ORIGINAL CONTRIBUTION. Evaluation of CSF-tau and CSF-A 42 as Diagnostic Markers for Alzheimer Disease in Clinical Practice

Similar documents
Cerebrospinal fluid tau protein as a biochemical marker for Alzheimer s disease: a community based follow up study

Biochemical Diagnosis of Alzheimer Disease by Measuring the Cerebrospinal Fluid Ratio of Phosphorylated tau Protein to -Amyloid Peptide 42

V. Senanarong, N. Siwasariyanon, L. Washirutmangkur, N. Poungvarin, C. Ratanabunakit, N. Aoonkaew, and S. Udomphanthurak

ORIGINAL CONTRIBUTION. Correlation of Longitudinal Cerebrospinal Fluid Biomarkers With Cognitive Decline in Healthy Older Adults

ORIGINAL CONTRIBUTION. Cerebrospinal Fluid -Amyloid 42 and Tau Proteins as Biomarkers of Alzheimer-Type Pathologic

Differential Diagnosis of Alzheimer Disease With Cerebrospinal Fluid Levels of Tau Protein Phosphorylated at Threonine 231

Biomarkers for Alzheimer s disease

Association of Cerebrospinal Fluid Tau Protein in Patients with Alzheimer s and Non Alzheimer s Dementias in a Tertiary Level Hospital in Bangladesh

1. Introduction. 2. Patients and Methods

Papers. Detection of Alzheimer s disease and dementia in the preclinical phase: population based cohort study. Abstract.

The Brain Injury Biomarker VLP-1 Is Increased in the Cerebrospinal Fluid of Alzheimer Disease Patients

ORIGINAL CONTRIBUTION. Plasma and Cerebrospinal Fluid Levels of Amyloid Proteins 1-40 and 1-42 in Alzheimer Disease

Mild Cognitive Impairment (MCI)

Dementia. Stephen S. Flitman, MD Medical Director 21st Century Neurology

DISCLOSURES. Objectives. THE EPIDEMIC of 21 st Century. Clinical Assessment of Cognition: New & Emerging Tools for Diagnosing Dementia NONE TO REPORT

Dementia mimicking Alzheimer s disease Owing to a tau mutation: CSF and PET findings

September 26 28, 2013 Westin Tampa Harbour Island. Co-sponsored by

Mild Cognitive Impairment

ORIGINAL CONTRIBUTION

I n recent years, the concept of mild cognitive impairment

The current state of healthcare for Normal Aging, Mild Cognitive Impairment, & Alzheimer s Disease

Measurement of Phosphorylated Tau Epitopes in the Differential Diagnosis of Alzheimer Disease

CSF Biomarkers and Incipient Alzheimer Disease in Patients With Mild Cognitive Impairment

Clinical Study Cerebrospinal Fluid Levels of sappα and sappβ in Lewy Body and Alzheimer s Disease: Clinical and Neurochemical Correlates

Estimating the Validity of the Korean Version of Expanded Clinical Dementia Rating (CDR) Scale

ORIGINAL CONTRIBUTION. Comparison of the Short Test of Mental Status and the Mini-Mental State Examination in Mild Cognitive Impairment

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

WHAT IS DEMENTIA? An acquired syndrome of decline in memory and other cognitive functions sufficient to affect daily life in an alert patient

Assessment at the bedside or in the clinic using the history, examination and laboratory tests to distinguish between different types of dementia

CSF diagnosis of Alzheimer s disease and dementia with Lewy bodies

Both total and phosphorylated tau are increased in Alzheimer s disease

Cerebrospinal Fluid Protein Biomarkers for Alzheimer s Disease

APPENDIX SUPPLEMENTARY METHODS Control group criteria for each center... 2 strem2 ELISA protocol... 3 APPENDIX TABLES... 4

NACC Minimum Data Set (MDS) Public Data Element Dictionary

USE OF LABORATORY AND IMAGING INVESTIGATIONS IN DEMENTIA

Form D1: Clinician Diagnosis

Anosognosia, or loss of insight into one s cognitive

ORIGINAL CONTRIBUTION

Dementia and Healthy Ageing : is the pathology any different?

Brain imaging for the diagnosis of people with suspected dementia

Cerebrospinal Fluid Biomarker Signature in Alzheimer s Disease Neuroimaging Initiative Subjects

Psychiatry Research 120 (2003) Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany d

Use of cerebrospinal fluid biomarker analysis for improving Alzheimer s disease diagnosis in a non-specialized setting

How can the new diagnostic criteria improve patient selection for DM therapy trials

CSF Aβ1-42 predicts cognitive impairment in de novo PD patients

Introduction, use of imaging and current guidelines. John O Brien Professor of Old Age Psychiatry University of Cambridge

Longitudinal cerebrospinal fluid biomarker measurements in preclinical sporadic Alzheimer s disease: A prospective 9-year study

USE OF BIOMARKERS TO DISTINGUISH SUBTYPES OF DEMENTIA. SGEC Webinar Handouts 1/18/2013

ORIGINAL CONTRIBUTION. Diagnostic Validity of the Dementia Questionnaire for Alzheimer Disease

Fact Sheet Alzheimer s disease

Regulatory Challenges across Dementia Subtypes European View

Clinical Diagnosis. Step 1: Dementia or not? Diagnostic criteria for dementia (DSM-IV)

Lewy body disease (LBD) is the second most common

Comments to this discussion are invited on the Alzforum Webinar page. Who Should Use the New Diagnostic Guidelines? The Debate Continues

Erin Cullnan Research Assistant, University of Illinois at Chicago

ORIGINAL CONTRIBUTION. Apolipoprotein E 4 Is a Determinant for Alzheimer-Type Pathologic Features in Tauopathies, Synucleinopathies,

ALZHEIMER S DISEASE. Mary-Letitia Timiras M.D. Overlook Hospital Summit, New Jersey

Caring Sheet #11: Alzheimer s Disease:

I n the past three decades various cognitive screening

Alzheimer s Disease. Clinical characteristics of late-onset Alzheimer s disease (LOAD) A/Prof David Darby

LUP. Lund University Publications Institutional Repository of Lund University

Diagnosis and Treatment of Alzhiemer s Disease

EFFECT OF DIFFERENT DIAGNOSTIC CRITERIA ON THE PREVALENCE OF DEMENTIA. Special Article

Validity of Family History for the Diagnosis of Dementia Among Siblings of Patients With Late-onset Alzheimer s Disease

EPIDEMIOLOGY AND RISK FACTORS OF DEMENTIA

Ruolo dei biomarcatori come criterio di supporto nella diagnostica delle demenze ad esordio precoce

Assessing and Managing the Patient with Cognitive Decline

Role of TDP-43 in Non-Alzheimer s and Alzheimer s Neurodegenerative Diseases

Yin-Hui Siow MD, FRCPC Director of Nuclear Medicine Southlake Regional Health Centre

Vascular dementia (VaD) is preceded by several years of

Practice parameter: Diagnosis of dementia (an evidence-based review)

NEXT-Link DEMENTIA. A network of Danish memory clinics YOUR CLINICAL RESEARCH PARTNER WITHIN ALZHEIMER S DISEASE AND OTHER DEMENTIA DISEASES.

DEMENTIA 101: WHAT IS HAPPENING IN THE BRAIN? Philip L. Rambo, PhD

Delirium, Apo-E status, and AD CSF biomarkers

Accelerated Memory Decline in Alzheimer s Disease With Apolipoprotein e4 Allele

ORIGINAL CONTRIBUTION. APOE Genotype, Family History of Dementia, and Alzheimer Disease Risk

Validation of amyloid-b peptides in CSF diagnosis of neurodegenerative dementias

Dementia. Assessing Brain Damage. Mental Status Examination

Type 2 Diabetes and Brain Disease in Older Adults. Erin L. Abner, PhD, MPH Asst. Professor University Of Kentucky

Dementia Past, Present and Future

Frontotemporal dementia and dementia with Lewy bodies in a case-control study of Alzheimer s disease

ORIGINAL CONTRIBUTION. Neuropathologic Outcome of Mild Cognitive Impairment Following Progression to Clinical Dementia

Improving diagnosis of Alzheimer s disease and lewy body dementia. Brain TLC October 2018

Clinicopathologic and genetic aspects of hippocampal sclerosis. Dennis W. Dickson, MD Mayo Clinic, Jacksonville, Florida USA

Alzheimer's disease (AD), also known as Senile Dementia of the Alzheimer Type (SDAT) or simply Alzheimer s is the most common form of dementia.

ORIGINAL CONTRIBUTION. Attention and Fluctuating Attention in Patients With Dementia With Lewy Bodies and Alzheimer Disease

Accuracy of the Clinical Diagnosis of Alzheimer Disease at National Institute on Aging Alzheimer Disease Centers, 2005Y2010

REGULAR RESEARCH ARTICLES

Baseline Characteristics of Patients Attending the Memory Clinic Serving the South Shore of Boston

Neuropsychiatric Manifestations in Vascular Cognitive Impairment Patients with and without Dementia

Brain Health and Risk Factors for Dementia

Quantitative analysis for a cube copying test

ORIGINAL CONTRIBUTION. Diagnostic Accuracy of Dementia With Lewy Bodies. to be the second

ORIGINAL CONTRIBUTION. The Relative Frequency of Dementia of Unknown Etiology Increases With Age and Is Nearly 50% in Nonagenarians

212 Index C-SB-13,

New life Collage of nursing Karachi

Transcription:

ORIGINAL CONTRIBUTION Evaluation of CSF-tau and CSF-A 42 as Diagnostic Markers for Alzheimer Disease in Clinical Practice Niels Andreasen, MD, PhD; Lennart Minthon, MD, PhD; Pia Davidsson, PhD; Eugeen Vanmechelen, PhD; Hugo Vanderstichele, PhD; Bengt Winblad, MD, PhD; Kaj Blennow, MD, PhD Objective: To evaluate the diagnostic potential of cerebrospinal fluid (CSF) levels of tau and -amyloid protein ending at amino acid 42 (A 42) as biomarkers for Alzheimer disease (AD) in clinical practice. Design: A 1-year prospective study. Setting: Community population based sample of all consecutive patients admitted for investigation of cognitive symptoms to the Piteå River Valley Hospital, Piteå, Sweden. Patients: A total of 241 patients with probable AD (n=105), possible AD (n=58), vascular dementia (n=23), mild cognitive impairment (n=20), Lewy body dementia (n=9), other neurological disorders (n=3), and psychiatric disorders (n=5) and nondemented individuals (n=18). Main Outcome Measures: Cerebrospinal fluid tau and CSF-A 42 were assayed each week as routine clinical neurochemical analyses. Sensitivity and specificity were defined using the regression line from 100 control subjects from a multicenter study. Positive and negative predictive values were calculated for different prevalence rates of AD. Results: We found increased CSF-tau and decreased CSF- A 42 levels in probable and possible AD. Sensitivity was 94% for probable AD, 88% for possible AD, and 75% for mild cognitive impairment, whereas specificity was 100% for psychiatric disorders and 89% for nondemented. Specificity was lower in Lewy body dementia (67%) mainly because of low CSF-A 42 levels and in vascular dementia (48%) mainly because of high CSF-tau levels. Sensitivity for CSF-tau and CSF-A 42 increased in patients with AD possessing the ApoE 4 allele, approaching 100%. At a prevalence of AD of 45%, the positive predictive value was 90% and the negative predictive value was 95%. Conclusions: Cerebrospinal fluid tau and CSF-A 42 have so far been studied in research settings, under conditions providing data on the optimal performance. We examined a prospective patient sample, with assays run in clinical routine, giving figures closer to the true performance of CSF-tau and CSF-A 42. The predictive value for AD was greater than 90%. Therefore, these biomarkers may have a role in the clinical workup of patients with cognitive impairment, especially to differentiate early AD from normal aging and psychiatric disorders. Arch Neurol. 2001;58:373-379 The affiliations of the authors appear in the acknowledgment section at the end of the article. THE CLINICAL diagnosis of sporadic Alzheimer disease (AD) is based on the identification of dementia with a clinical profile suggestive of AD from the medical history and clinical examination together with the exclusion of other causes of dementia using brain imaging and laboratory tests. 1 There are no established (ie, used in clinical routine) biochemical markers to identify AD. Such biochemical markers might increase diagnostic accuracy, especially early in the course of the disease, when clinical symptoms might be mild and vague and overlap with cognitive changes accompanying aging and other brain disorders. Especially in view of future disease-modifying compounds, which are likely to have their maximal benefit before neurodegeneration is widespread, there is a great need for reliable biochemical diagnostic markers of AD. For editorial comment see page 349 A diagnostic marker for AD should reflect a central pathogenic process of the disease, ie, the degeneration of the neurons and their synapses and the defining lesion s senile plaques (SPs) and neurofibrillary tangles. 2 Two such biomarkers are tau and -amyloid protein ending at amino acid 42 (A 42). The cerebrospinal fluid 373

PATIENTS AND METHODS STUDY POPULATION This investigation was part of the longitudinal geriatric population study in Piteå, Sweden, 12 with a population of approximately 60000 individuals. All individuals with cognitive impairment must be referred for medical examination at the hospital. Patients were admitted from the local general practitioner or the community health service. The study included all consecutive patients (N=265) admitted during 1 year (September 1, 1998, to August 31, 1999). A lumbar puncture (LP) was performed on all patients who accepted (n=241; acceptance rate, 91%). Clinical evaluation was performed in a standardized way, and all data were recorded in research protocols. 12 Diagnostic evaluation in all patients included a clinical examination (detailed medical history and somatic, neuropsychiatric, and neurological status), a neuropsychologic test battery, assessment of activities of daily living, routine blood tests to exclude secondary dementias (eg, vitamin B 12, folate, albumin, calcium, and thyroid-stimulating hormone), routine CSF tests to identify blood-brain barrier damage and infectious and inflammatory disorders, an electroencephalogram (to evaluate -frequency and focal abnormalities), and a computed tomographic scan (to evaluate cortical atrophy, white matter lesions, and infarcts and lacunas). Clinical diagnoses were based on summarized information from the diagnostic evaluation and were made by one of us (N.A., a geriatrician). The presence or absence of dementia was diagnosed according to the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), criteria. 13 Probable and possible AD were diagnosed according to the National Institute of Neurological and Communicative Disorders and Stroke Alzheimer s Disease and Related Disorders Association (NINCDS-ADRDA) criteria, 1 and vascular dementia (VAD) according to the National Institute of Neurological Disorders and Stroke Association Internationale pour la Recherche et l Enseignement en Neuroscience criteria. 14 Patients with probable AD had an insidious onset and even progression of dementia, which could not be explained by systemic or brain disorders other than AD. No patient had prominent frontal lobe symptoms or history, clinical, or brain imaging signs of cerebrovascular disease, except for mild white matter lesions. Vascular dementia was diagnosed in patients with a history of transitory ischemic attacks or stroke episodes with a temporal relation to development of dementia together with computed tomographic and magnetic resonance tomographic findings of lacunas, infarcts, and often moderate or marked white matter lesions. The possible AD group preferentially consisted of patients with mixed AD/VAD, ie, those with even progression of dementia but with a history of transitory ischemic attacks or stroke episodes without temporal relation to development of dementia or with accidental findings of lacunas, minor infarcts, or moderate white matter lesions on computed tomography. Patients with clinical findings suggestive of AD but also with signs of other degenerative disorders (eg, frontal lobe symptoms) were also diagnosed as having possible AD. Mild cognitive impairment (MCI) was diagnosed in patients with memory impairment but no other symptoms of dementia according to established criteria. 15 Of patients with MCI, 3 (15%) of 20 progressed to AD with dementia during follow-up. Lewy body dementia (LBD) was diagnosed according to consensus criteria. 16 The nondemented group included individuals with subjective minor memory complaints but without objective signs of memory (CSF) level of tau has been suggested to reflect neuronal and axonal degeneration 3 or possibly formation of neurofibrillary tangles, 4 whereas the CSF-A 42 level might reflect the deposition of A into SPs, with lower levels remaining in the CSF. 5 Several previous studies have found increased CSF-tau 6,7 and reduced CSF-A 42 5,8,9 levels in AD. A large multicenter study 10 found that the combination of CSF-tau and CSF-A 42 gave approximately 85% sensitivity and specificity for AD. However, all previous studies are based on patient series from research centers with analyses run at a single occasion in research laboratories. To further evaluate the clinical usefulness of CSF markers, sensitivity and specificity data must be calculated on consecutive patients and biochemical analyses must be run in routine clinical neurochemistry. In a recent study, Andreasen et al 11 showed that CSF-tau has high sensitivity for AD, also, in clinical practice. In this study, we present data for the combination of CSF-tau and CSF-A 42 as diagnostic markers for AD based on all patients admitted for dementia examination to Piteå River Valley Hospital, Piteå, Sweden, during a 1-year period, where CSF analyses were run each week in routine clinical neurochemistry. RESULTS The coefficient of variance for the internal control samples, run on 76 different ELISA plates during 1 year, was 18.9% for CSF-tau and 10.7% for CSF-A 42 for the normal control and 10.1% for CSF-tau and 11.0% for CSF-A 42 for the AD control. We also studied the analytical variation for the CSF-A 42 assay and the stability of CSF-A 42 by reanalyzing 41 stored ( 6 months) CSF samples on 1 ELISA plate. The correlation between A 42 run in clinical routine at different times during 1 year and the same samples rerun at one occasion was high (r=0.96; P.001) (Figure 1). Of 241 patients included in the study, 10 (4.1%) had post-lp headaches (mild in 4 patients, moderate in 4, and severe in 2). There was a significant increase in the level of CSFtau in the probable AD group compared with the VAD (P=.001), MCI (P=.04), LBD (P.001), and nondemented (P.001) groups. An increase in CSF-tau levels was also found in the possible AD group compared with the LBD (P=.002), nondemented (P.001), and VAD (P=.04) groups. No significant differences were found among the other diagnostic groups (Table 2). 374

impairment or dementia symptoms at the clinical examination or neuropsychological assessment. In the nondemented group, an outcome observation and criteria was that no progression was found during follow-up. All other clinical diagnoses were made according to established criteria (DSM-IV 13 and International Classification of Diseases, 10th Revision [ICD-10] 17 ). All clinical diagnoses and evaluations were made without knowledge of the results of the biochemical analyses and vice versa. The clinical characteristics of the patients are given in Table 1. Severity of dementia was evaluated using the Mini-Mental State Examination. 18 The ethics committees in Umeå and Göteborg, Sweden, approved the study. The regression line from a large multicenter study 10 in which the same enzyme-linked immunosorbent assays (ELISAs) for tau and A 42 determinations were used defined the cutoff levels for CSF-tau and CSF-A 42 (A 42=240+1.18 tau). This study 10 included CSF samples from 100 healthy volunteers or patients without brain disorders. CSF ANALYSES Samples of CSF were taken in polypropylene tubes to avoid absorption of A into the test tubes 5 and were sent by ordinary mail to the Clinical Neurochemistry Laboratory at Sahlgren s University Hospital in Mölndal, Sweden. After arrival (the day after LP), samples were aliquoted and frozen pending biochemical analyses, which were performed within 1 week. The incidence of post-lp headache was recorded prospectively. Post-LP headache was graded on a scale from 0 to 3 (0, absent; 1, mild headache with duration 2 days; 2, moderate headache with duration 2 days requiring administration of oral analgesics; and 3, severe headache with duration 2 days requiring treatment with an epidural blood patch). The level of CSF-tau was determined using an ELISA (Innotest htau-ag; Innogenetics NV, Gent, Belgium) constructed to measure both normal tau and phosphorylated tau. 6,19 The level of CSF-A 42 was determined using an ELISA (INNOTEST -amyloid (1-42) ; Innogenetics) specific for A 42. 5,20 Assays of CSF-tau and CSF-A 42 were run as routine clinical neurochemical analyses. Analyses were run every week, and all samples were run in duplicate. Two CSF pools were made for use as internal controls: a normal pool (CSF samples from patients with psychiatric or minor neurological disorders) with a mean tau value of 288 pg/ml and a mean A 42 value of 700 pg/ml and an AD pool with a mean tau level of 904 pg/ml and a mean A 42 level of 383 pg/ml. Control pools were stored at 80 C and were run on every ELISA plate analyzed (n=76). ApoE GENOTYPING Apolipoprotein E genotyping was performed by polymerase chain reaction followed by minisequencing as described previously. 21 STATISTICAL ANALYSIS Comparisons between groups were performed using factor analysis of variance with post hoc analyses (Tukey honestly significant difference test for unequal N). The Pearson correlation coefficient was used for correlations. Sensitivity (ie, the proportion of patients with AD and high tau and low A 42 levels) and specificity (ie, the proportion of other patients with normal tau and A 42 levels) were calculated using the cutoff line from a multicenter study. 10 There was a marked decrease in CSF-A 42 levels in the probable AD group compared with the VAD (P=.006), psychiatric disorders (P=.003), and nondemented (P.001) groups. A decrease in the CSF-A 42 level was also found in the possible AD group compared with the psychiatric disorders (P=.02) and nondemented (P.001) groups, in the MCI group compared with the nondemented group (P=.006), and in the LBD group compared with the nondemented group (P=.004). No significant differences were found among the other diagnostic groups (Table 2). Within the AD group, there were no significant correlations between age and either CSF-tau (r= 0.10; P=.32) or CSF-A 42 (r=0.003; P=.98). However, because there were significant differences in age among the diagnostic groups, we performed multiple analyses of variance with CSF-tau or CSF-A 42 as dependent variables and age as a covariate, which showed an effect by diagnosis (P.001) but not by age for CSF-tau (P=.83) or CSF-A 42 (P=.54). Sensitivity and specificity data for the combination of CSF-tau and CSF-A 42 using the cutoff line from the multicenter study 10 are presented in Table 2, and the individual values are given in Figure 2. Sensitivity was 94% for probable AD, 88% for possible AD, and 75% for MCI (Table 2). Specificity was 100% for psychiatric disorders and 89% for the nondemented group (Table 2). Specificity was lower in the LBD group (67%) mainly because of low CSF-A 42 levels. The lowest separation was found in the VAD group, with a specificity of 48% mainly because of high CSF-tau levels. Sensitivity for the combination of CSF-tau and CSF- A 42 in patients possessing the ApoE 4 allele increased from 94% to 99% (73/74) for probable AD, from 88% to 100% (27/27) for possible AD, and from 75% to 88% (7/8) for MCI (Figure 2). In VAD, all 3 ApoE 4 positive patients had pathologic values for CSF-tau and CSF-A 42 (Figure 2). Positive and negative predictive values for the combination of tau and A 42 at different disease prevalences are given in Figure 3. The prevalence of probable AD was 105 (44%) of 241, resulting in a positive predictive value of 90% and a negative predictive value of 95%. Positive and negative predictive values were 82% and 97%, respectively, at a prevalence of 30% and 73% and 98%, respectively, at a prevalence of 20% (Figure 3). There were no significant differences in CSF-tau levels between patients without vs with the ApoE 4 allele in the probable AD (892±590 vs 730±319 pg/ml; P=.23), possible AD (716±310 vs 680±234 pg/ml; P=.23), VAD 375

Table 1. Basic Clinical Characteristics of the Diagnostic Groups* Diagnostic Group Patients, No. (n = 241) Sex, M/F, No. Age, Mean ± SD, y Duration, Mean ± SD, y MMSE Score, Mean ± SD Probable AD 105 39/66 75.9 ± 6.8 3.3 ± 2.9 22.8 ± 4.9 Possible AD 58 34/24 77.4 ± 6.4 3.4 ± 2.4 21.6 ± 5.1 VAD 23 14/9 76.5 ± 7.4 3.2 ± 2.9 22.3 ± 5.0 MCI 20 8/12 70.9 ± 6.4 2.6 ± 2.7 27.8 ± 2.0 LBD 9 5/4 74.6 ± 7.0 2.1 ± 1.0 20.9 ± 4.9 Other neurological disorders 3 2/1 64.7 ± 10.1 3.5 ± 2.8 24.3 ± 2.5 Psychiatric disorders 5 3/2 67.8 ± 4.7 3.3 ± 4.6 25.4 ± 4.4 Nondemented 18 5/13 61.8 ± 12.1 2.8 ± 1.6 28.4 ± 2.2 *MMSE indicates Mini-Mental State Examination; AD, Alzheimer disease; VAD, vascular dementia; MCI, mild cognitive impairment; and LBD, Lewy body dementia. Significances for age: nondemented vs probable AD, P.001; possible AD, P.001; VAD, P.001; and LBD, P =.002. Possible AD vs MCI, P =.01. Significances for MMSE: nondemented vs probable AD, P.001; possible AD, P.001; VAD, P.001; and LBD, P =.003. MCI vs probable AD, P.001; possible AD, P.001; VAD, P =.002; and LBD, P =.009. Includes frontotemporal dementia (n = 1), cerebrovascular disorder (n = 1), and Creutzfeldt-Jakob disease (n = 1). Includes depression (n = 3) and alcoholism (n = 2). CSF-Aβ42, pg/ml, Rerun on 1 Plate 2000 1500 1000 500 CSF-Aβ42, pg/ml, in Clinical Routine During 1 y Figure 1. Analytical variation and storage stability of cerebrospinal fluid -amyloid protein ending at amino acid 42 (CSF-A 42). Samples of CSF were assayed in clinical routine during 1 year and reanalyzed on 1 enzyme-linked immunosorbent assay plate (n=41; r=0.96, P.001). (486±284 vs 338±257 pg/ml; P=.34), or LBD (262±105 vs 188±70 pg/ml; P=.23) groups. In contrast, there were significant differences in CSF-A 42 levels between patients without vs with the ApoE 4 allele in the probable AD (622±228 vs 482±137 pg/ml; P.001), possible AD (649±251 vs 484±153 pg/ml; P=.004), and VAD (762±303 vs 322±62 pg/ml; P.001) groups. No significant difference was found in the LBD group (624±203 vs 456±53 pg/ml; P=.22), possibly because of the low number of patients (n=6 vs n=3). COMMENT We evaluated the utility of the combination of CSF-tau and CSF-A 42 as diagnostic markers for AD in clinical practice. All patients admitted for evaluation of suspected dementia to a community hospital during 1 year were included. Assays of CSF samples were run each week as routine analyses in a clinical neurochemical laboratory. This setting gives the opportunity to further evaluate the diagnostic potential of diagnostic markers for AD. Samples of CSF were sent at room temperature over a substantial distance (approximately 1600 km). Reanalysis of CSF-A 42 on a single occasion gave values similar to those obtained at several runs during 1 year. The stability of the ELISAs, as determined by running both high and low control samples on each plate, was also acceptable and in the range expected for immunoassays. These findings suggest that the present procedure for handling and analyzing CSF samples for routine analyses is accurate and that the ELISAs are robust. We found an increase in CSF-tau and a decrease in CSF-A 42 levels in AD, in agreement with results of several previous studies. 5-11 Using the cutoff line from a multicenter study, 10 the sensitivity to identify AD was high, greater than 90%, and the positive and negative predictive values for AD were both high. Furthermore, sensitivity increased if the ApoE genotype also was taken into consideration. Academic centers report accuracy rates for the clinical diagnosis of AD of 65% to 90%, 22-24 although some studies have reported lower figures. 25 Thus, higher sensitivity figures than those obtained in the present study might not be expected for diagnostic markers when evaluated in clinically diagnosed patients. Specificity was high to differentiate AD from psychiatric disorders and nondemented. However, specificity was lower in the LBD group mainly because several patients had low CSF-A 42 levels. This might be a consequence of patients with LBD harboring SPs in the brain. 26 The lowest specificity was found in the VAD group. One possible explanation is that patients with VAD, in addition to cerebrovascular abnormalities, might have concomitant AD pathologic findings, which is impossible to exclude clinically. Neuropathologic studies 27,28 have found that a high proportion of patients with clinically diagnosed VAD (40%-80%) has notable concomitant AD pathologic findings. Indeed, the lowest CSF-A 42 levels in VAD were found in patients with the ApoE 4 allele, raising the question of whether these patients har- 376

Table 2. Cerebrospinal Fluid (CSF) Levels of tau and A 42 in the Diagnostic Groups* Diagnostic Group Patients, No. (n = 241) CSF-tau, Mean ± SD, pg/ml CSF-A 42, Mean ± SD, pg/ml Sensitivity for a Diagnosis of AD, % (95% CI) Specificity for a Diagnosis of Non-AD, % (95% CI) Probable AD 105 759 ± 417 523 ± 180 94 (88-97) NA Possible AD 58 699 ± 275 572 ± 225 88 (77-94) NA VAD 23 461 ± 280 704 ± 321 NA 48 (29-67) MCI 20 517 ± 190 640 ± 260 75 (53-89) NA LBD 9 238 ± 97 568 ± 183 NA 67 (35-88) Other neurological disorders Frontotemporal dementia 1 961 1060 NA NA Creutzfeldt-Jakob disease 1 3280 464 NA NA Cerebrovascular disorder 1 392 467 NA NA Psychiatric disorders 5 400 ± 115 901 ± 109 NA 100 (57-100) Nondemented 18 264 ± 102 897 ± 242 NA 89 (67-97) *A 42 indicates -amyloid protein ending at amino acid 42; AD, Alzheimer disease; CI, confidence interval; VAD, vascular dementia; MCI, mild cognitive impairment; LBD, Lewy body dementia; and NA, not applicable. Significances for CSF-tau: probable AD vs VAD, P =.001; MCI, P =.04; LBD, P.001; and nondemented, P.001. Possible AD vs LBD, P =.002; nondemented, P.001; and VAD, P =.04. Significances for CSF-A 42: probable AD vs VAD, P =.006; psychiatric disorders, P =.003; nondemented, P.001. Possible AD vs psychiatric disorders, P =.02; and nondemented, P.001. MCI vs nondemented, P =.006. LBD vs nondemented, P =.004. 2000 Probable AD Possible AD Vascular Dementia 1500 Aβ42, pg/ml 1000 500 0 2000 Mild Cognitive Impairment Lewy Body Dementia Other Neurological Disorders Psychiatric Disorders Nondemented 1500 Aβ42, pg/ml 1000 500 tau, pg/ml tau, pg/ml tau, pg/ml Figure 2. Individual values for cerebrospinal fluid tau and -amyloid protein ending at amino acid 42 (A 42) in the different diagnostic groups. The cutoff line (A 42=240+1.18 tau) is from a large multicenter study. 10 Black circles and squares indicate patients possessing the ApoE 4 allele. bor concomitant AD pathologic findings. It is clear that studies with neuropathologically confirmed cases are needed to determine with certainty the sensitivity and specificity of CSF-tau and CSF-A 42 as diagnostic markers for AD. Also, the 3 patients with other neurological disorders had abnormal CSF markers. The highest CSF-tau level in the present study was found in a patient with Creutzfeldt-Jakob disease (CJD), in agreement with results of previous studies. 29 The level of CSF-tau has been suggested to reflect neuronal and axonal degeneration, 6 which is very intense in CJD. The patient with CJD had an even higher CSF-tau value (14600 pg/ml) at follow-up 1 month later. Thus, very high CSF-tau 377

Predictive Value, % 100 90 80 70 60 50 40 30 20 10 Positive Negative 0 10 20 30 40 50 60 70 80 90 100 Prevalence of AD, % Figure 3. Positive and negative predictive values for the combination of cerebrospinal fluid tau and -amyloid protein ending at amino acid 42 at different prevalence rates of Alzheimer disease (AD). The shaded area is the approximate prevalence of AD in different series from the literature (40%-60%). levels may raise suspicion of CJD, although the sensitivity of CSF-tau to identify CJD has to be further evaluated. The patient with CJD also had low a CSF-A 42 level, also in agreement with results of a previous study, 30 supporting the fact that a low CSF-A 42 level is not specific for AD and questioning the mechanism for the reduction of CSF-A 42 levels in AD, which has been suggested to be a consequence of deposition of the A into SPs. 5,8 In the present study, we found high sensitivity for the combination of CSF-tau and CSF-A 42 for AD, whereas specificity was lower, especially for some other dementias and neurological disorders. Although this reduces the clinical diagnostic utility, we think that this drawback can, at least partly, be overcome by using CSF markers together with the summarized information gained from the clinical examination. 31 We suggest that AD can be diagnosed on the basis of a combination of (1) characteristic symptoms of, in the initial stage, memory disturbances and, later on, parietal symptoms; (2) characteristic brain imaging findings, eg, parietotemporal blood flow defect on single-photon emission computed tomography and hippocampal and cortical atrophy together with absence of cerebrovascular changes on computed tomographic or magnetic resonance tomographic scans; and (3) a characteristic pattern of CSF biomarkers (high CSF-tau and low CSF- A 42 values together with normal blood-brain barrier function and absence of pleocytosis or intrathecal immunoglobulin production) and other biochemical tools, eg, ApoE genotyping. 31 As an analogy, the clinical diagnosis of myocardial infarction is based on the combination of clinical symptoms, electrocardiographic findings, and biochemical markers (eg, creatine kinase). Furthermore, the effect of the lower specificity on the clinical usefulness of CSF-tau and CSF-A 42 might be overestimated because not all disorders in which abnormal levels of these biomarkers can be found are important (ie, difficult) differential diagnoses of AD, eg, acute stroke 32 or human immunodeficiency virus dementia. 33 Instead, CSF-tau and CSF-A 42 might have their major use as an adjunct to help to differentiate AD from the most problematic differential diagnoses, especially ageassociated memory impairment, depressive pseudodementia, Parkinson disease, progressive supranuclear palsy, and alcoholic dementia. Lumbar puncture is easy to perform, with a low risk for complications. 34 In the present study, the incidence of post-lp headache was low, also in clinical routine evaluation of patients admitted for cognitive impairment. Therefore, LP can be regarded as a feasible, moderately invasive test with a low risk for complications that can be included in the clinical diagnostic workup. In our view, CSF biomarkers might be especially important to be able to start treatment early in the course of the disease, when age-associated memory impairment and depressive pseudodementia are some of the most problematic differential diagnoses. 35 In a recent study, 36 we showed that the combination of CSF-tau and CSF-A 42 also might help identify patients with MCI who will develop AD. In summary, CSF biomarkers for AD so far have been studied in research settings under conditions providing data on their optimal performance. We evaluated the combination of CSF-tau and CSF-A 42 prospectively in a community-based sample of patients, and ELISAs were run each week in clinical neurochemical routine. Also, under these conditions, these biomarkers have positive and negative predictive values for AD greater than 90% and therefore might have a role in the clinical workup of patients with cognitive impairment, especially to differentiate early AD from normal aging and psychiatric disorders such as depressive pseudodementia. Accepted for publication July 5, 2000. From the Department of Rehabilitation, Piteå River Valley Hospital, Piteå, Sweden (Dr Andreasen); the Department of Psychiatry, Neuropsychiatric Clinic, Malmö University Hospital, Malmö, Sweden (Dr Minthon); the Department of Clinical Neuroscience, Unit of Neurochemistry, University of Göteborg, Sahlgren s University Hospital, Mölndal, Sweden (Drs Davidsson and Blennow); Innogenetics NV, Gent, Belgium (Drs Vanmechelen and Vanderstichele); the Section of Geriatric Medicine, Department of Clinical Neuroscience and Family Medicine, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden (Dr Winblad); and The Medical Research Council, Stockholm, Sweden (Dr Blennow). Dr Winblad is now with the Department of Clinical Neuroscience, Occupational Therapy and Elderly Care Research, Division of Geriatric Medicine, Karolinska Institute, Huddinge University Hospital, Stockholm. This work was supported by grants 11560 and 12103 from the Swedish Medical Research Council, Stockholm; by Alzheimerfonden, Lund, Sweden; by Stiftelsen för Gamla Tjänarinnor, Stockholm, Sweden; by the Tore Nilssons Fond för Medicinsk Forskning, Stockholm; by the Norrbottens Läns Landstings FoU Fond, Luleå Sweden; by Svenska Läkaresällskapet, Stockholm; and by Åke Wibergs Stiftelse, Stockholm. We are grateful to everyone at the CSF Protein Section at the Neurochemistry Laboratory, University of 378

Göteborg, Sahlgren s University Hospital, and to Christina Sjödin for her skillful technical assistance. Corresponding author and reprints: Niels Andreasen, MD, PhD, Department of Rehabilitation, Piteå River Valley Hospital, PO Box 715, SE-94128 Piteå, Sweden (e-mail: Niels.Andreasen@nll.se). REFERENCES 1. 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 the Department of Health and Human Services Task Force on Alzheimer s disease. Neurology. 1984;34:939-944. 2. The Ronald and Nancy Reagan Research Institute of the Alzheimer s Association and the National Institute on Aging Working Group. Consensus report of the Working Group on Molecular and Biochemical Markers of Alzheimer s Disease. Neurobiol Aging. 1998;19:109-116. 3. Andreasen N, Vanmechelen E, Van de Voorde E, et al. Cerebrospinal fluid tau protein as a biochemical marker for Alzheimer s disease: a community-based follow-up study. J Neurol Neurosurg Psychiatry. 1998;64:298-305. 4. Tapiola T, Overmyer M, Lehtovirta M, et al. The level of cerebrospinal fluid tau correlates with neurofibrillary tangles in Alzheimer s disease. Neuroreport. 1997; 8:3961-3963. 5. Andreasen N, Hesse C, Davidsson P, et al. Cerebrospinal fluid -amyloid (1-42) in Alzheimer s disease: differences between early- and late-onset AD and stability during the course of disease. Arch Neurol. 1999;56:673-680. 6. Blennow K, Wallin A, Ågren H, Spenger C, Siegfried J, Vanmechelen E. Tau protein in cerebrospinal fluid: a biochemical marker for axonal degeneration in Alzheimer s disease? Mol Chem Neuropathol. 1995;26:231-245. 7. Vigo-Pelfrey C, Seubert P, Barbour R, et al. Elevation of microtubule-associated protein tau in the cerebrospinal fluid of patients with Alzheimer s disease. Neurology. 1995;45:788-793. 8. Motter R, Vigo-Pelfrey C, Kholodenko D, et al. Reduction of -amyloid peptide 42 in the cerebrospinal fluid of patients with Alzheimer s disease. Ann Neurol. 1995;38:643-648. 9. Galasko D, Clark C, Chang L, et al. High cerebrospinal fluid tau and low amyloid 42 levels in the clinical diagnosis of Alzheimer disease and relation to apolipoprotein E genotype. Arch Neurol. 1998;55:937-945. 10. Hulstaert F, Blennow K, Ivanoiu A, et al. Improved discrimination of Alzheimer s disease patients from other subject groups using the combined measure of -amyloid(1-42) and tau in CSF: a multicenter study. Neurology. 1999;52:1555-1562. 11. Andreasen N, Minthon L, Clarberg A, et al. Sensitivity, specificity, and stability of CSF-tau in AD in a community-based patient sample. Neurology. 1999;53: 1488-1494. 12. Andreasen N, Sjödin C, Blennow K, Winblad B, Svärsudd K. Prevalence and incidence of memory recall disturbances in a geographically defined general population: the Piteå Dementia Project. Neuroepidemiology. 1999;18:144-155. 13. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition. Washington, DC: American Psychiatric Association; 1994. 14. Roman GC, Tatemichi TK, Erkinjuntti T, et al. Vascular dementia: diagnostic criteria for research studies: report of the NINDS-AIREN International Workshop. Neurology. 1993;43:243-245. 15. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Kokmen E, Tangelos EG. Aging, memory, and mild cognitive impairment. Int Psychogeriatr. 1997;9(suppl 1):65-69. 16. McKeith IG, Perry EK, Perry RH, for the Consortium on Dementia With Lewy Bodies. Report of the second Dementia With Lewy Body International Workshop: diagnosis and treatment. Neurology. 1999;53:902-905. 17. Klassifikation av Sjukdomar och Hälsoproblem 1997: Swedish Version of International Classification of Diseases, Ninth Revision (ICD-10). Stockholm, Sweden: National Board of Health and Welfare; 1996. 18. Folstein M, Folstein S, McHugh P. Mini-Mental State : a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975; 12:189-198. 19. Vandermeeren M, Mercken M, Vanmechelen E, et al. Detection of tau proteins in normal and Alzheimer s disease cerebrospinal fluid with a sensitive sandwich enzyme-linked immunosorbent assay. J Neurochem. 1993;61:1828-1834. 20. Vanderstichele H, Blennow K, Wallin A, et al. Development of a specific diagnostic test for measurement of -amyloid (1-42) in CSF. In: Fisher A, Hanin I, Yoshida M, eds. Alzheimer s and Parkinson s Diseases. New York, NY: Plenum Publishing Corp; 1998:773-778. 21. Blennow K, Ricksten A, Prince J, et al. No association between the 2- macroglobulin (A2M) deletion and Alzheimer s disease, and no change in A2M mrna, protein, or protein expression. J Neural Transm. 2000;107:1065-1079. 22. Tierney MC, Fisher RH, Lewis AJ, et al. The NINCDS-ADRDA Work Group criteria for the clinical diagnosis of probable Alzheimer s disease: a clinicopathologic study of 57 cases. Neurology. 1988;38:359-364. 23. Galasko D, Hansen LA, Katzman R, et al. Clinical-neuropathological correlations in Alzheimer s disease and related dementias. Arch Neurol. 1994;51:888-895. 24. Klatka LA, Schiffer RB, Powers JM, Kazee AM. Incorrect diagnosis of Alzheimer s disease: a clinicopathologic study. Arch Neurol. 1996;53:35-42. 25. Mayeux R, Saunders AM, Shea S, et al, for the Alzheimer s Disease Centers Consortium on Apolipoprotein E and Alzheimer s Disease. Utility of the apolipoprotein E genotype in the diagnosis of Alzheimer s disease. N Engl J Med. 1998; 338:506-511. 26. Perry EK, Irving D, Kerwin JM, et al. Cholinergic transmitter and neurotrophic activities in Lewy body dementia: similarity to Parkinson s and distinction from Alzheimer disease. Alzheimer Dis Assoc Disord. 1993;7:69-79. 27. Jellinger KA, Bancher C. Neuropathology of Alzheimer s disease: a critical update. J Neural Transm. 1998;54:77-95. 28. Kosunen O, Soininen H, Paljarvi L, Heinonen O, Talasniemi S, Riekkinen PJ Sr. Diagnostic accuracy of Alzheimer s disease: a neuropathological study. Acta Neuropathol (Berl). 1996;91:185-193. 29. Otto M, Wiltfang J, Tumani H, et al. Elevated levels of tau-protein in cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. Neurosci Lett. 1997;225:210-212. 30. Otto M, Esselmann H, Schulz-Shaeffer W, et al. Decreased -amyloid1-42 in cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. Neurology. 2000; 54:1099-1102. 31. Blennow K, Vanmechelen E. Combination of the different biological markers for increasing specificity of in vivo Alzheimer s testing. J Neural Transm. 1998;53: 223-235. 32. Andersson LM, Blennow K, Fuchs D, Svennerholm B, Gisslén M. Increased cerebrospinal fluid protein tau concentration in neuro-aids. J Neurol Sci. 1999; 171:92-96. 33. Hesse C, Rosengren L, Vanmechelen E, et al. Cerebrospinal fluid markers for Alzheimer s disease evaluated after acute ischaemic stroke. J Alzheimer Dis. In press. 34. Blennow K, Wallin A, Häger O. Low frequency of post-lumbar puncture headache in demented patients. Acta Neurol Scand. 1993;88:221-223. 35. Zapotoczky HG. Problems of differential diagnosis between depressive pseudodementia and Alzheimer s disease. In: Jellinger K, Fazekas F, Windisch M, eds. Ageing and Dementia. New York, NY: Springer-Verlag NY Inc; 1998:91-95. 36. Andreasen N, Minthon L, Vanmechelen E, et al. Cerebrospinal fluid tau and A 42 as predictors of development of Alzheimer s disease in patients with mild cognitive impairment. Neurosci Lett. 1999;273:5-8. 379