polypeptides tau are altered in Alzheimer paired helical filaments. Brain Res 1988;464:43-52. 29

Chapter ONE 17. Iqbal K, Grundke-Iqbal I, Zaidi T, Merz PA, Wen GY, Shaikh SS, Wisniewski HM, Alafuzoff I, Winblad B. Defective brain microtubule assembly in Alzheimer s disease. Lancet 1986;2:421-6. 18. Iqbal K, Grundke-Iqbal I, Smith AJ, George L, Tung YC, Zaidi T. Identification and localization of a tau peptide to paired helical filaments of Alzheimer disease. Proc Natl Acad Sci U S A 1989;86:5646-50. 19. Hansson O, Zetterberg H, Buchhave P, Andreasson U, Londos E, Minthon L, Blennow K. Prediction of Alzheimer s disease using the CSF Abeta42/Abeta40 ratio in patients with mild cognitive impairment. Dement Geriatr Cogn Disord 2007;23:316-20. 20. Kanai M, Matsubara E, Isoe K, Urakami K, Nakashima K, Arai H, Sasaki H, Abe K, Iwatsubo T, Kosaka T, Watanabe M, Tomidokoro Y, Shizuka M, Mizushima K, Nakamura T, Igeta Y, Ikeda Y, Amari M, Kawarabayashi T, Ishiguro K, Harigaya Y, Wakabayashi K, Okamoto K, Hirai S, Shoji M. Longitudinal study of cerebrospinal fluid levels of tau, A beta1-40, and A beta1-42(43) in Alzheimer s disease: a study in Japan. Ann Neurol 1998;44:17-26. 21. Lewczuk P, Esselmann H, Otto M, Maler JM, Henkel AW, Henkel MK, Eikenberg O, Antz C, Krause WR, Reulbach U, Kornhuber J, Wiltfang J. Neurochemical diagnosis of Alzheimer s dementia by CSF Abeta42, Abeta42/ Abeta40 ratio and total tau. Neurobiol Aging 2004;25:273-81. 22. Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43). Neuron 1994;13:45-53. 23. Verbeek MM, Eikelenboom P, De Waal RM. Differences between the pathogenesis of senile plaques and congophilic angiopathy in Alzheimer disease. J Neuropathol Exp Neurol 1997;56:751-61. 24. Rensink AA, De Waal RM, Kremer B, Verbeek MM. Pathogenesis of cerebral amyloid angiopathy. Brain Res Brain Res Rev 2003;43:207-23. 25. Gravina SA, Ho L, Eckman CB, Long KE, Otvos L, Jr., Younkin LH, Suzuki N, Younkin SG. Amyloid beta protein (A beta) in Alzheimer s disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at A beta 40 or A beta 42(43). J Biol Chem 1995;270:7013-6. 26. Braak E, Griffing K, Arai K, Bohl J, Bratzke H, Braak H. Neuropathology of Alzheimer s disease: what is new since A. Alzheimer? Eur Arch Psychiatry Clin Neurosci 1999;249 Suppl 3:14-22. 27. Crowther RA. Structural aspects of pathology in Alzheimer s disease. Biochim Biophys Acta 1990;1096:1-9. 28. www.cbo.nl/downloads/387/rl_dement_2005.pdf. CBO 2005. 29. Riedijk S, Duivenvoorden H, Van SJ, Niermeijer M, Tibben A. Sense of competence in a Dutch sample of informal caregivers of frontotemporal dementia patients. Dement Geriatr Cogn Disord 2009;27:337-43. 30. de Rotrou J, Cantegreil I, Faucounau V, Wenisch E, Chausson C, Jegou D, Grabar S, Rigaud AS. Do patients diagnosed with Alzheimer s disease benefit from a psycho-educational programme for family caregivers? A randomised controlled study. Int J Geriatr Psychiatry 2011;26:833-42. 31. Aarsland D, Kurz M, Beyer M, Bronnick K, Piepenstock NS, Ballard C. Early discriminatory diagnosis of dementia with Lewy bodies. The emerging role of CSF and imaging biomarkers. Dement Geriatr Cogn Disord 2008;25:195-205. 32. Olichney JM, Galasko D, Salmon DP, Hofstetter CR, Hansen LA, Katzman R, Thal LJ. Cognitive decline is faster in Lewy body variant than in Alzheimer s disease. Neurology 1998;51:351-7. 33. McKeith I, Fairbairn A, Perry R, Thompson P, Perry E. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ 1992;305:673-8. 34. Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH. The diagnosis of mild cognitive impairment due to Alzheimer s disease: recommendations from the National Institute on Aging-Alzheimer s Association workgroups on diagnostic guidelines for Alzheimer s disease. Alzheimers Dement 2011;7:270-9. 35. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR, Jr., Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y, Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH. Toward defining the preclinical stages of Alzheimer s disease: recommendations from the National Institute on Aging-Alzheimer s Association workgroups on diagnostic guidelines for Alzheimer s disease. Alzheimers Dement 2011;7:280-92. 36. Kazee AM, Eskin TA, Lapham LW, Gabriel KR, McDaniel KD, Hamill RW. Clinicopathologic correlates in Alzheimer disease: assessment of clinical and pathologic diagnostic criteria. Alzheimer Dis Assoc Disord 1993;7:152-64. 30

Introduction and Outline 37. Mendez MF, Mastri AR, Sung JH, Frey WH. Clinically diagnosed Alzheimer disease: neuropathologic findings in 650 cases. Alzheimer Dis Assoc Disord 1992;6:35-43. 38. Nelson PT, Jicha GA, Kryscio RJ, Abner EL, Schmitt FA, Cooper G, Xu LO, Smith CD, Markesbery WR. Low sensitivity in clinical diagnoses of dementia with Lewy bodies. J Neurol 2010;257:359-66. 39. Molsa PK, Paljarvi L, Rinne JO, Rinne UK, Sako E. Validity of clinical diagnosis in dementia: a prospective clinicopathological study. J Neurol Neurosurg Psychiatry 1985;48:1085-90. 40. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001;69:89-95. 41. Dubois B, Feldman HH, Jacova C, DeKosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, Meguro K, O Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Stern Y, Visser PJ, Scheltens P. Research criteria for the diagnosis of Alzheimer s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol 2007;6:734-46. 42. Dubois B, Feldman HH, Jacova C, Cummings JL, DeKosky ST, Barberger-Gateau P, Delacourte A, Frisoni G, Fox NC, Galasko D, Gauthier S, Hampel H, Jicha GA, Meguro K, O Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Sarazin M, de Souza LC, Stern Y, Visser PJ, Scheltens P. Revising the definition of Alzheimer s disease: a new lexicon. Lancet Neurol 2010;9:1118-27. 43. Consensus report of the Working Group on: Molecular and Biochemical Markers of Alzheimer s Disease. The Ronald and Nancy Reagan Research Institute of the Alzheimer s Association and the National Institute on Aging Working Group. Neurobiol Aging 1998;19:109-16. 44. Jack CR, Jr., Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ. Hypothetical model of dynamic biomarkers of the Alzheimer s pathological cascade. Lancet Neurol 2010;9:119-28. 45. de Leon MJ, George AE, Stylopoulos LA, Smith G, Miller DC. Early marker for Alzheimer s disease: the atrophic hippocampus. Lancet 1989;2:672-3. 46. Ball MJ, Fisman M, Hachinski V, Blume W, Fox A, Kral VA, Kirshen AJ, Fox H, Merskey H. A new definition of Alzheimer s disease: a hippocampal dementia. Lancet 1985;1:14-6. 47. Scheltens P, Leys D, Barkhof F, Huglo D, Weinstein HC, Vermersch P, Kuiper M, Steinling M, Wolters EC, Valk J. Atrophy of medial temporal lobes on MRI in probable Alzheimer s disease and normal ageing: diagnostic value and neuropsychological correlates. J Neurol Neurosurg Psychiatry 1992;55:967-72. 48. de Leon MJ, George AE, Golomb J, Tarshish C, Convit A, Kluger A, De SS, McRae T, Ferris SH, Reisberg B, Ince C, Rusinek H, Bobinski M, Quinn B, Miller DC, Wisniewski HM. Frequency of hippocampal formation atrophy in normal aging and Alzheimer s disease. Neurobiol Aging 1997;18:1-11. 49. Jack CR, Jr., Dickson DW, Parisi JE, Xu YC, Cha RH, O Brien PC, Edland SD, Smith GE, Boeve BF, Tangalos EG, Kokmen E, Petersen RC. Antemortem MRI findings correlate with hippocampal neuropathology in typical aging and dementia. Neurology 2002;58:750-7. 50. Ibanez V, Pietrini P, Alexander GE, Furey ML, Teichberg D, Rajapakse JC, Rapoport SI, Schapiro MB, Horwitz B. Regional glucose metabolic abnormalities are not the result of atrophy in Alzheimer s disease. Neurology 1998;50:1585-93. 51. Hoffman JM, Welsh-Bohmer KA, Hanson M, Crain B, Hulette C, Earl N, Coleman RE. FDG PET imaging in patients with pathologically verified dementia. J Nucl Med 2000;41:1920-8. 52. Jagust W, Reed B, Mungas D, Ellis W, DeCarli C. What does fluorodeoxyglucose PET imaging add to a clinical diagnosis of dementia? Neurology 2007;69:871-7. 53. Palmert MR, Podlisny MB, Witker DS, Oltersdorf T, Younkin LH, Selkoe DJ, Younkin SG. The beta-amyloid protein precursor of Alzheimer disease has soluble derivatives found in human brain and cerebrospinal fluid. Proc Natl Acad Sci U S A 1989;86:6338-42. 54. Seubert P, Vigo-Pelfrey C, Esch F, Lee M, Dovey H, Davis D, Sinha S, Schlossmacher M, Whaley J, Swindlehurst C,. Isolation and quantification of soluble Alzheimer s beta-peptide from biological fluids. Nature 1992;359:325-7. 55. Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, McKay DM, Tintner R, Frangione B,. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science 1992;258:126-9. 56. Vandermeeren M, Mercken M, Vanmechelen E, Six J, van d, V, Martin JJ, Cras P. Detection of tau proteins in normal and Alzheimer s disease cerebrospinal fluid with a sensitive sandwich enzyme-linked immunosorbent assay. J Neurochem 1993;61:1828-34. 31

Chapter ONE 57. Weidemann A, Konig G, Bunke D, Fischer P, Salbaum JM, Masters CL, Beyreuther K. Identification, biogenesis, and localization of precursors of Alzheimer s disease A4 amyloid protein. Cell 1989;57:115-26. 58. Galasko D, Chang L, Motter R, Clark CM, Kaye J, Knopman D, Thomas R, Kholodenko D, Schenk D, Lieberburg I, Miller B, Green R, Basherad R, Kertiles L, Boss MA, Seubert P. High cerebrospinal fluid tau and low amyloid beta42 levels in the clinical diagnosis of Alzheimer disease and relation to apolipoprotein E genotype. Arch Neurol 1998;55:937-45. 59. Motter R, Vigo-Pelfrey C, Kholodenko D, Barbour R, Johnson-Wood K, Galasko D, Chang L, Miller B, Clark C, Green R,. Reduction of beta-amyloid peptide42 in the cerebrospinal fluid of patients with Alzheimer s disease. Ann Neurol 1995;38:643-8. 60. Shoji M, Matsubara E, Kanai M, Watanabe M, Nakamura T, Tomidokoro Y, Shizuka M, Wakabayashi K, Igeta Y, Ikeda Y, Mizushima K, Amari M, Ishiguro K, Kawarabayashi T, Harigaya Y, Okamoto K, Hirai S. Combination assay of CSF tau, A beta 1-40 and A beta 1-42(43) as a biochemical marker of Alzheimer s disease. J Neurol Sci 1998;158:134-40. 61. Tamaoka A, Sawamura N, Fukushima T, Shoji S, Matsubara E, Shoji M, Hirai S, Furiya Y, Endoh R, Mori H. Amyloid beta protein 42(43) in cerebrospinal fluid of patients with Alzheimer s disease. J Neurol Sci 1997;148:41-5. 62. Vigo-Pelfrey C, Seubert P, Barbour R, Blomquist C, Lee M, Lee D, Coria F, Chang L, Miller B, Lieberburg I,. Elevation of microtubule-associated protein tau in the cerebrospinal fluid of patients with Alzheimer s disease. Neurology 1995;45:788-93. 63. Blennow K. Cerebrospinal fluid protein biomarkers for Alzheimer s disease. NeuroRx 2004;1:213-25. 64. de Jong D, Kremer BP, Olde Rikkert MG, Verbeek MM. Current state and future directions of neurochemical biomarkers for Alzheimer s disease. Clin Chem Lab Med 2007;45:1421-34. 65. Vanmechelen E, Vanderstichele H, Hulstaert F, Andreasen N, Minthon L, Winblad B, Davidsson P, Blennow K. Cerebrospinal fluid tau and beta-amyloid(1-42) in dementia disorders. Mech Ageing Dev 2001;122:2005-11. 66. Verbeek MM, de Jong D, Kremer HP. Brain-specific proteins in cerebrospinal fluid for the diagnosis of neurodegenerative diseases. Ann Clin Biochem 2003;40:25-40. 67. de Jong D, Jansen RW, Kremer BP, Verbeek MM. Cerebrospinal fluid amyloid beta42/phosphorylated tau ratio discriminates between Alzheimer s disease and vascular dementia. J Gerontol A Biol Sci Med Sci 2006;61:755-8. 68. Mukaetova-Ladinska EB, Monteith R, Perry EK. Cerebrospinal fluid biomarkers for dementia with lewy bodies. Int J Alzheimers Dis 2010;2010:536538. 69. Schoonenboom NS, Pijnenburg YA, Mulder C, Rosso SM, van Elk EJ, Van Kamp GJ, Van Swieten JC, Scheltens P. Amyloid beta(1-42) and phosphorylated tau in CSF as markers for early-onset Alzheimer disease. Neurology 2004;62:1580-4. 70. Verwey NA, van der Flier WM, Blennow K, Clark C, Sokolow S, De Deyn PP, Galasko D, Hampel H, Hartmann T, Kapaki E, Lannfelt L, Mehta PD, Parnetti L, Petzold A, Pirttila T, Saleh L, Skinningsrud A, Swieten JC, Verbeek MM, Wiltfang J, Younkin S, Scheltens P, Blankenstein MA. A worldwide multicentre comparison of assays for cerebrospinal fluid biomarkers in Alzheimer s disease. Ann Clin Biochem 2009;46:235-40. 71. Mulder C, Verwey NA, van der Flier WM, Bouwman FH, Kok A, van Elk EJ, Scheltens P, Blankenstein MA. Amyloid-beta(1-42), total tau, and phosphorylated tau as cerebrospinal fluid biomarkers for the diagnosis of Alzheimer disease. Clin Chem 2010;56:248-53. 72. Mattsson N, Andreasson U, Persson S, Arai H, Batish SD, Bernardini S, Bocchio-Chiavetto L, Blankenstein MA, Carrillo MC, Chalbot S, Coart E, Chiasserini D, Cutler N, Dahlfors G, Duller S, Fagan AM, Forlenza O, Frisoni GB, Galasko D, Galimberti D, Hampel H, Handberg A, Heneka MT, Herskovits AZ, Herukka SK, Holtzman DM, Humpel C, Hyman BT, Iqbal K, Jucker M, Kaeser SA, Kaiser E, Kapaki E, Kidd D, Klivenyi P, Knudsen CS, Kummer MP, Lui J, Llado A, Lewczuk P, Li QX, Martins R, Masters C, McAuliffe J, Mercken M, Moghekar A, Molinuevo JL, Montine TJ, Nowatzke W, O Brien R, Otto M, Paraskevas GP, Parnetti L, Petersen RC, Prvulovic D, de Reus HP, Rissman RA, Scarpini E, Stefani A, Soininen H, Schroder J, Shaw LM, Skinningsrud A, Skrogstad B, Spreer A, Talib L, Teunissen C, Trojanowski JQ, Tumani H, Umek RM, Van BB, Vanderstichele H, Vecsei L, Verbeek MM, Windisch M, Zhang J, Zetterberg H, Blennow K. The Alzheimer s Association external quality control program for cerebrospinal fluid biomarkers. Alzheimers Dement 2011;7:386-95. 73. Sojkova J, Driscoll I, Iacono D, Zhou Y, Codispoti KE, Kraut MA, Ferrucci L, Pletnikova O, Mathis CA, Klunk WE, O Brien RJ, Wong DF, Troncoso JC, Resnick SM. In vivo fibrillar beta-amyloid detected using [11C]PiB positron emission tomography and neuropathologic assessment in older adults. Arch Neurol 2011;68:232-40. 32