1
|
Wang Z, Lewis V, Stehmann C, Varghese S, Senesi M, McGlade A, Ellett LJ, Doecke JD, Eratne D, Velakoulis D, Masters CL, Collins SJ, Li Q. Alzheimer's disease biomarker utilization at first referral enhances differential diagnostic precision with simultaneous exclusion of Creutzfeldt-Jakob disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12548. [PMID: 38352040 PMCID: PMC10862167 DOI: 10.1002/dad2.12548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
Most suspected Creutzfeldt-Jakob disease (CJD) cases are eventually diagnosed with other disorders. We assessed the utility of investigating Alzheimer's disease (AD) biomarkers and neurofilament light (NfL) in patients when CJD is suspected. The study cohort consisted of cerebrospinal fluid (CSF) samples referred for CJD biomarker screening wherein amyloid beta 1-42 (Aβ1-42), phosphorylated tau 181 (p-tau181), and total tau (t-tau) could be assessed via Elecsys immunoassays (n = 419) and NfL via enzyme-linked immunosorbent assay (ELISA; n = 161). In the non-CJD sub cohort (n = 371), 59% (219/371) had A+T- (abnormal Aβ1-42 only) and 21% (79/371) returned A+T+ (abnormal Aβ1-42 and p-tau181). In the 48 CJD subjects, a similar AD biomarker profile distribution was observed. To partially address the prevalence of likely pre-symptomatic AD, NfL was utilized to assess for neuronal damage. NfL was abnormal in 76% (25/33) of A+T- subjects 40 to 69 years of age, 80% (20/25) of whom had normal t-tau. This study reinforces AD as an important differential diagnosis of suspected CJD, highlighting that incorporating AD biomarkers and NfL at initial testing is worthwhile.
Collapse
Affiliation(s)
- Zitianyu Wang
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | - Victoria Lewis
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Department of Medicine, Clinical Sciences Building, Royal Melbourne Hospital (RMH)The University of MelbourneParkvilleAustralia
| | - Christiane Stehmann
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | - Shiji Varghese
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | - Matteo Senesi
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Department of Medicine, Clinical Sciences Building, Royal Melbourne Hospital (RMH)The University of MelbourneParkvilleAustralia
| | - Amelia McGlade
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | - Laura J. Ellett
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | | | - Dhamidhu Eratne
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Neuropsychiatry, John Cade BuildingRoyal Melbourne HospitalParkvilleAustralia
| | - Dennis Velakoulis
- Neuropsychiatry, John Cade BuildingRoyal Melbourne HospitalParkvilleAustralia
| | - Colin L. Masters
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
| | - Steven J. Collins
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Australian National Creutzfeldt‐Jakob Disease Registry (ANCJDR), The Florey InstituteThe University of MelbourneParkvilleAustralia
- Department of Medicine, Clinical Sciences Building, Royal Melbourne Hospital (RMH)The University of MelbourneParkvilleAustralia
| | - Qiao‐Xin Li
- National Dementia Diagnostics Laboratory (NDDL), The Florey InstituteThe University of MelbourneParkvilleAustralia
| |
Collapse
|
2
|
Papaliagkas V, Kalinderi K, Vareltzis P, Moraitou D, Papamitsou T, Chatzidimitriou M. CSF Biomarkers in the Early Diagnosis of Mild Cognitive Impairment and Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24108976. [PMID: 37240322 DOI: 10.3390/ijms24108976] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a rapidly growing disease that affects millions of people worldwide, therefore there is an urgent need for its early diagnosis and treatment. A huge amount of research studies are performed on possible accurate and reliable diagnostic biomarkers of AD. Due to its direct contact with extracellular space of the brain, cerebrospinal fluid (CSF) is the most useful biological fluid reflecting molecular events in the brain. Proteins and molecules that reflect the pathogenesis of the disease, e.g., neurodegeneration, accumulation of Abeta, hyperphosphorylation of tau protein and apoptosis may be used as biomarkers. The aim of the current manuscript is to present the most commonly used CSF biomarkers for AD as well as novel biomarkers. Three CSF biomarkers, namely total tau, phospho-tau and Abeta42, are believed to have the highest diagnostic accuracy for early AD diagnosis and the ability to predict AD development in mild cognitive impairment (MCI) patients. Moreover, other biomarkers such as soluble amyloid precursor protein (APP), apoptotic proteins, secretases and inflammatory and oxidation markers are believed to have increased future prospects.
Collapse
Affiliation(s)
- Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrion University Campus, 57400 Sindos, Greece
| | - Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Patroklos Vareltzis
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despoina Moraitou
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theodora Papamitsou
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Chatzidimitriou
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrion University Campus, 57400 Sindos, Greece
| |
Collapse
|
3
|
Mohammadi Z, Alizadeh H, Marton J, Cumming P. The Sensitivity of Tau Tracers for the Discrimination of Alzheimer's Disease Patients and Healthy Controls by PET. Biomolecules 2023; 13:290. [PMID: 36830659 PMCID: PMC9953528 DOI: 10.3390/biom13020290] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperphosphorylated tau aggregates, also known as neurofibrillary tangles, are a hallmark neuropathological feature of Alzheimer's disease (AD). Molecular imaging of tau by positron emission tomography (PET) began with the development of [18F]FDDNP, an amyloid β tracer with off-target binding to tau, which obtained regional specificity through the differing distributions of amyloid β and tau in AD brains. A concerted search for more selective and affine tau PET tracers yielded compounds belonging to at least eight structural categories; 18F-flortaucipir, known variously as [18F]-T807, AV-1451, and Tauvid®, emerged as the first tau tracer approved by the American Food and Drug Administration. The various tau tracers differ concerning their selectivity over amyloid β, off-target binding at sites such as monoamine oxidase and neuromelanin, and degree of uptake in white matter. While there have been many reviews of molecular imaging of tau in AD and other conditions, there has been no systematic comparison of the fitness of the various tracers for discriminating between AD patient and healthy control (HC) groups. In this narrative review, we endeavored to compare the binding properties of the various tau tracers in vitro and the effect size (Cohen's d) for the contrast by PET between AD patients and age-matched HC groups. The available tracers all gave good discrimination, with Cohen's d generally in the range of two-three in culprit brain regions. Overall, Cohen's d was higher for AD patient groups with more severe illness. Second-generation tracers, while superior concerning off-target binding, do not have conspicuously higher sensitivity for the discrimination of AD and HC groups. We suppose that available pharmacophores may have converged on a maximal affinity for tau fibrils, which may limit the specific signal imparted in PET studies.
Collapse
Affiliation(s)
- Zohreh Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hadi Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Straße 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
4
|
Botella Lucena P, Vanherle S, Lodder C, Gutiérrez de Ravé M, Stancu IC, Lambrichts I, Vangheluwe R, Bruffaerts R, Dewachter I. Blood-based Aβ42 increases in the earliest pre-pathological stage before decreasing with progressive amyloid pathology in preclinical models and human subjects: opening new avenues for prevention. Acta Neuropathol 2022; 144:489-508. [PMID: 35796870 PMCID: PMC9381631 DOI: 10.1007/s00401-022-02458-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022]
Abstract
Blood-based (BB) biomarkers for Aβ and tau can indicate pathological processes in the brain, in the early pathological, even pre-symptomatic stages in Alzheimer’s disease. However, the relation between BB biomarkers and AD-related processes in the brain in the earliest pre-pathology stage before amyloid pathology develops, and their relation with total brain concentrations of Aβ and tau, is poorly understood. This stage presents a critical window for the earliest prevention of AD. Preclinical models with well-defined temporal progression to robust amyloid and tau pathology provide a unique opportunity to study this relation and were used here to study the link between BB biomarkers with AD-related processes in pre- and pathological stages. We performed a cross-sectional study at different ages assessing the link between BB concentrations and AD-related processes in the brain. This was complemented with a longitudinal analysis and with analysis of age-related changes in a small cohort of human subjects. We found that BB-tau concentrations increased in serum, correlating with progressive development of tau pathology and with increasing tau aggregates and p-tau concentrations in brain in TauP301S mice (PS19) developing tauopathy. BB-Aβ42 concentrations in serum decreased between 4.5 and 9 months of age, correlating with the progressive development of robust amyloid pathology in APP/PS1 (5xFAD) mice, in line with previous findings. Most importantly, BB-Aβ42 concentrations significantly increased between 1.5 and 4.5 months, i.e., in the earliest pre-pathological stage, before robust amyloid pathology develops in the brain, indicating biphasic BB-Aβ42 dynamics. Furthermore, increasing BB-Aβ42 in the pre-pathological phase, strongly correlated with increasing Aβ42 concentrations in brain. Our subsequent longitudinal analysis of BB-Aβ42 in 5xFAD mice, confirmed biphasic BB-Aβ42, with an initial increase, before decreasing with progressive robust pathology. Furthermore, in human samples, BB-Aβ42 concentrations were significantly higher in old (> 60 years) compared to young (< 50 years) subjects, as well as to age-matched AD patients, further supporting age-dependent increase of Aβ42 concentrations in the earliest pre-pathological phase, before amyloid pathology. Also BB-Aβ40 concentrations were found to increase in the earliest pre-pathological phase both in preclinical models and human subjects, while subsequent significantly decreasing concentrations in the pathological phase were characteristic for BB-Aβ42. Together our data indicate that BB biomarkers reflect pathological processes in brain of preclinical models with amyloid and tau pathology, both in the pathological and pre-pathological phase. Our data indicate a biphasic pattern of BB-Aβ42 in preclinical models and a human cohort. And most importantly, we here show that BB-Aβ increased and correlated with increasing concentrations of Aβ in the brain, in the earliest pre-pathological stage in a preclinical model. Our data thereby identify a novel critical window for prevention, using BB-Aβ as marker for accumulating Aβ in the brain, in the earliest pre-pathological stage, opening new avenues for personalized early preventive strategies against AD, even before amyloid pathology develops.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Sarah Vanherle
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Chritica Lodder
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | | | - Ilie-Cosmin Stancu
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Ivo Lambrichts
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Riet Vangheluwe
- Neurology Department, ZOL Genk General Hospital, Genk, Belgium
| | - Rose Bruffaerts
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium.,Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU, 3000, Leuven, Belgium.,Department of Neurology, University Hospitals, 3000, Leuven, Belgium.,Computational Neurology, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium.
| |
Collapse
|
5
|
Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis 2022; 37:67-104. [PMID: 34719771 DOI: 10.1007/s11011-021-00800-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases (NDs) are characterised by progressive dysfunction of synapses, neurons, glial cells and their networks. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormalities. The most common neurodegenerative disorders are amyloidosis, tauopathies, a-synucleinopathy, and TAR DNA-binding protein 43 (TDP-43) proteopathy. The protein abnormalities in these disorders have abnormal conformational properties along with altered cellular mechanisms, and they exhibit motor deficit, mitochondrial malfunction, dysfunctions in autophagic-lysosomal pathways, synaptic toxicity, and more emerging mechanisms such as the roles of stress granule pathways and liquid-phase transitions. Finally, for each ND, microglial cells have been reported to be implicated in neurodegeneration, in particular, because the microglial responses can shift from neuroprotective to a deleterious role. Growing experimental evidence suggests that abnormal protein conformers act as seed material for oligomerization, spreading from cell to cell through anatomically connected neuronal pathways, which may in part explain the specific anatomical patterns observed in brain autopsy sample. In this review, we mention the human pathology of select neurodegenerative disorders, focusing on how neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis) represent a great healthcare problem worldwide and are becoming prevalent because of the increasing aged population. Despite many studies have focused on their etiopathology, the exact cause of these diseases is still largely unknown and until now with the only available option of symptomatic treatments. In this review, we aim to report the systematic and clinically correlated potential biomarker candidates. Although future studies are necessary for their use in early detection and progression in humans affected by NDs, the promising results obtained by several groups leads us to this idea that biomarkers could be used to design a potential therapeutic approach and preclinical clinical trials for the treatments of NDs.
Collapse
Affiliation(s)
- Nishit Pathak
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Ishi Tandon
- Amity University Jaipur, Rajasthan, Jaipur, Rajasthan, India
| | - Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, Kansei Behavioural and Brain Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Cao Hongyi
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
6
|
Kim SH, Lee EH, Kim HJ, Kim AR, Kim YE, Lee JH, Yoon MY, Koh SH. Development of a Low-Molecular-Weight Aβ42 Detection System Using a Enzyme-Linked Peptide Assay. Biomolecules 2021; 11:1818. [PMID: 34944462 PMCID: PMC8699310 DOI: 10.3390/biom11121818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that is the most common cause of dementia. The incidence of AD is rapidly rising because of the aging of the world population. Because AD is presently incurable, early diagnosis is very important. The disease is characterized by pathological changes such as deposition of senile plaques and decreased concentration of the amyloid-beta 42 (Aβ42) peptide in the cerebrospinal fluid (CSF). The concentration of Aβ42 in the CSF is a well-studied AD biomarker. The specific peptide probe was screened through four rounds of biopanning, which included the phage display process. The screened peptide showed strong binding affinity in the micromolar range, and the enzyme-linked peptide assay was optimized using the peptide we developed. This diagnostic method showed specificity toward Aβ42 in the presence of other proteins. The peptide-binding site was also estimated using molecular docking analysis. Finally, the diagnostic method we developed could significantly distinguish patients who were classified based on amyloid PET images.
Collapse
Affiliation(s)
- Sang-Heon Kim
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Eun-Hye Lee
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
| | - Hyung-Ji Kim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - A-Ru Kim
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Ye-Eun Kim
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Korea
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - Moon-Young Yoon
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Seong-Ho Koh
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Korea
| |
Collapse
|
7
|
Park S, Kim Y. Bias-generating factors in biofluid amyloid-β measurements for Alzheimer's disease diagnosis. Biomed Eng Lett 2021; 11:287-295. [PMID: 34616582 DOI: 10.1007/s13534-021-00201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia worldwide, yet the dearth of readily accessible diagnostic biomarkers is a substantial hindrance towards progressing to effective preventive and therapeutic approaches. Due to a long delay between cerebral amyloid-β (Aβ) accumulation and the onset of cognitive impairments, biomarkers that reflect Aβ pathology and enable routine screening for disease progression are of urgent need for application in the clinical diagnosis of AD. According to accumulating evidences, cerebrospinal fluid (CSF) and plasma offer windows to the brain as they allow monitoring of biochemical changes in the brain. Considering the high availability and accuracy in depicting Aβ deposition in the brain, Aβ levels in CSF and plasma are regarded as promising fluid biomarkers for the diagnosis of AD patients at an early stage. However, clinical data with intra- and interindividual variations in the concentrations of CSF and plasma Aβ implicate the need to reevaluate current Aβ detection methods and establish a standardized operating procedure. Therefore, this review introduces three bias-generating factors in biofluid Aβ measurement that may hamper the accurate Aβ quantification and how such complications can be overcome for the widespread implementation of fluid Aβ detection in clinical practice.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| |
Collapse
|
8
|
Jung NY, Kim ES, Kim HS, Jeon S, Lee MJ, Pak K, Lee JH, Lee YM, Lee K, Shin JH, Ko JK, Lee JM, Yoon JA, Hwang C, Choi KU, Lee EC, Seong JK, Huh GY, Kim DS, Kim EJ. Comparison of Diagnostic Performances Between Cerebrospinal Fluid Biomarkers and Amyloid PET in a Clinical Setting. J Alzheimers Dis 2021; 74:473-490. [PMID: 32039853 DOI: 10.3233/jad-191109] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The diagnostic performances of cerebrospinal fluid (CSF) biomarkers and amyloid positron emission tomography (PET) were compared by examining the association and concordance or discordance between CSF Aβ1-42 and amyloid PET, after determining our own cut-off values for CSF Alzheimer's disease (AD) biomarkers. Furthermore, we evaluated the ability of CSF biomarkers and amyloid PET to predict clinical progression. CSF Aβ1-42, t-tau, and p-tau levels were analyzed in 203 individuals [27 normal controls, 38 mild cognitive impairment (MCI), 62 AD dementia, and 76 patients with other neurodegenerative diseases] consecutively recruited from two dementia clinics. We used both visual and standardized uptake value ratio (SUVR)-based amyloid PET assessments for analyses. The association of CSF biomarkers with amyloid PET SUVR, hippocampal atrophy, and cognitive function were investigated by linear regression analysis, and the risk of conversion from MCI to AD dementia was assessed using a Cox proportional hazards model. CSF p-tau/Aβ1-42 and t-tau/Aβ1-42 exhibited the best diagnostic accuracies among the CSF AD biomarkers examined. Correlations were observed between CSF biomarkers and global SUVR, hippocampal volume, and cognitive function. Overall concordance and discordance between CSF Aβ1-42 and amyloid PET was 77% and 23%, respectively. Baseline positive CSF Aβ1-42 for MCI demonstrated a 5.6-fold greater conversion risk than negative CSF Aβ1-42 . However, amyloid PET findings failed to exhibit significant prognostic value. Therefore, despite presence of a significant correlation between the CSF Aβ1-42 level and SUVR of amyloid PET, and a relevant concordance between CSF Aβ1-42 and amyloid PET, baseline CSF Aβ1-42 better predicted AD conversion.
Collapse
Affiliation(s)
- Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Eun Soo Kim
- Department of Anesthesia and Pain Medicine, Pusan National University Hospital, School of Medicine, Pusan National University, Busan, Republic of Korea
| | - Hyang-Sook Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sumin Jeon
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jae-Hyeok Lee
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Young Min Lee
- Department of Psychiatry, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Kangyoon Lee
- Department of Psychiatry, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jin-Hong Shin
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jun Kyeung Ko
- Department of Neurosurgery, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jae Meen Lee
- Department of Neurosurgery, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jin A Yoon
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Chungsu Hwang
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Kyung-Un Choi
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Eun Chong Lee
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
| | - Joon-Kyung Seong
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
| | - Gi Yeong Huh
- Department of Forensic Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dae-Seong Kim
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| |
Collapse
|
9
|
Boerwinkle AH, Wisch JK, Chen CD, Gordon BA, Butt OH, Schindler SE, Sutphen C, Flores S, Dincer A, Benzinger TLS, Fagan AM, Morris JC, Ances BM. Temporal Correlation of CSF and Neuroimaging in the Amyloid-Tau-Neurodegeneration Model of Alzheimer Disease. Neurology 2021; 97:e76-e87. [PMID: 33931538 DOI: 10.1212/wnl.0000000000012123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/23/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate temporal correlations between CSF and neuroimaging (PET and MRI) measures of amyloid, tau, and neurodegeneration in relation to Alzheimer disease (AD) progression. METHODS A total of 371 cognitively unimpaired and impaired participants enrolled in longitudinal studies of AD had both CSF (β-amyloid [Aβ]42, phosphorylated tau181, total tau, and neurofilament light chain) and neuroimaging (Pittsburgh compound B [PiB] PET, flortaucipir PET, and structural MRI) measures. The pairwise time interval between CSF and neuroimaging measures was binned into 2-year periods. Spearman correlations identified the time bin when CSF and neuroimaging measures most strongly correlated. CSF and neuroimaging measures were then binarized as biomarker-positive or biomarker-negative using Gaussian mixture modeling. Cohen kappa coefficient identified the time bin when CSF measures best agreed with corresponding neuroimaging measures when determining amyloid, tau, and neurodegeneration biomarker positivity. RESULTS CSF Aβ42 and PiB PET showed maximal correlation when collected within 6 years of each other (R ≈ -0.5). CSF phosphorylated tau181 and flortaucipir PET showed maximal correlation when CSF was collected 4 to 8 years prior to PET (R ≈ 0.4). CSF neurofilament light chain and cortical thickness showed low correlation, regardless of time interval (R avg ≈ -0.3). Similarly, CSF total tau and cortical thickness had low correlation, regardless of time interval (R avg < -0.2). CONCLUSIONS CSF Aβ42 and PiB PET best agree when acquired in close temporal proximity, whereas CSF phosphorylated tau precedes flortaucipir PET by 4 to 8 years. CSF and neuroimaging measures of neurodegeneration have low correspondence and are not interchangeable at any time interval.
Collapse
Affiliation(s)
- Anna H Boerwinkle
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Julie K Wisch
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Charles D Chen
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Brian A Gordon
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Omar H Butt
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Suzanne E Schindler
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Courtney Sutphen
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Shaney Flores
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Aylin Dincer
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Tammie L S Benzinger
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Anne M Fagan
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - John C Morris
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO
| | - Beau M Ances
- From the Departments of Neurology (A.H.B., J.K.W., O.H.B., S.E.S., C.S., A.M.F., J.C.M., B.M.A.) and Radiology (C.D.C., B.A.G., S.F., A.D., T.L.S.B.), Washington University in St. Louis, MO.
| |
Collapse
|
10
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Lee JC, Kim HY, Lee S, Shin J, Kim HV, Kim K, Baek S, Lee D, Jeon H, Kim D, Yang S, Han G, Park K, Choi J, Park J, Moss JA, Janda KD, Kim Y. Discovery of Chemicals to Either Clear or Indicate Amyloid Aggregates by Targeting Memory‐Impairing Anti‐Parallel Aβ Dimers. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Jinny Claire Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
- Department of Chemistry Department of Immunology and Microbial Science The Skaggs Institute for Chemical Biology The Worm Institute for Research and Medicine (WIRM) The Scripps Research Institute La Jolla CA 92037 USA
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - Sejin Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
- KIST School University of Science and Technology (UST) Korea Institute of Science and Technology (KIST) Seoul 02792 South Korea
| | - Jisu Shin
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - Hyunjin Vincent Kim
- KIST School University of Science and Technology (UST) Korea Institute of Science and Technology (KIST) Seoul 02792 South Korea
| | - Kyeonghwan Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - Seungyeop Baek
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
- Department of Biotechnology Yonsei University Seoul 03722 South Korea
| | - Donghee Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - Hanna Jeon
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - DaWon Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| | - Seung‐Hoon Yang
- Department of Medical Biotechnology Dongguk University Gyeonggi-do 10326 South Korea
| | - Gyoonhee Han
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
- Department of Biotechnology Yonsei University Seoul 03722 South Korea
| | - Keunwan Park
- Natural Product Informatics Research Center Korea Institute of Science and Technology (KIST) Gangwon-do 25451 South Korea
| | | | | | - Jason A. Moss
- Department of Chemistry Department of Immunology and Microbial Science The Skaggs Institute for Chemical Biology The Worm Institute for Research and Medicine (WIRM) The Scripps Research Institute La Jolla CA 92037 USA
| | - Kim D. Janda
- Department of Chemistry Department of Immunology and Microbial Science The Skaggs Institute for Chemical Biology The Worm Institute for Research and Medicine (WIRM) The Scripps Research Institute La Jolla CA 92037 USA
| | - YoungSoo Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences Yonsei University 85 Songdogwahak-ro, Yeonsu-gu Incheon 21983 South Korea
| |
Collapse
|
12
|
Lee JC, Kim HY, Lee S, Shin J, Kim HV, Kim K, Baek S, Lee D, Jeon H, Kim D, Yang SH, Han G, Park K, Choi J, Park J, Moss JA, Janda KD, Kim Y. Discovery of Chemicals to Either Clear or Indicate Amyloid Aggregates by Targeting Memory-Impairing Anti-Parallel Aβ Dimers. Angew Chem Int Ed Engl 2020; 59:11491-11500. [PMID: 32233096 DOI: 10.1002/anie.202002574] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/31/2020] [Indexed: 01/20/2023]
Abstract
Amyloid-β (Aβ) oligomers are implicated in Alzheimer disease (AD). However, their unstable nature and heterogeneous state disrupts elucidation of their explicit role in AD progression, impeding the development of tools targeting soluble Aβ oligomers. Herein parallel and anti-parallel variants of Aβ(1-40) dimers were designed and synthesized, and their pathogenic properties in AD models characterized. Anti-parallel dimers induced cognitive impairments with increased amyloidogenesis and cytotoxicity, and this dimer was then used in a screening platform. Through screening, two FDA-approved drugs, Oxytetracycline and Sunitinib, were identified to dissociate Aβ oligomers and plaques to monomers in 5XFAD transgenic mice. In addition, fluorescent Astrophloxine was shown to detect aggregated Aβ in brain tissue and cerebrospinal fluid samples of AD mice. This screening platform provides a stable and homogeneous environment for observing Aβ interactions with dimer-specific molecules.
Collapse
Affiliation(s)
- Jinny Claire Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea.,Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Sejin Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea.,KIST School, University of Science and Technology (UST), Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Jisu Shin
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Hyunjin Vincent Kim
- KIST School, University of Science and Technology (UST), Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Kyeonghwan Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Seungyeop Baek
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea.,Department of Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Donghee Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Hanna Jeon
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - DaWon Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| | - Seung-Hoon Yang
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, 10326, South Korea
| | - Gyoonhee Han
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea.,Department of Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Keunwan Park
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangwon-do, 25451, South Korea
| | | | | | - Jason A Moss
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kim D Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - YoungSoo Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, South Korea
| |
Collapse
|
13
|
Korecka M, Figurski MJ, Landau SM, Brylska M, Alexander J, Blennow K, Zetterberg H, Jagust WJ, Trojanowski JQ, Shaw LM. Analytical and Clinical Performance of Amyloid-Beta Peptides Measurements in CSF of ADNIGO/2 Participants by an LC-MS/MS Reference Method. Clin Chem 2020; 66:587-597. [PMID: 32087019 PMCID: PMC7108496 DOI: 10.1093/clinchem/hvaa012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/20/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) amyloid-β1-42 (Aβ42) reliably detects brain amyloidosis based on its high concordance with plaque burden at autopsy and with amyloid positron emission tomography (PET) ligand retention observed in several studies. Low CSF Aβ42 concentrations in normal aging and dementia are associated with the presence of fibrillary Aβ across brain regions detected by amyloid PET imaging. METHODS An LC-MS/MS reference method for Aβ42, modified by adding Aβ40 and Aβ38 peptides to calibrators, was used to analyze 1445 CSF samples from ADNIGO/2 participants. Seventy runs were completed using 2 different lots of calibrators. For preparation of Aβ42 calibrators and controls spiking solution, reference Aβ42 standard with certified concentration was obtained from EC-JRC-IRMM (Belgium). Aβ40 and Aβ38 standards were purchased from rPeptide. Aβ42 calibrators' accuracy was established using CSF-based Aβ42 Certified Reference Materials (CRM). RESULTS CRM-adjusted Aβ42 calibrator concentrations were calculated using the regression equation Y (CRM-adjusted) = 0.89X (calibrators) + 32.6. Control samples and CSF pools yielded imprecision ranging from 6.5 to 10.2% (Aβ42) and 2.2 to 7.0% (Aβ40). None of the CSF pools showed statistically significant differences in Aβ42 concentrations across 2 different calibrator lots. Comparison of Aβ42 with Aβ42/Aβ40 showed that the ratio improved concordance with concurrent [18F]-florbetapir PET as a measure of fibrillar Aβ (n = 766) from 81 to 88%. CONCLUSIONS Long-term performance assessment substantiates our modified LC-MS/MS reference method for 3 Aβ peptides. The improved diagnostic performance of the CSF ratio Aβ42/Aβ40 suggests that Aβ42 and Aβ40 should be measured together and supports the need for an Aβ40 CRM.
Collapse
Affiliation(s)
- Magdalena Korecka
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michal J Figurski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA
| | - Magdalena Brylska
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob Alexander
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
14
|
Carandini T, Arighi A, Sacchi L, Fumagalli GG, Pietroboni AM, Ghezzi L, Colombi A, Scarioni M, Fenoglio C, De Riz MA, Marotta G, Scarpini E, Galimberti D. Testing the 2018 NIA-AA research framework in a retrospective large cohort of patients with cognitive impairment: from biological biomarkers to clinical syndromes. ALZHEIMERS RESEARCH & THERAPY 2019; 11:84. [PMID: 31615545 PMCID: PMC6794758 DOI: 10.1186/s13195-019-0543-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/27/2019] [Indexed: 12/29/2022]
Abstract
Background According to the 2018 NIA-AA research framework, Alzheimer’s disease (AD) is not defined by the clinical consequences of the disease, but by its underlying pathology, measured by biomarkers. Evidence of both amyloid-β (Aβ) and phosphorylated tau protein (p-tau) deposition—assessed interchangeably with amyloid-positron emission tomography (PET) and/or cerebrospinal fluid (CSF) analysis—is needed to diagnose AD in a living person. Our aim was to test the new NIA-AA research framework in a large cohort of cognitively impaired patients to evaluate correspondence between the clinical syndromes and the underlying pathologic process testified by biomarkers. Methods We retrospectively analysed 628 subjects referred to our centre in suspicion of dementia, who underwent CSF analysis, together with neuropsychological assessment and neuroimaging, and were diagnosed with different neurodegenerative dementias according to current criteria, or as cognitively unimpaired. Subjects were classified considering CSF biomarkers, and the prevalence of normal, AD-continuum and non-AD profiles in each clinical syndrome was calculated. The positivity threshold of each CSF biomarker was first assessed by receiver operating characteristic analysis, using Aβ-positive/negative status as determined by amyloid-PET visual reads. The agreement between CSF and amyloid-PET data was also evaluated. Results Among patients with a clinical diagnosis of AD, 94.1% were in the AD-continuum, whereas 5.5% were classified as non-AD and 0.4% were normal. The AD-continuum profile was found also in 26.2% of frontotemporal dementia, 48.6% of Lewy body dementia, 25% of atypical parkinsonism and 44.7% of vascular dementia. Biomarkers’ profile did not differ in amnestic and not amnestic mild cognitive impairment. CSF Aβ levels and amyloid-PET tracer binding negatively correlated, and the concordance between the two Aβ biomarkers was 89%. Conclusions The examination of the 2018 NIA-AA research framework in our clinical setting revealed a good, but incomplete, correspondence between the clinical syndromes and the underlying pathologic process measured by CSF biomarkers. The AD-continuum profile resulted to be a sensitive, but non-specific biomarker with regard to the clinical AD diagnosis. CSF and PET Aβ biomarkers were found to be not perfectly interchangeable to quantify the Aβ burden, possibly because they measure different aspects of AD pathology.
Collapse
Affiliation(s)
- Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy. .,Dino Ferrari Center, University of Milan, Milan, Italy.
| | - Andrea Arighi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Luca Sacchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Giorgio G Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Anna M Pietroboni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Laura Ghezzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Annalisa Colombi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Marta Scarioni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | | | - Milena A De Riz
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Giorgio Marotta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| |
Collapse
|
15
|
Tariciotti L, Casadei M, Honig LS, Teich AF, McKhann Ii GM, Tosto G, Mayeux R. Clinical Experience with Cerebrospinal Fluid Aβ42, Total and Phosphorylated Tau in the Evaluation of 1,016 Individuals for Suspected Dementia. J Alzheimers Dis 2019; 65:1417-1425. [PMID: 30149454 PMCID: PMC6218126 DOI: 10.3233/jad-180548] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Elevated total tau (tTau), 181-phosphorylated phosphorylated tau (pTau), and low amyloid-β42 (Aβ42) in cerebrospinal fluid (CSF) represent a diagnostic biomarker for Alzheimer’s disease (AD). Objective: The goal was to determine the overall accuracy of CSF Aβ42, tTau, pTau, and the Aβ42/total tau index (ATI) in a non-research, clinical setting for the diagnosis of AD. Methods: From medical records in 1,016 patients that had CSF studies for dementia over a 12-year period (2005 to 2017), we calculated the sensitivity and specificity of CSF Aβ42, tTau, and pTau and the ATI in relation to the final clinical diagnosis. Results: Compared with non-demented patients and patients with other dementias or mild cognitive impairment (MCI), the sensitivity and specificity of the recommended ATI and pTau cut-offs (ATI < 1.0 and pTau >61 pg/ml) for the diagnosis of AD were 0.88 and 0.72, respectively. Similar results were obtained comparing AD with non-demented patients only (0.88, 0.82) and AD with other types of dementia (0.81, 0.77). A subgroup of patients with presumed normal pressure hydrocephalus (n = 154) were biopsied at the time of shunt placement. Using the pathological manifestations of AD as the standard, the sensitivity was 0.83 while the specificity was 0.72. Conclusions: In a non-research setting, CSF biomarkers for AD showed a high sensitivity in accordance with previous studies, but modest specificity differentiating AD from other types of dementia or MCI. This study of unselected patients provides a valid and realistic assessment of the diagnostic accuracy of these CSF biomarkers in clinical practice.
Collapse
Affiliation(s)
| | | | - Lawrence S Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, College of Physicians and Surgeons of Columbia University and The New York Presbyterian Hospital, New York, NY, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Guy M McKhann Ii
- Department of Neurosurgery, College of Physicians and Surgeons of Columbia University and The New York Presbyterian Hospital, New York, NY, USA
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, College of Physicians and Surgeons of Columbia University and The New York Presbyterian Hospital, New York, NY, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Neurology, College of Physicians and Surgeons of Columbia University and The New York Presbyterian Hospital, New York, NY, USA.,Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
16
|
Diagnosis of Alzheimer's disease utilizing amyloid and tau as fluid biomarkers. Exp Mol Med 2019; 51:1-10. [PMID: 31073121 PMCID: PMC6509326 DOI: 10.1038/s12276-019-0250-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023] Open
Abstract
Current technological advancements in clinical and research settings have permitted a more intensive and comprehensive understanding of Alzheimer’s disease (AD). This development in knowledge regarding AD pathogenesis has been implemented to produce disease-modifying drugs. The potential for accessible and effective therapeutic methods has generated a need for detecting this neurodegenerative disorder during early stages of progression because such remedial effects are more profound when implemented during the initial, prolonged prodromal stages of pathogenesis. The aggregation of amyloid-β (Aβ) and tau isoforms are characteristic of AD; thus, they are considered core candidate biomarkers. However, research attempting to establish the reliability of Aβ and tau as biomarkers has culminated in an amalgamation of contradictory results and theories regarding the biomarker concentrations necessary for an accurate diagnosis. In this review, we consider the capabilities and limitations of fluid biomarkers collected from cerebrospinal fluid, blood, and oral, ocular, and olfactory secretions as diagnostic tools for AD, along with the impact of the integration of these biomarkers in clinical settings. Furthermore, the evolution of diagnostic criteria and novel research findings are discussed. This review is a summary and reflection of the ongoing concerted efforts to establish fluid biomarkers as a diagnostic tool and implement them in diagnostic procedures. Markers from body fluids could help clinicians diagnose Alzheimer’s disease before cognitive decline appears. After numerous setbacks in treating advanced Alzheimer’s, researchers are eager to identify biological indicators that facilitate earlier disease detection and interception. A review by YoungSoo Kim and colleagues at Yonsei University in South Korea, explores the promise of ‘fluid biomarkers,’ which enables diagnosis using cerebrospinal fluid (CSF), blood, oral, ocular, and olfactory fluid samples. Shifts in CSF levels of amyloid beta and tau, two proteins central to Alzheimer’s pathology, can reliably monitor at-risk individuals. Although CSF collection is unpleasant for patients, it remains more promising than blood, where current data for candidate fluid biomarkers are relatively inconclusive. In this review, investigations to discover safer, cheaper, and more reliable diagnostic tools to shift treatment from alleviation to prevention are introduced.
Collapse
|
17
|
Manca C, Rivasseau Jonveaux T, Roch V, Marie PY, Karcher G, Lamiral Z, Malaplate C, Verger A. Amyloid PETs are commonly negative in suspected Alzheimer’s disease with an increase in CSF phosphorylated-tau protein concentration but an Aβ42 concentration in the very high range: a prospective study. J Neurol 2019; 266:1685-1692. [DOI: 10.1007/s00415-019-09315-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/24/2022]
|
18
|
Hrubešová K, Fousková M, Habartová L, Fišar Z, Jirák R, Raboch J, Setnička V. Search for biomarkers of Alzheimer's disease: Recent insights, current challenges and future prospects. Clin Biochem 2019; 72:39-51. [PMID: 30953619 DOI: 10.1016/j.clinbiochem.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
Due to the trend of prolonged lifespan leading to higher incidence of age-related diseases, the demand for reliable biomarkers of dementia rises. In this review, we present novel biomarkers of high potential, especially those found in blood, urine or saliva, which could lead to a more comfortable patient experience and better time- and cost-effectivity, compared to the currently used diagnostic methods. We focus on biomarkers that might allow for the detection of Alzheimer's disease before its clinical manifestations. Such biomarkers might be helpful for better understanding the etiology of the disease and identifying its risk factors. Moreover, it could be a base for developing new treatment or at least help to prolong the presymptomatic stage in patients suffering from Alzheimer's disease. As potential candidates, we present, for instance, neurofilament light in both cerebrospinal fluid and blood plasma or amyloid β in plasma. Above all, we provide an overview of different approaches to the diagnostics, analyzing patient's biofluids as a whole using molecular spectroscopy. Infrared and Raman spectroscopy and especially chiroptical methods provide information not only on the chemical composition, but also on molecular structure. Therefore, these techniques are promising for the diagnostics of Alzheimer's disease, as the accumulation of amyloid β in abnormal conformation is one of the hallmarks of this disease.
Collapse
Affiliation(s)
- Kateřina Hrubešová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Markéta Fousková
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
19
|
Salvadó G, Molinuevo JL, Brugulat-Serrat A, Falcon C, Grau-Rivera O, Suárez-Calvet M, Pavia J, Niñerola-Baizán A, Perissinotti A, Lomeña F, Minguillon C, Fauria K, Zetterberg H, Blennow K, Gispert JD. Centiloid cut-off values for optimal agreement between PET and CSF core AD biomarkers. Alzheimers Res Ther 2019; 11:27. [PMID: 30902090 PMCID: PMC6429814 DOI: 10.1186/s13195-019-0478-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/27/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND The Centiloid scale has been developed to standardize measurements of amyloid PET imaging. Reference cut-off values of this continuous measurement enable the consistent operationalization of decision-making for multicentre research studies and clinical trials. In this study, we aimed at deriving reference Centiloid thresholds that maximize the agreement against core Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers in two large independent cohorts. METHODS A total of 516 participants of the ALFA+ Study (N = 205) and ADNI (N = 311) underwent amyloid PET imaging ([18F]flutemetamol and [18F]florbetapir, respectively) and core AD CSF biomarker determination using Elecsys® tests. Tracer uptake was quantified in Centiloid units (CL). Optimal Centiloid cut-offs were sought that maximize the agreement between PET and dichotomous determinations based on CSF levels of Aβ42, tTau, pTau, and their ratios, using pre-established reference cut-off values. To this end, a receiver operating characteristic analysis (ROC) was conducted, and Centiloid cut-offs were calculated as those that maximized the Youden's J Index or the overall percentage agreement recorded. RESULTS All Centiloid cut-offs fell within the range of 25-35, except for CSF Aβ42 that rendered an optimal cut-off value of 12 CL. As expected, the agreement of tau/Aβ42 ratios was higher than that of CSF Aβ42. Centiloid cut-off robustness was confirmed even when established in an independent cohort and against variations of CSF cut-offs. CONCLUSIONS A cut-off of 12 CL matches previously reported values derived against postmortem measures of AD neuropathology. Together with these previous findings, our results flag two relevant inflection points that would serve as boundary of different stages of amyloid pathology: one around 12 CL that marks the transition from the absence of pathology to subtle pathology and another one around 30 CL indicating the presence of established pathology. The derivation of robust and generalizable cut-offs for core AD biomarkers requires cohorts with adequate representation of intermediate levels. TRIAL REGISTRATION ALFA+ Study, NCT02485730 ALFA PET Sub-study, NCT02685969.
Collapse
Affiliation(s)
- Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- CIBER de Bioengeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
| | - Javier Pavia
- CIBER de Bioengeniería, Biomateriales y Nanomedicina, Madrid, Spain
- Nuclear Medicine Department, Hospital Clínic, Barcelona, Spain
- Instititut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | | | | | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- CIBER de Bioengeniería, Biomateriales y Nanomedicina, Madrid, Spain
| |
Collapse
|
20
|
Shahpasand-Kroner H, Klafki HW, Bauer C, Schuchhardt J, Hüttenrauch M, Stazi M, Bouter C, Wirths O, Vogelgsang J, Wiltfang J. A two-step immunoassay for the simultaneous assessment of Aβ38, Aβ40 and Aβ42 in human blood plasma supports the Aβ42/Aβ40 ratio as a promising biomarker candidate of Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2018; 10:121. [PMID: 30526652 PMCID: PMC6286509 DOI: 10.1186/s13195-018-0448-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/12/2018] [Indexed: 12/30/2022]
Abstract
Background The quantification of amyloid-beta (Aβ) peptides in blood plasma as potential biomarkers of Alzheimer’s disease (AD) is hampered by very low Aβ concentrations and the presence of matrix components that may interfere with the measurements. Methods We developed a two-step immunoassay for the simultaneous measurement of the relative levels of Aβ38, Aβ40 and Aβ42 in human EDTA plasma. The assay was employed for the study of 23 patients with dementia of the Alzheimer’s type (AD-D) and 17 patients with dementia due to other reasons (OD). We examined relationships with the clinical diagnosis, cerebral Aβ load as quantified by amyloid-positron emission tomography, apolipoprotein E genotype, Aβ levels and Tau protein in cerebrospinal fluid. Results Preconcentration of plasma Aβ peptides by immunoprecipitation substantially facilitated their immunological measurements. The Aβ42/Aβ40 and Aβ42/Aβ38 ratios were statistically significantly lower in the AD-D patients than in the OD group. The areas under the receiver operating characteristic curves reached 0.87 for the Aβ42/Aβ40 ratio and 0.80 for the Aβ42/Aβ38 ratio. Conclusions The measurement of plasma Aβ peptides with an immunological assay can be improved by preconcentration via immunoprecipitation with an antibody against the Aβ amino-terminus and elution of the captured peptides by heating in a mild detergent-containing buffer. Our findings support the Aβ42/Aβ40 ratio in blood plasma as a promising AD biomarker candidate which correlates significantly with the validated core biomarkers of AD. Further studies will be needed for technical advancement of the assay and validation of the biomarker findings. Electronic supplementary material The online version of this article (10.1186/s13195-018-0448-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hedieh Shahpasand-Kroner
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, D-37075, Goettingen, Germany.,Present address: Department of Neurology, David Geffen School of Medicine, Neuroscience Research Building, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Hans-W Klafki
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany
| | - Chris Bauer
- MicroDiscovery GmbH, Marienburger Strasse 1, D-10405, Berlin, Germany
| | | | - Melanie Hüttenrauch
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany
| | - Martina Stazi
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Goettingen (UMG), Georg-August-University, Robert-Koch-Str. 40, D-37075, Goettingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University, Von-Siebold-Str. 5, D-37075, Goettingen, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, D-37075, Goettingen, Germany. .,Department of Medical Science, iBiMED, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
21
|
Huang CC, Huang WM, Chen CH, Jhou ZY, The Alzheimer's Disease Neuroimaging Initiative, Lin CP. The Combination of Functional and Structural MRI Is a Potential Screening Tool in Alzheimer's Disease. Front Aging Neurosci 2018; 10:251. [PMID: 30297997 PMCID: PMC6160579 DOI: 10.3389/fnagi.2018.00251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/31/2018] [Indexed: 11/13/2022] Open
Abstract
Introduction: This study aimed to survey the discrimination power of parameters from cerebrospinal fluid (CSF) biomarkers, fluorodeoxyglucose uptake on PET (FDG-PET), structural magnetic resonance imaging (MRI), and functional MRI in high- and low-risk subjects or in converters and stable subjects of normal and mild cognitive impairment (MCI) statuses. Methods: We used baseline resting-state functional MRI (rfMRI) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset to analyze functional networks and recorded subjects' characteristics and results of the CSF study, FDG-PET, and structural MRI from the ADNI website. All parameters were evaluated based on the between-group difference among normal (NC), MCI, and Alzheimer's disease (AD) groups. The parameters other than CSF results were included to study the difference between high- and low-AD-risk subjects in NC or MCI groups, based on CSF results. On the basis of two-year follow-up conditions, all parameters were compared between stable subjects and converters in NC and MCI. Results: CSF biomarkers, FDG-PET, structural MRI, and functional MRI are all able to differentiate AD from MCI or NC but not between MCI and NC. As compared with low-AD-risk subjects, high-risk subjects present decreased FDG-PET in both MCI and NC groups but structural MRI change only in MCI status and rfMRI alteration only in NC status. As compared with stable subjects, converters have decreased FDG-PET, functional network changes, and structural changes in both MCI and NC groups. Conclusion: The combination of functional and structural MRI is a safer screening tool but with similar power as FDG-PET to reflect CSF change in the AD pathological process and to identify high-risk subjects and converters in NC and MCI.
Collapse
Affiliation(s)
- Chun-Chao Huang
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Wei-Ming Huang
- Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Chia-Hung Chen
- Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Zong-Yi Jhou
- Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - The Alzheimer's Disease Neuroimaging Initiative
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
22
|
Vatsadze SZ, Eremina OE, Veselova IA, Kalmykov SN, Nenajdenko VG. 18F-Labelled catecholamine type radiopharmaceuticals in the diagnosis of neurodegenerative diseases and neuroendocrine tumours: approaches to synthesis and development prospects. RUSSIAN CHEMICAL REVIEWS 2018. [DOI: 10.1070/rcr4752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Longitudinal structural cerebral changes related to core CSF biomarkers in preclinical Alzheimer's disease: A study of two independent datasets. NEUROIMAGE-CLINICAL 2018; 19:190-201. [PMID: 30023169 PMCID: PMC6050455 DOI: 10.1016/j.nicl.2018.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/08/2018] [Accepted: 04/14/2018] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is characterized by an accumulation of β-amyloid (Aβ42) accompanied by brain atrophy and cognitive decline. Several recent studies have shown that Aβ42 accumulation is associated with gray matter (GM) changes prior to the development of cognitive impairment, in the so-called preclinical stage of the AD (pre-AD). It also has been proved that the GM atrophy profile is not linear, both in normal ageing but, especially, on AD. However, several other factors may influence this association and may have an impact on the generalization of results from different samples. In this work, we estimate differences in rates of GM volume change in cognitively healthy elders in association with baseline core cerebrospinal fluid (CSF) AD biomarkers, and assess to what these differences are sample dependent. We report the dependence of atrophy rates, measured in a two-year interval, on Aβ42, computed both over continuous and categorical values of Aβ42, at voxel-level (p < 0.001; k < 100) and corrected for sex, age and education. Analyses were performed jointly and separately, on two samples. The first sample was formed of 31 individuals (22 Ctrl and 9 pre-AD), aged 60–80 and recruited at the Hospital Clinic of Barcelona. The second sample was a replica of the first one with subjects selected from the ADNI dataset. We also investigated the dependence of the GM atrophy rate on the basal levels of continuous p-tau and on the p-tau/Aβ42 ratio. Correlation analyses on the whole sample showed a dependence of GM atrophy rates on Aβ42 in medial and orbital frontal, precuneus, cingulate, medial temporal regions and cerebellum. Correlations with p-tau were located in the left hippocampus, parahippocampus and striatal nuclei whereas correlation with p-tau/Aβ42 was mainly found in ventral and medial temporal areas. Regarding analyses performed separately, we found a substantial discrepancy of results between samples, illustrating the complexities of comparing two independent datasets even when using the same inclusion criteria. Such discrepancies may lead to significant differences in the sample size needed to detect a particular reduction on cerebral atrophy rates in prevention trials. Higher cognitive reserve and more advanced pathological progression in the ADNI sample could partially account for the observed discrepancies. Taken together, our findings in these two samples highlight the importance of comparing and merging independent datasets to draw more robust and generalizable conclusions on the structural changes in the preclinical stages of AD. GM atrophy rates depends differently on values of CSF Aβ42 than on CSF p-tau in the preclinical stage of AD. Discrepant results were obtained. Although nominally equivalent, samples might reflect different time-windows in the AD continuum. It is necessary a further effort to standardize CSF-biomarkers measures and thresholds to make different samples to be directly comparable.
Collapse
Key Words
- AD, Alzheimer's disease
- ADNI, Alzheimer's Disease Neuroimaging Initiative
- Alzheimer's disease
- Aβ42, amyloid beta
- CDR, Clinical Dementia Rating
- CSF biomarkers
- CSF, Cerebro-Spinal Fluid
- Ctrl, control
- DI, divergences of the longitudinal deformations
- ELISA, Enzyme-Linked ImmunoSorbent Assay
- FWE, Family Wise Error
- GM, gray matter
- HCB, Hospital Clinic Barcelona
- L, left
- Longitudinal VBM
- MMSE, Mini Mental State examination
- PLR, pairwise longitudinal registration
- Preclinical Alzheimer's disease
- R, right
- ROI, region of interest
- TIV, total intracranial volume
- VBM, voxel-based morphometry
- WM, white matter
- p-tau, phosphorylated tau
- preAD, preclinical Alzheimer's disease
- t-tau, total tau
Collapse
|
24
|
Niemantsverdriet E, Ottoy J, Somers C, De Roeck E, Struyfs H, Soetewey F, Verhaeghe J, Van den Bossche T, Van Mossevelde S, Goeman J, De Deyn PP, Mariën P, Versijpt J, Sleegers K, Van Broeckhoven C, Wyffels L, Albert A, Ceyssens S, Stroobants S, Staelens S, Bjerke M, Engelborghs S. The Cerebrospinal Fluid Aβ1-42/Aβ1-40 Ratio Improves Concordance with Amyloid-PET for Diagnosing Alzheimer's Disease in a Clinical Setting. J Alzheimers Dis 2018; 60:561-576. [PMID: 28869470 PMCID: PMC5611891 DOI: 10.3233/jad-170327] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Evidence suggests that the concordance between amyloid-PET and cerebrospinal fluid (CSF) amyloid-β (Aβ) increases when the CSF Aβ1–42/Aβ1–40 ratio is used as compared to CSF Aβ1–42 levels alone. Objective: In order to test this hypothesis, we set up a prospective longitudinal study comparing the concordance between different amyloid biomarkers for Alzheimer’s disease (AD) in a clinical setting. Methods: Seventy-eight subjects (AD dementia (n = 17), mild cognitive impairment (MCI, n = 48), and cognitively healthy controls (n = 13)) underwent a [18F]Florbetapir ([18F]AV45) PET scan, [18F]FDG PET scan, MRI scan, and an extensive neuropsychological examination. In a large subset (n = 67), a lumbar puncture was performed and AD biomarkers were analyzed (Aβ1–42, Aβ1–40, T-tau, P-tau181). Results: We detected an increased concordance in the visual and quantitative (standardized uptake value ratio (SUVR) and total volume of distribution (VT)) [18F]AV45 PET measures when the CSF Aβ1–42/Aβ1–40 was applied compared to Aβ1–42 alone. CSF biomarkers were stronger associated to [18F]AV45 PET for SUVR values when considering the total brain white matter as reference region instead of cerebellar grey matter Conclusions: The concordance between CSF Aβ and [18F]AV45 PET increases when the CSF Aβ1–42/Aβ1–40 ratio is applied. This finding is of most importance for the biomarker-based diagnosis of AD as well as for selection of subjects for clinical trials with potential disease-modifying therapies for AD.
Collapse
Affiliation(s)
- Ellis Niemantsverdriet
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Julie Ottoy
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Charisse Somers
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Ellen De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Faculty of Psychology and Educational Sciences, Vrije Universiteit Brussels, Brussels, Belgium
| | - Hanne Struyfs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Femke Soetewey
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Tobi Van den Bossche
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Sara Van Mossevelde
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Johan Goeman
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Peter Paul De Deyn
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Peter Mariën
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.,Clinical and Experimental Neurolinguistics, CLIN, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jan Versijpt
- Vrije Universiteit Brussel(VUB), University Hospital Brussels (UZ Brussel), Department of Neurology, Brussels, Belgium
| | - Kristel Sleegers
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium.,Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Adrien Albert
- Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sarah Ceyssens
- Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium.,Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| |
Collapse
|
25
|
Schindler SE, Gray JD, Gordon BA, Xiong C, Batrla-Utermann R, Quan M, Wahl S, Benzinger TLS, Holtzman DM, Morris JC, Fagan AM. Cerebrospinal fluid biomarkers measured by Elecsys assays compared to amyloid imaging. Alzheimers Dement 2018; 14:1460-1469. [PMID: 29501462 DOI: 10.1016/j.jalz.2018.01.013] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/15/2017] [Accepted: 01/26/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Levels of amyloid β peptide 42 (Aβ42), total tau, and phosphorylated tau-181 are well-established cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease, but variability in manual plate-based assays has limited their use. We examined the relationship between CSF biomarkers, as measured by a novel automated immunoassay platform, and amyloid positron emission tomography. METHODS CSF samples from 200 individuals underwent separate analysis for Aβ42, total tau, and phosphorylated tau-181 with automated Roche Elecsys assays. Aβ40 was measured with a commercial plate-based assay. Positron emission tomography with Pittsburgh Compound B was performed less than 1 year from CSF collection. RESULTS Ratios of CSF biomarkers (total tau/Aβ42, phosphorylated tau-181/Aβ42, and Aβ42/Aβ40) best discriminated Pittsburgh Compound B-positive from Pittsburgh Compound B-negative individuals. DISCUSSION CSF biomarkers and amyloid positron emission tomography reflect different aspects of Alzheimer's disease brain pathology, and therefore, less-than-perfect correspondence is expected. Automated assays are likely to increase the utility of CSF biomarkers.
Collapse
Affiliation(s)
- Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Julia D Gray
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | - Tammie L S Benzinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Lewczuk P, Matzen A, Blennow K, Parnetti L, Molinuevo JL, Eusebi P, Kornhuber J, Morris JC, Fagan AM. Cerebrospinal Fluid Aβ42/40 Corresponds Better than Aβ42 to Amyloid PET in Alzheimer's Disease. J Alzheimers Dis 2018; 55:813-822. [PMID: 27792012 PMCID: PMC5147502 DOI: 10.3233/jad-160722] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Decreased concentrations of amyloid-β 1-42 (Aβ42) in cerebrospinal fluid (CSF) and increased retention of Aβ tracers in the brain on positron emission tomography (PET) are considered the earliest biomarkers of Alzheimer’s disease (AD). However, a proportion of cases show discrepancies between the results of the two biomarker modalities which may reflect inter-individual differences in Aβ metabolism. The CSF Aβ42/40 ratio seems to be a more accurate biomarker of clinical AD than CSF Aβ42 alone. Objective: We tested whether CSF Aβ42 alone or the Aβ42/40 ratio corresponds better with amyloid PET status and analyzed the distribution of cases with discordant CSF-PET results. Methods: CSF obtained from a mixed cohort (n = 200) of cognitively normal and abnormal research participants who had undergone amyloid PET within 12 months (n = 150 PET-negative, n = 50 PET-positive according to a previously published cut-off) was assayed for Aβ42 and Aβ40 using two recently developed immunoassays. Optimal CSF cut-offs for amyloid positivity were calculated, and concordance was tested by comparison of the areas under receiver operating characteristic (ROC) curves (AUC) and McNemar’s test for paired proportions. Results: CSF Aβ42/40 corresponded better than Aβ42 with PET results, with a larger proportion of concordant cases (89.4% versus 74.9%, respectively, p < 0.0001) and a larger AUC (0.936 versus 0.814, respectively, p < 0.0001) associated with the ratio. For both CSF biomarkers, the percentage of CSF-abnormal/PET-normal cases was larger than that of CSF-normal/PET-abnormal cases. Conclusion: The CSF Aβ42/40 ratio is superior to Aβ42 alone as a marker of amyloid-positivity by PET. We hypothesize that this increase in performance reflects the ratio compensating for general between-individual variations in CSF total Aβ.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Medical University of Białystok, and Department of Biochemical Diagnostics, University Hospital of Bialystok, Bialystok, Poland
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Lucilla Parnetti
- Section of Neurology, Center for Memory Disturbances, University of Perugia, Italy
| | - Jose Luis Molinuevo
- Alzheimer's disease and other cognitive disorders unit, Neurology Service, Hospital Clínic de Barcelona - Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Paolo Eusebi
- Section of Neurology, Center for Memory Disturbances, University of Perugia, Italy
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - John C Morris
- The Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- The Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
27
|
Villemagne VL, Doré V, Burnham SC, Masters CL, Rowe CC. Imaging tau and amyloid-β proteinopathies in Alzheimer disease and other conditions. Nat Rev Neurol 2018; 14:225-236. [DOI: 10.1038/nrneurol.2018.9] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
28
|
Wu KY, Hsiao IT, Chen CH, Liu CY, Hsu JL, Huang SY, Yen TC, Lin KJ. Plasma Aβ analysis using magnetically-labeled immunoassays and PET 18F-florbetapir binding in non-demented patients with major depressive disorder. Sci Rep 2018; 8:2739. [PMID: 29426824 PMCID: PMC5807319 DOI: 10.1038/s41598-018-21140-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/30/2018] [Indexed: 01/25/2023] Open
Abstract
An increased level of brain amyloid deposition and a decreased level of cerebral spinal fluid (CSF) Aβ42 are currently considered reliable biomarkers of Alzheimer’s disease (AD); however, the usefulness of plasma Aβ levels are not well-established. This study investigated the relationships between plasma Aβ levels and cerebral amyloidosis in 36 non-demented patients with major depressive disorder (MDD). All participants underwent 18F-florbetapir PET imaging and provided a blood sample at the same time for immunomagnetic reduction assay to measure the plasma levels of Aβ40 and Aβ42. We found inverse associations of the plasma Aβ42 level and the Aβ42/Aβ40 ratio, and a positive association of the plasma Aβ40 level, with cerebral amyloid deposition in the precuneus, parietal and posterior cingulate cortex. Subgroup analyses in subjects with higher 18F-florbetapir uptake values or MDD with amnestic mild cognitive impairment revealed more pervasive relationships of plasma Aβ measures with 18F-florbetapir binding across the brain regions examined. The study suggested that regional brain amyloid deposition in terms of 18F-florbetapir PET uptake had weak-to-moderate associations with plasma Aβ42 and Aβ40 levels, and the Aβ42/Aβ40 ratio. Validation in a larger population of subjects of known cerebral amyloidosis status is needed. Careful interpretation of plasma data is warranted.
Collapse
Affiliation(s)
- Kuan-Yi Wu
- Department of Psychiatry, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Ing-Tsung Hsiao
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Chia-Yih Liu
- Department of Psychiatry, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology and Dementia Center, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan.,Graduate Institute of Humanities in Medicine and Brain and Consciousness Research Center, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Yao Huang
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan. .,Department of Medical Imaging and Radiological Sciences and Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan.
| |
Collapse
|
29
|
van der Flier WM, Scheltens P. Amsterdam Dementia Cohort: Performing Research to Optimize Care. J Alzheimers Dis 2018; 62:1091-1111. [PMID: 29562540 PMCID: PMC5870023 DOI: 10.3233/jad-170850] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2017] [Indexed: 01/01/2023]
Abstract
The Alzheimer center of the VU University Medical Center opened in 2000 and was initiated to combine both patient care and research. Together, to date, all patients forming the Amsterdam Dementia Cohort number almost 6,000 individuals. In this cohort profile, we provide an overview of the results produced based on the Amsterdam Dementia Cohort. We describe the main results over the years in each of these research lines: 1) early diagnosis, 2) heterogeneity, and 3) vascular factors. Among the most important research efforts that have also impacted patients' lives and/or the research field, we count the development of novel, easy to use diagnostic measures such as visual rating scales for MRI and the Amsterdam IADL Questionnaire, insight in different subgroups of AD, and findings on incidence and clinical sequelae of microbleeds. Finally, we describe in the outlook how our research endeavors have improved the lives of our patients.
Collapse
Affiliation(s)
- Wiesje M. van der Flier
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Pereira JB, Strandberg TO, Palmqvist S, Volpe G, van Westen D, Westman E, Hansson O. Amyloid Network Topology Characterizes the Progression of Alzheimer's Disease During the Predementia Stages. Cereb Cortex 2018; 28:340-349. [PMID: 29136123 PMCID: PMC6454565 DOI: 10.1093/cercor/bhx294] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence showing that the accumulation of the amyloid-β (Aβ) peptide into extracellular plaques is a central event in Alzheimer's disease (AD). These abnormalities can be detected as lowered levels of Aβ42 in the cerebrospinal fluid (CSF) and are followed by increased amyloid burden on positron emission tomography (PET) several years before the onset of dementia. The aim of this study was to assess amyloid network topology in nondemented individuals with early stage Aβ accumulation, defined as abnormal CSF Aβ42 levels and normal Florbetapir PET (CSF+/PET-), and more advanced Aβ accumulation, defined as both abnormal CSF Aβ42 and Florbetapir PET (CSF+/PET+). The amyloid networks were built using correlations in the mean 18F-florbetapir PET values between 72 brain regions and analyzed using graph theory analyses. Our findings showed an association between early amyloid stages and increased covariance as well as shorter paths between several brain areas that overlapped with the default-mode network (DMN). Moreover, we found that individuals with more advanced amyloid accumulation showed more widespread changes in brain regions both within and outside the DMN. These findings suggest that amyloid network topology could potentially be used to assess disease progression in the predementia stages of AD.
Collapse
Affiliation(s)
- Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Tor Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, MalmöSweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Sweden
| | - Giovanni Volpe
- Department of Physics, Göteborg University, Göteborg, Sweden
| | - Danielle van Westen
- Department of Clinical Sciences Lund, Diagnostic Radiology, Lund University, Lund, Sweden
- Imaging and Function, Skåne University Health Care, Lund, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, MalmöSweden
| | | |
Collapse
|
31
|
Pawlowski M, Meuth SG, Duning T. Cerebrospinal Fluid Biomarkers in Alzheimer's Disease-From Brain Starch to Bench and Bedside. Diagnostics (Basel) 2017; 7:diagnostics7030042. [PMID: 28703785 PMCID: PMC5617942 DOI: 10.3390/diagnostics7030042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/21/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease is the most common cause of dementia. Over the last three decades, research has advanced dramatically and provided a detailed understanding of the molecular events underlying the pathogenesis of Alzheimer’s disease. In parallel, assays for the detection of biomarkers that reflect the typical Alzheimer’s disease-associated pathology have been developed and validated in myriads of clinical studies. Such biomarkers complement clinical diagnosis and improve diagnostic accuracy. The use of biomarkers will become even more important with the advent of disease-modifying therapies. Such therapies will likely be most beneficial when administered early in the disease course. Here, we summarise the development of the core Alzheimer’s disease cerebrospinal fluid biomarkers: amyloid-β and tau. We provide an overview of their role in cellular physiology and Alzheimer’s disease pathology, and embed their development as cerebrospinal fluid biomarkers into the historical context of Alzheimer’s disease research. Finally, we summarise recommendations for their use in clinical practice, and outline perspectives for novel cerebrospinal fluid candidate biomarkers.
Collapse
Affiliation(s)
- Matthias Pawlowski
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| | - Sven G Meuth
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| | - Thomas Duning
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany.
| |
Collapse
|
32
|
Mo Y, Stromswold J, Wilson K, Holder D, Sur C, Laterza O, Savage MJ, Struyk A, Scheltens P, Teunissen CE, Burke J, Macaulay SL, Bråthen G, Sando SB, White LR, Weiss C, Cowes A, Bush MM, DeSilva G, Darby DG, Rainey-Smith SR, Surls J, Sagini E, Tanen M, Altman A, Luthman J, Egan MF. A multinational study distinguishing Alzheimer's and healthy patients using cerebrospinal fluid tau/Aβ42 cutoff with concordance to amyloid positron emission tomography imaging. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2017; 6:201-209. [PMID: 28349119 PMCID: PMC5357677 DOI: 10.1016/j.dadm.2017.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction Changes in cerebrospinal fluid (CSF) tau and amyloid β (Aβ)42 accompany development of Alzheimer's brain pathology. Robust tau and Aβ42 immunoassays were developed to establish a tau/Aβ42 cutoff distinguishing mild-to-moderate Alzheimer's disease (AD) subjects from healthy elderly control (HC) subjects. Methods A CSF tau/Aβ42 cutoff criteria was chosen, which distinguished the groups and maximized concordance with amyloid PET. Performance was assessed using an independent validation cohort. Results A tau/Aβ42 = 0.215 cutoff provided 94.8% sensitivity and 77.7% specificity. Concordance with PET visual reads was estimated at 86.9% in a ∼50% PET positive population. In the validation cohort, the cutoff demonstrated 78.4% sensitivity and 84.9% specificity to distinguish the AD and HC populations. Discussion A tau/Aβ42 cutoff with acceptable sensitivity and specificity distinguished HC from mild-to-moderate AD subjects and maximized concordance to brain amyloidosis. The defined cutoff demonstrated that CSF analysis may be useful as a surrogate to imaging assessment of AD pathology.
Collapse
Affiliation(s)
- Yi Mo
- Merck & Co. Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | | | | | - Philip Scheltens
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - James Burke
- Duke Neurology, Duke University, Durham, NC, USA
| | - S Lance Macaulay
- Commonwealth Scientific and Industrial Research Organization, Parkville, Victoria, Australia
| | - Geir Bråthen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology, University Hospital of Trondheim, Trondheim, Norway
| | - Sigrid Botne Sando
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology, University Hospital of Trondheim, Trondheim, Norway
| | - Linda R White
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology, University Hospital of Trondheim, Trondheim, Norway
| | | | | | | | | | - David G Darby
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia, Australia
| | | | | | | | | | | | | |
Collapse
|
33
|
Abe Y, Kimura N, Takahashi R, Gotou M, Mizukami K, Uchida H, Matsubara E. Relationship between cytokine levels in the cerebrospinal fluid and 11C-Pittsburgh compound B retention in patients with mild cognitive impairment. Geriatr Gerontol Int 2017; 17:1907-1913. [PMID: 28261965 DOI: 10.1111/ggi.12991] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 11/01/2016] [Accepted: 11/22/2016] [Indexed: 01/05/2023]
Abstract
AIM In the present study, we examined the relationship between cytokine levels in the cerebrospinal fluid (CSF) and 11 C-Pittsburgh compound B (PiB) retention in patients with mild cognitive impairment. METHODS A total of 33 participants (12 men and 21 women; mean age 76.5 years) with mild cognitive impairment underwent neuropsychological assessments, PiB positron emission tomography and analysis of cytokine levels in the CSF. The CSF levels of 48 cytokines and growth factors were measured using multiplex immunoassays. PiB retention was assessed based on a standardized uptake value ratio. Mild cognitive impairment participants were classified as PiB-positive and PiB-negative, with a cut-off level of 1.4. We compared the CSF cytokine levels and Alzheimer's disease biomarkers, including β-amyloid 1-42, total tau and tau phosphorylated at threonine 181, between the two subgroups, and evaluated the correlation between PiB retention or CSF Alzheimer's disease biomarkers and CSF cytokine levels. RESULTS Cytokine levels in the CSF did not differ between the two subgroups. Macrophage inflammatory protein-1β levels in the CSF significantly correlated with PiB retention only in the PiB-positive subgroup, whereas stem cell growth factor-β levels significantly correlated with PiB retention in the PiB-negative subgroup. Furthermore, stem cell growth factor-β levels significantly correlated with total tau and tau phosphorylated at threonine 181 levels in only the PiB-negative subgroup. CONCLUSION The present findings suggest that macrophage inflammatory protein-1β and stem cell growth factor-β are associated with chronic inflammatory processes accompanied by amyloid deposition or AD pathophysiology. Geriatr Gerontol Int 2017; 17: 1907-1913.
Collapse
Affiliation(s)
- Yoshitake Abe
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Noriyuki Kimura
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ryuichi Takahashi
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan.,Department of Neurology, Hyogo Prefectural Rehabilitation Hospital, Hyougo, Japan
| | - Megumi Gotou
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ken Mizukami
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Hirotatsu Uchida
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| | - Etsuro Matsubara
- Department of Neurology, Oita University, Faculty of Medicine, Oita, Japan
| |
Collapse
|
34
|
Almdahl IS, Lauridsen C, Selnes P, Kalheim LF, Coello C, Gajdzik B, Møller I, Wettergreen M, Grambaite R, Bjørnerud A, Bråthen G, Sando SB, White LR, Fladby T. Cerebrospinal Fluid Levels of Amyloid Beta 1-43 Mirror 1-42 in Relation to Imaging Biomarkers of Alzheimer's Disease. Front Aging Neurosci 2017; 9:9. [PMID: 28223932 PMCID: PMC5293760 DOI: 10.3389/fnagi.2017.00009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/12/2017] [Indexed: 11/24/2022] Open
Abstract
Introduction: Amyloid beta 1-43 (Aβ43), with its additional C-terminal threonine residue, is hypothesized to play a role in early Alzheimer’s disease pathology possibly different from that of amyloid beta 1-42 (Aβ42). Cerebrospinal fluid (CSF) Aβ43 has been suggested as a potential novel biomarker for predicting conversion from mild cognitive impairment (MCI) to dementia in Alzheimer’s disease. However, the relationship between CSF Aβ43 and established imaging biomarkers of Alzheimer’s disease has never been assessed. Materials and Methods: In this observational study, CSF Aβ43 was measured with ELISA in 89 subjects; 34 with subjective cognitive decline (SCD), 51 with MCI, and four with resolution of previous cognitive complaints. All subjects underwent structural MRI; 40 subjects on a 3T and 50 on a 1.5T scanner. Forty subjects, including 24 with SCD and 12 with MCI, underwent 18F-Flutemetamol PET. Seventy-eight subjects were assessed with 18F-fluorodeoxyglucose PET (21 SCD/7 MCI and 11 SCD/39 MCI on two different scanners). Ten subjects with SCD and 39 with MCI also underwent diffusion tensor imaging. Results: Cerebrospinal fluid Aβ43 was both alone and together with p-tau a significant predictor of the distinction between SCD and MCI. There was a marked difference in CSF Aβ43 between subjects with 18F-Flutemetamol PET scans visually interpreted as negative (37 pg/ml, n = 27) and positive (15 pg/ml, n = 9), p < 0.001. Both CSF Aβ43 and Aβ42 were negatively correlated with standardized uptake value ratios for all analyzed regions; CSF Aβ43 average rho -0.73, Aβ42 -0.74. Both CSF Aβ peptides correlated significantly with hippocampal volume, inferior parietal and frontal cortical thickness and axial diffusivity in the corticospinal tract. There was a trend toward CSF Aβ42 being better correlated with cortical glucose metabolism. None of the studied correlations between CSF Aβ43/42 and imaging biomarkers were significantly different for the two Aβ peptides when controlling for multiple testing. Conclusion: Cerebrospinal fluid Aβ43 appears to be strongly correlated with cerebral amyloid deposits in the same way as Aβ42, even in non-demented patients with only subjective cognitive complaints. Regarding imaging biomarkers, there is no evidence from the present study that CSF Aβ43 performs better than the classical CSF biomarker Aβ42 for distinguishing SCD and MCI.
Collapse
Affiliation(s)
- Ina S Almdahl
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Camilla Lauridsen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Per Selnes
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Lisa F Kalheim
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| | - Christopher Coello
- Preclinical PET/CT, Institute of Basic Medical Sciences, University of Oslo Oslo, Norway
| | | | - Ina Møller
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim Trondheim, Norway
| | - Marianne Wettergreen
- Department of Neurology, Akershus University HospitalLørenskog, Norway; Department of Clinical Molecular Biology (EpiGen), Institute of Clinical Medicine, University of Oslo - Akershus University HospitalLørenskog, Norway
| | - Ramune Grambaite
- Department of Neurology, Akershus University Hospital Lørenskog, Norway
| | - Atle Bjørnerud
- The Intervention Centre, Oslo University Hospital Oslo, Norway
| | - Geir Bråthen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Sigrid B Sando
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Linda R White
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology and Clinical Neurophysiology, University Hospital of TrondheimTrondheim, Norway
| | - Tormod Fladby
- Division of Medicine and Laboratory Sciences, Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway; Department of Neurology, Akershus University HospitalLørenskog, Norway
| |
Collapse
|
35
|
Villemagne VL, Doré V, Bourgeat P, Burnham SC, Laws S, Salvado O, Masters CL, Rowe CC. Aβ-amyloid and Tau Imaging in Dementia. Semin Nucl Med 2017; 47:75-88. [DOI: 10.1053/j.semnuclmed.2016.09.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
36
|
Leuzy A, Chiotis K, Hasselbalch SG, Rinne JO, de Mendonça A, Otto M, Lleó A, Castelo-Branco M, Santana I, Johansson J, Anderl-Straub S, von Arnim CAF, Beer A, Blesa R, Fortea J, Herukka SK, Portelius E, Pannee J, Zetterberg H, Blennow K, Nordberg A. Pittsburgh compound B imaging and cerebrospinal fluid amyloid-β in a multicentre European memory clinic study. Brain 2016; 139:2540-53. [PMID: 27401520 PMCID: PMC4995359 DOI: 10.1093/brain/aww160] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/02/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to assess the agreement between data on cerebral amyloidosis, derived using Pittsburgh compound B positron emission tomography and (i) multi-laboratory INNOTEST enzyme linked immunosorbent assay derived cerebrospinal fluid concentrations of amyloid-β42; (ii) centrally measured cerebrospinal fluid amyloid-β42 using a Meso Scale Discovery enzyme linked immunosorbent assay; and (iii) cerebrospinal fluid amyloid-β42 centrally measured using an antibody-independent mass spectrometry-based reference method. Moreover, we examined the hypothesis that discordance between amyloid biomarker measurements may be due to interindividual differences in total amyloid-β production, by using the ratio of amyloid-β42 to amyloid-β40 Our study population consisted of 243 subjects from seven centres belonging to the Biomarkers for Alzheimer's and Parkinson's Disease Initiative, and included subjects with normal cognition and patients with mild cognitive impairment, Alzheimer's disease dementia, frontotemporal dementia, and vascular dementia. All had Pittsburgh compound B positron emission tomography data, cerebrospinal fluid INNOTEST amyloid-β42 values, and cerebrospinal fluid samples available for reanalysis. Cerebrospinal fluid samples were reanalysed (amyloid-β42 and amyloid-β40) using Meso Scale Discovery electrochemiluminescence enzyme linked immunosorbent assay technology, and a novel, antibody-independent, mass spectrometry reference method. Pittsburgh compound B standardized uptake value ratio results were scaled using the Centiloid method. Concordance between Meso Scale Discovery/mass spectrometry reference measurement procedure findings and Pittsburgh compound B was high in subjects with mild cognitive impairment and Alzheimer's disease, while more variable results were observed for cognitively normal and non-Alzheimer's disease groups. Agreement between Pittsburgh compound B classification and Meso Scale Discovery/mass spectrometry reference measurement procedure findings was further improved when using amyloid-β42/40 Agreement between Pittsburgh compound B visual ratings and Centiloids was near complete. Despite improved agreement between Pittsburgh compound B and centrally analysed cerebrospinal fluid, a minority of subjects showed discordant findings. While future studies are needed, our results suggest that amyloid biomarker results may not be interchangeable in some individuals.
Collapse
Affiliation(s)
- Antoine Leuzy
- 1 Department of Neurobiology, Care Science, and Society, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- 1 Department of Neurobiology, Care Science, and Society, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Steen G Hasselbalch
- 2 Danish Dementia Research Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Juha O Rinne
- 3 Division of Clinical Neurosciences, Turku University Hospital, University of Turku, Turku, Finland 4 Turku PET Centre, University of Turku, Turku, Finland
| | - Alexandre de Mendonça
- 5 Department of Neurology and Laboratory of Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Markus Otto
- 6 Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Alberto Lleó
- 7 Department of Neurology, Institut d'Investigacions Biomèdiques, Hospital de Sant Pau, Barcelona, Spain 8 Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Miguel Castelo-Branco
- 9 Institute for Nuclear Sciences Applied to Health (ICNAS), Brain Imaging Network of Portugal, Coimbra, Portugal 10 Institute for Biomedical Imaging and Life Sciences (IBILI) and Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- 11 Department of Neurology, Coimbra University Hospital, Coimbra, Portugal 12 Centre for Neuroscience and Cell Biology (CNC), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | | | - Ambros Beer
- 13 Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Rafael Blesa
- 7 Department of Neurology, Institut d'Investigacions Biomèdiques, Hospital de Sant Pau, Barcelona, Spain 8 Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Juan Fortea
- 7 Department of Neurology, Institut d'Investigacions Biomèdiques, Hospital de Sant Pau, Barcelona, Spain 8 Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Sanna-Kaisa Herukka
- 14 Department of Neurology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Erik Portelius
- 15 Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Josef Pannee
- 15 Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- 15 Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden 16 Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- 15 Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Agneta Nordberg
- 1 Department of Neurobiology, Care Science, and Society, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden 17 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
37
|
Villemagne VL, Chételat G. Neuroimaging biomarkers in Alzheimer's disease and other dementias. Ageing Res Rev 2016; 30:4-16. [PMID: 26827785 DOI: 10.1016/j.arr.2016.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/16/2022]
Abstract
In vivo imaging of β-amyloid (Aβ) has transformed the assessment of Aβ pathology and its changes over time, extending our insight into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional or global Aβ burden in the brain. This knowledge is essential for therapeutic trial recruitment and for the evaluation of anti-Aβ treatments. Although cross sectional evaluation of Aβ burden does not strongly correlate with cognitive impairment, it does correlate with cognitive (especially memory) decline and with a higher risk for conversion to AD in the aging population and MCI subjects. This suggests that Aβ deposition is a protracted pathological process starting well before the onset of symptoms. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of Aβ are required to confirm this hypothesis and further elucidate the role of Aβ deposition in the course of Alzheimer's disease.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Victoria 3084, Australia; Department of Medicine, University of Melbourne, Austin Health, Victoria 3084, Australia; The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France.
| | - Gaël Chételat
- The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France; Université de Caen Basse-Normandie, Unité Mixte de Recherche (UMR), S1077 Caen, France; Ecole Pratique des Hautes Etudes, UMR-S1077, 14000 Caen, France; Unité 1077, Centre Hospitalier Universitaire de Caen, 14000 Caen, France
| |
Collapse
|
38
|
Declercq LD, Vandenberghe R, Van Laere K, Verbruggen A, Bormans G. Drug Development in Alzheimer's Disease: The Contribution of PET and SPECT. Front Pharmacol 2016; 7:88. [PMID: 27065872 PMCID: PMC4814730 DOI: 10.3389/fphar.2016.00088] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
Clinical trials aiming to develop disease-altering drugs for Alzheimer’s disease (AD), a neurodegenerative disorder with devastating consequences, are failing at an alarming rate. Poorly defined inclusion-and outcome criteria, due to a limited amount of objective biomarkers, is one of the major concerns. Non-invasive molecular imaging techniques, positron emission tomography and single photon emission (computed) tomography (PET and SPE(C)T), allow visualization and quantification of a wide variety of (patho)physiological processes and allow early (differential) diagnosis in many disorders. PET and SPECT have the ability to provide biomarkers that permit spatial assessment of pathophysiological molecular changes and therefore objectively evaluate and follow up therapeutic response, especially in the brain. A number of specific PET/SPECT biomarkers used in support of emerging clinical therapies in AD are discussed in this review.
Collapse
Affiliation(s)
- Lieven D Declercq
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven Leuven, Belgium
| | - Alfons Verbruggen
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| |
Collapse
|
39
|
Palmqvist S, Mattsson N, Hansson O. Cerebrospinal fluid analysis detects cerebral amyloid-β accumulation earlier than positron emission tomography. Brain 2016; 139:1226-36. [PMID: 26936941 PMCID: PMC4806222 DOI: 10.1093/brain/aww015] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/23/2015] [Indexed: 11/13/2022] Open
Abstract
Cerebral accumulation of amyloid-β is thought to be the starting mechanism in Alzheimer's disease. Amyloid-β can be detected by analysis of cerebrospinal fluid amyloid-β42 or amyloid positron emission tomography, but it is unknown if any of the methods can identify an abnormal amyloid accumulation prior to the other. Our aim was to determine whether cerebrospinal fluid amyloid-β42 change before amyloid PET during preclinical stages of Alzheimer's disease. We included 437 non-demented subjects from the prospective, longitudinal Alzheimer's Disease Neuroimaging Initiative (ADNI) study. All underwent (18)F-florbetapir positron emission tomography and cerebrospinal fluid amyloid-β42 analysis at baseline and at least one additional positron emission tomography after a mean follow-up of 2.1 years (range 1.1-4.4 years). Group classifications were based on normal and abnormal cerebrospinal fluid and positron emission tomography results at baseline. We found that cases with isolated abnormal cerebrospinal fluid amyloid-β and normal positron emission tomography at baseline accumulated amyloid with a mean rate of 1.2%/year, which was similar to the rate in cases with both abnormal cerebrospinal fluid and positron emission tomography (1.2%/year, P = 0.86). The mean accumulation rate of those with isolated abnormal cerebrospinal fluid was more than three times that of those with both normal cerebrospinal fluid and positron emission tomography (0.35%/year, P = 0.018). The group differences were similar when analysing yearly change in standardized uptake value ratio of florbetapir instead of percentage change. Those with both abnormal cerebrospinal fluid and positron emission tomography deteriorated more in memory and hippocampal volume compared with the other groups (P < 0.001), indicating that they were closer to Alzheimer's disease dementia. The results were replicated after adjustments of different factors and when using different cut-offs for amyloid-β abnormality including a positron emission tomography classification based on the florbetapir uptake in regions where the initial amyloid-β accumulation occurs in Alzheimer's disease. This is the first study to show that individuals who have abnormal cerebrospinal amyloid-β42 but normal amyloid-β positron emission tomography have an increased cortical amyloid-β accumulation rate similar to those with both abnormal cerebrospinal fluid and positron emission tomography and higher rate than subjects where both modalities are normal. The results indicate that cerebrospinal fluid amyloid-β42 becomes abnormal in the earliest stages of Alzheimer's disease, before amyloid positron emission tomography and before neurodegeneration starts.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden Department of Neurology, Skåne University Hospital, Sweden
| | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden Memory Clinic, Skåne University Hospital, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden Memory Clinic, Skåne University Hospital, Sweden
| | | |
Collapse
|
40
|
Nabers A, Ollesch J, Schartner J, Kötting C, Genius J, Haußmann U, Klafki H, Wiltfang J, Gerwert K. An infrared sensor analysing label-free the secondary structure of the Abeta peptide in presence of complex fluids. JOURNAL OF BIOPHOTONICS 2016; 9:224-234. [PMID: 25808829 DOI: 10.1002/jbio.201400145] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/19/2015] [Accepted: 03/01/2015] [Indexed: 06/04/2023]
Abstract
The secondary structure change of the Abeta peptide to beta-sheet was proposed as an early event in Alzheimer's disease. The transition may be used for diagnostics of this disease in an early state. We present an Attenuated Total Reflection (ATR) sensor modified with a specific antibody to extract minute amounts of Abeta peptide out of a complex fluid. Thereby, the Abeta peptide secondary structure was determined in its physiological aqueous environment by FTIR-difference-spectroscopy. The presented results open the door for label-free Alzheimer diagnostics in cerebrospinal fluid or blood. It can be extended to further neurodegenerative diseases. An immunologic ATR-FTIR sensor for Abeta peptide secondary structure analysis in complex fluids is presented.
Collapse
Affiliation(s)
- Andreas Nabers
- Protein Research Unit Ruhr within Europe (PURE), Ruhr-University Bochum, Department of Biophysics ND04-596, Universitätsstraße 150, 44780, Bochum, Germany
| | - Julian Ollesch
- Protein Research Unit Ruhr within Europe (PURE), Ruhr-University Bochum, Department of Biophysics ND04-596, Universitätsstraße 150, 44780, Bochum, Germany
| | - Jonas Schartner
- Protein Research Unit Ruhr within Europe (PURE), Ruhr-University Bochum, Department of Biophysics ND04-596, Universitätsstraße 150, 44780, Bochum, Germany
| | - Carsten Kötting
- Protein Research Unit Ruhr within Europe (PURE), Ruhr-University Bochum, Department of Biophysics ND04-596, Universitätsstraße 150, 44780, Bochum, Germany
| | - Just Genius
- Protein Research Unit Ruhr within Europe (PURE), LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty University of Duisburg-Essen, 45147, Essen, Germany
| | - Ute Haußmann
- Protein Research Unit Ruhr within Europe (PURE), LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty University of Duisburg-Essen, 45147, Essen, Germany
| | - Hans Klafki
- Protein Research Unit Ruhr within Europe (PURE), LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty University of Duisburg-Essen, 45147, Essen, Germany
| | - Jens Wiltfang
- Protein Research Unit Ruhr within Europe (PURE), LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Medical Faculty University of Duisburg-Essen, 45147, Essen, Germany
- University Medical Center, Department of Psychiatry and Psychotherapy, Georg-August-University, 37073, Göttingen, Germany
| | - Klaus Gerwert
- Protein Research Unit Ruhr within Europe (PURE), Ruhr-University Bochum, Department of Biophysics ND04-596, Universitätsstraße 150, 44780, Bochum, Germany.
| |
Collapse
|
41
|
Lauridsen C, Sando SB, Shabnam A, Møller I, Berge G, Grøntvedt GR, Bakken IJ, Salvesen Ø, Bråthen G, White LR. Cerebrospinal Fluid Levels of Amyloid Beta 1-43 in Patients with Amnestic Mild Cognitive Impairment or Early Alzheimer's Disease: A 2-Year Follow-Up Study. Front Aging Neurosci 2016; 8:30. [PMID: 26973507 PMCID: PMC4772322 DOI: 10.3389/fnagi.2016.00030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/08/2016] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Biomarkers that will reliably predict the onset of Alzheimer's disease (AD) are urgently needed. Although cerebrospinal fluid (CSF) amyloid beta 1-42 (Aβ42), total tau, and phosphorylated tau can be used to complement the clinical diagnosis of AD, amnestic mild cognitive impairment (aMCI), the prodromal phase of AD, is heterogeneous. Biomarkers should be able to determine which patients with aMCI are at greatest risk of AD. Histological studies and animal models indicate that amyloid beta 1-43 (Aβ43) aggregates early, and may play a role in the pathological process of AD. We have examined levels of CSF Aβ43 in a 2-year longitudinal study of aMCI and early AD. MATERIALS AND METHODS Cerebrospinal fluid was collected at baseline, and after one and 2 years from patients with AD (n = 19), and patients with aMCI (n = 42). Of these, 21 progressed to AD during the 2 years of study, whereas 21 did not. Controls (n = 32) were lumbar punctured at baseline only. CSF analyses of Aβ43, Aβ42, and total tau were carried out with ELISA. RESULTS At baseline, CSF Aβ43, CSF Aβ42 and ratios with total tau could be used to separate controls from all three patient groups. CSF Aβ43, but not Aβ42, could separate patients with aMCI who progressed to AD during the 2 years of follow-up, from those that did not. The CSF total tau/Aβ43 ratio had a slightly but significantly larger area under the receiver operating characteristic curve when compared to the CSF total tau/Aβ42 ratio. CSF Aβ43 levels, but not Aβ42 levels, decreased from baseline to 2 years in the AD group. DISCUSSION AND CONCLUSION CSF Aβ43 was demonstrated to be significantly reduced in patients already by the time that aMCI or AD was diagnosed, compared to controls, and this change must have occurred during the preclinical period. Since our results suggested that CSF Aβ43 distinguishes between subgroups of patients with aMCI better than CSF Aβ42, it may prove to be a useful additional biomarker for identifying aMCI patients at greatest risk of AD.
Collapse
Affiliation(s)
- Camilla Lauridsen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Sigrid B Sando
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology, University Hospital of TrondheimTrondheim, Norway
| | - Adiba Shabnam
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Ina Møller
- Department of Neurology, University Hospital of Trondheim Trondheim, Norway
| | - Guro Berge
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Gøril R Grøntvedt
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology, University Hospital of TrondheimTrondheim, Norway
| | | | - Øyvind Salvesen
- Unit for Applied Clinical Research, Faculty of Medicine, Norwegian University of Science and Technology Trondheim, Norway
| | - Geir Bråthen
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology, University Hospital of TrondheimTrondheim, Norway
| | - Linda R White
- Department of Neuroscience, Faculty of Medicine, Norwegian University of Science and TechnologyTrondheim, Norway; Department of Neurology, University Hospital of TrondheimTrondheim, Norway
| |
Collapse
|
42
|
Rethinking on the concept of biomarkers in preclinical Alzheimer's disease. Neurol Sci 2016; 37:663-72. [PMID: 26792010 DOI: 10.1007/s10072-016-2477-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/09/2016] [Indexed: 01/01/2023]
Abstract
The neuropathological processes eventually leading to Alzheimer's disease (AD) are thought to start decades before the appearance of clinical symptoms and the clinical diagnosis of AD dementia. The term "preclinical AD" has been recently introduced to identify this "silent stage" of AD, when the disease is already present, but symptoms are not yet clinically evident. Advances in AD biomarkers have dramatically improved the ability to detect AD pathological processes in vivo in cognitively intact subjects, thus demonstrating the presence of AD pathology in the preclinical phase. This review focuses on the recent advances in the field of neuroimaging and CSF AD biomarkers specifically in the preclinical phase of AD, and aims to discuss the significance that such biomarkers could have in cognitively intact subjects. Even though the use of such biomarkers in AD preclinical phase has contributed to improve our understanding of AD early pathological processes, it raised also a number of new challenges that still remain to be overcome, such as a better definition of the clinical and individual significance of currently known biomarkers in preclinical stages and the development of novel biomarkers of different early AD-related events.
Collapse
|
43
|
Sepulcre J, Masdeu JC. Advanced Neuroimaging Methods Towards Characterization of Early Stages of Alzheimer's Disease. Methods Mol Biol 2016; 1303:509-519. [PMID: 26235088 DOI: 10.1007/978-1-4939-2627-5_31] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In the past 5 years, imaging network properties in the brain of patients with Alzheimer's disease (AD) has revolutionized our understanding of this disorder. Postmortem data had already suggested that the damage spreads along functional neural networks, but postmortem studies do not provide information on the temporal evolution of the damage in the same patient, essential to determine spreading. These data can be provided by functional and structural neuroimaging, which allow for the visualization over time of the progressive damage inflicted by AD. Functional networks can be mapped by determining the synchrony across brain regions of the blood oxygenation level dependence (BOLD) signal on functional magnetic resonance imaging (MRI) during quiet wakefulness. Other less extensively used techniques are also useful. For instance, amyloid deposition can be imaged and its progression mapped to determine whether it follows brain networks, and, if so, which are affected earliest. Network patterns of neurobiological changes, including tau deposition, may prove critical to our understanding of the neurobiology of AD and therefore open the way for therapeutic interventions.
Collapse
Affiliation(s)
- Jorge Sepulcre
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
44
|
Abstract
The aim of this study was to examine cross-sectionally whether higher cardiorespiratory fitness (CRF) might favorably modify amyloid-β (Aβ)-related decrements in cognition in a cohort of late-middle-aged adults at risk for Alzheimer's disease (AD). Sixty-nine enrollees in the Wisconsin Registry for Alzheimer's Prevention participated in this study. They completed a comprehensive neuropsychological exam, underwent 11C Pittsburgh Compound B (PiB)-PET imaging, and performed a graded treadmill exercise test to volitional exhaustion. Peak oxygen consumption (VO2peak) during the exercise test was used as the index of CRF. Forty-five participants also underwent lumbar puncture for collection of cerebrospinal fluid (CSF) samples, from which Aβ42 was immunoassayed. Covariate-adjusted regression analyses were used to test whether the association between Aβ and cognition was modified by CRF. There were significant VO2peak*PiB-PET interactions for Immediate Memory (p=.041) and Verbal Learning & Memory (p=.025). There were also significant VO2peak*CSF Aβ42 interactions for Immediate Memory (p<.001) and Verbal Learning & Memory (p<.001). Specifically, in the context of high Aβ burden, that is, increased PiB-PET binding or reduced CSF Aβ42, individuals with higher CRF exhibited significantly better cognition compared with individuals with lower CRF. In a late-middle-aged, at-risk cohort, higher CRF is associated with a diminution of Aβ-related effects on cognition. These findings suggest that exercise might play an important role in the prevention of AD.
Collapse
|
45
|
Zwan MD, Rinne JO, Hasselbalch SG, Nordberg A, Lleó A, Herukka SK, Soininen H, Law I, Bahl JMC, Carter SF, Fortea J, Blesa R, Teunissen CE, Bouwman FH, van Berckel BNM, Visser PJ. Use of amyloid-PET to determine cutpoints for CSF markers: A multicenter study. Neurology 2015; 86:50-8. [PMID: 26468410 DOI: 10.1212/wnl.0000000000002081] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/28/2015] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES To define CSF β-amyloid 1-42 (Aβ42) cutpoints to detect cortical amyloid deposition as assessed by 11C-Pittsburgh compound B ([11C]PiB)-PET and to compare these calculated cutpoints with cutpoints currently used in clinical practice. METHODS We included 433 participants (57 controls, 99 with mild cognitive impairment, 195 with Alzheimer disease [AD] dementia, and 82 with non-AD dementia) from 5 European centers. We calculated for each center and for the pooled cohort CSF Aβ42 and Aβ42/tau ratio cutpoints for cortical amyloid deposition based on visual interpretation of [11C]PiB-PET images. RESULTS Amyloid-PET-based calculated CSF Aβ42 cutpoints ranged from 521 to 616 pg/mL, whereas existing clinical-based cutpoints ranged from 400 to 550 pg/mL. Using the calculated cutpoint from the pooled sample (557 pg/mL), concordance between CSF Aβ42 and amyloid-PET was 84%. Similar concordance was found when using a dichotomized Aβ42/tau ratio. Exploratory analysis showed that participants with a positive amyloid-PET and normal CSF Aβ42 levels had higher CSF tau and phosphorylated tau levels and more often had mild cognitive impairment or AD dementia compared with participants who had negative amyloid-PET and abnormal CSF Aβ42 levels. CONCLUSIONS Amyloid-PET-based CSF Aβ42 cutpoints were higher and tended to reduce intercenter variability compared with clinical-based cutpoints. Discordant participants with normal CSF Aβ42 and a positive amyloid-PET may be more likely to have AD-related amyloid pathology than participants with abnormal CSF Aβ42 and a negative amyloid-PET. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that an amyloid-PET-based CSF Aβ42 cutpoint identifies individuals with amyloid deposition with a sensitivity of 87% and specificity of 80%.
Collapse
Affiliation(s)
- Marissa D Zwan
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands.
| | - Juha O Rinne
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Steen G Hasselbalch
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Agneta Nordberg
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Alberto Lleó
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Sanna-Kaisa Herukka
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Hilkka Soininen
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Ian Law
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Justyna M C Bahl
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Stephen F Carter
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Juan Fortea
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Rafael Blesa
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Charlotte E Teunissen
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Femke H Bouwman
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Bart N M van Berckel
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| | - Pieter J Visser
- From the Alzheimer Center & Department of Neurology (M.D.Z., F.H.B., P.J.V.) and Department of Clinical Chemistry (C.E.T.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands; Turku PET Centre and Department of Neurology (J.O.R.), University of Turku and Turku University Hospital, Finland; Danish Dementia Research Centre (S.G.H.), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of NVS (A.N., S.F.C.), Centre for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm; Department of Geriatric Medicine (A.N.), Karolinska University Hospital, Stockholm, Sweden; Memory Unit (A.L., J.F., R.B.), Department of Neurology, Hospital de Sant Pau, Barcelona; CIBERNED (A.L., J.F., R.B.), Center for Network Biomedical Research into Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain; Institute of Clinical Medicine-Neurology (S.-K.H., H.S., J.M.C.B.), University of Eastern Finland, Kuopio; Department of Clinical Physiology (I.L.), Nuclear Medicine and PET, Rigshospitalet, Copenhagen; Department of Autoimmunology and Biomarkers (J.M.C.B.), Statens Serum Institut, Copenhagen, Denmark; Wolfson Molecular Imaging Centre (S.F.C.), Institute of Brain Behaviour and Mental Health, University of Manchester, UK; Department of Radiology & Nuclear Medicine (B.N.M.v.B.), VU University Medical Center, Amsterdam; and Department of Psychiatry and Neuropsychology (P.J.V.), Maastricht University, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht, the Netherlands
| |
Collapse
|
46
|
Palmqvist S, Zetterberg H, Mattsson N, Johansson P, Minthon L, Blennow K, Olsson M, Hansson O. Detailed comparison of amyloid PET and CSF biomarkers for identifying early Alzheimer disease. Neurology 2015; 85:1240-9. [PMID: 26354982 PMCID: PMC4607601 DOI: 10.1212/wnl.0000000000001991] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/03/2015] [Indexed: 11/15/2022] Open
Abstract
Objective: To compare the diagnostic accuracy of CSF biomarkers and amyloid PET for diagnosing early-stage Alzheimer disease (AD). Methods: From the prospective, longitudinal BioFINDER study, we included 122 healthy elderly and 34 patients with mild cognitive impairment who developed AD dementia within 3 years (MCI-AD). β-Amyloid (Aβ) deposition in 9 brain regions was examined with [18F]-flutemetamol PET. CSF was analyzed with INNOTEST and EUROIMMUN ELISAs. The results were replicated in 146 controls and 64 patients with MCI-AD from the Alzheimer's Disease Neuroimaging Initiative study. Results: The best CSF measures for identifying MCI-AD were Aβ42/total tau (t-tau) and Aβ42/hyperphosphorylated tau (p-tau) (area under the curve [AUC] 0.93–0.94). The best PET measures performed similarly (AUC 0.92–0.93; anterior cingulate, posterior cingulate/precuneus, and global neocortical uptake). CSF Aβ42/t-tau and Aβ42/p-tau performed better than CSF Aβ42 and Aβ42/40 (AUC difference 0.03–0.12, p < 0.05). Using nonoptimized cutoffs, CSF Aβ42/t-tau had the highest accuracy of all CSF/PET biomarkers (sensitivity 97%, specificity 83%). The combination of CSF and PET was not better than using either biomarker separately. Conclusions: Amyloid PET and CSF biomarkers can identify early AD with high accuracy. There were no differences between the best CSF and PET measures and no improvement when combining them. Regional PET measures were not better than assessing the global Aβ deposition. The results were replicated in an independent cohort using another CSF assay and PET tracer. The choice between CSF and amyloid PET biomarkers for identifying early AD can be based on availability, costs, and doctor/patient preferences since both have equally high diagnostic accuracy. Classification of evidence: This study provides Class III evidence that amyloid PET and CSF biomarkers identify early-stage AD equally accurately.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ritter A, Cummings J. Fluid Biomarkers in Clinical Trials of Alzheimer's Disease Therapeutics. Front Neurol 2015; 6:186. [PMID: 26379620 PMCID: PMC4553391 DOI: 10.3389/fneur.2015.00186] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/10/2015] [Indexed: 02/02/2023] Open
Abstract
With the demographic shift of the global population toward longer life expectancy, the number of people living with Alzheimer’s disease (AD) has rapidly expanded and is projected to triple by the year 2050. Current treatments provide symptomatic relief but do not affect the underlying pathology of the disease. Therapies that prevent or slow the progression of the disease are urgently needed to avoid this growing public health emergency. Insights gained from decades of research have begun to unlock the pathophysiology of this complex disease and have provided targets for disease-modifying therapies. In the last decade, few therapeutic agents designed to modify the underlying disease process have progressed to clinical trials and none have been brought to market. With the focus on disease modification, biomarkers promise to play an increasingly important role in clinical trials. Six biomarkers have now been included in diagnostic criteria for AD and are regularly incorporated into clinical trials. Three biomarkers are neuroimaging measures – hippocampal atrophy measured by magnetic resonance imaging (MRI), amyloid uptake as measured by Pittsburg compound B positron emission tomography (PiB-PET), and decreased fluorodeoxyglucose (18F) uptake as measured by PET (FDG-PET) – and three are sampled from fluid sources – cerebrospinal fluid levels of amyloid β42 (Aβ42), total tau, and phosphorylated tau. Fluid biomarkers are important because they can provide information regarding the underlying biochemical processes that are occurring in the brain. The purpose of this paper is to review the literature regarding the existing and emerging fluid biomarkers and to examine how fluid biomarkers have been incorporated into clinical trials.
Collapse
Affiliation(s)
- Aaron Ritter
- Cleveland Clinic Lou Ruvo Center for Brain Health , Las Vegas, NV , USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health , Las Vegas, NV , USA
| |
Collapse
|
48
|
Toledo JB, Bjerke M, Da X, Landau SM, Foster NL, Jagust W, Jack C, Weiner M, Davatzikos C, Shaw LM, Trojanowski JQ. Nonlinear Association Between Cerebrospinal Fluid and Florbetapir F-18 β-Amyloid Measures Across the Spectrum of Alzheimer Disease. JAMA Neurol 2015; 72:571-81. [PMID: 25822737 DOI: 10.1001/jamaneurol.2014.4829] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IMPORTANCE Cerebrospinal fluid (CSF) and positron emission tomographic (PET) amyloid biomarkers have been proposed for the detection of Alzheimer disease (AD) pathology in living patients and for the tracking of longitudinal changes, but the relation between biomarkers needs further study. OBJECTIVE To determine the association between CSF and PET amyloid biomarkers (cross-sectional and longitudinal measures) and compare the cutoffs for these measures. DESIGN, SETTING, AND PARTICIPANTS Longitudinal clinical cohort study from 2005 to 2014 including 820 participants with at least 1 florbetapir F-18 (hereafter referred to as simply florbetapir)-PET scan and at least 1 CSF β-amyloid 1-42 (Aβ1-42) sample obtained within 30 days of each other (501 participants had a second PET scan after 2 years, including 150 participants with CSF Aβ1-42 measurements). Data were obtained from the Alzheimer's Disease Neuroimaging Initiative database. MAIN OUTCOMES AND MEASURES Four different PET scans processing pipelines from 2 different laboratories were compared. The PET cutoff values were established using a mixture-modeling approach, and different mathematical models were applied to define the association between CSF and PET amyloid measures. RESULTS The values of the CSF Aβ1-42 samples and florbetapir-PET scans showed a nonlinear association (R2 = 0.48-0.66), with the strongest association for values in the middle range. The presence of a larger dynamic range of florbetapir-PET scan values in the higher range compared with the CSF Aβ1-42 plateau explained the differences in correlation with cognition (R2 = 0.36 and R2 = 0.25, respectively). The APOE genotype significantly modified the association between both biomarkers. The PET cutoff values derived from an unsupervised classifier converged with previous PET cutoff values and the established CSF Aβ1-42 cutoff levels. There was no association between longitudinal Aβ1-42 levels and standardized uptake value ratios during follow-up. CONCLUSIONS AND RELEVANCE The association between both biomarkers is limited to a middle range of values, is modified by the APOE genotype, and is absent for longitudinal changes; 4 different approaches in 2 different platforms converge on similar pathological Aβ cutoff levels; and different pipelines to process PET scans showed correlated but not identical results. Our findings suggest that both biomarkers measure different aspects of AD Aβ pathology.
Collapse
Affiliation(s)
- Jon B Toledo
- Institute on Aging, Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Maria Bjerke
- Institute on Aging, Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Xiao Da
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley
| | - Norman L Foster
- Center for Alzheimer's Care, Imaging and Research, Department of Neurology, University of Utah, Salt Lake City
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley
| | - Clifford Jack
- Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael Weiner
- Center for Imaging of Neurodegenerative Diseases, Department of Radiology, San Francisco VA Medical Center/University of California, San Francisco
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Leslie M Shaw
- Institute on Aging, Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John Q Trojanowski
- Institute on Aging, Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | |
Collapse
|
49
|
Schindler SE, Fagan AM. Autosomal Dominant Alzheimer Disease: A Unique Resource to Study CSF Biomarker Changes in Preclinical AD. Front Neurol 2015; 6:142. [PMID: 26175713 PMCID: PMC4483518 DOI: 10.3389/fneur.2015.00142] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/12/2015] [Indexed: 12/27/2022] Open
Abstract
Our understanding of the pathogenesis of Alzheimer disease (AD) has been greatly influenced by investigation of rare families with autosomal dominant mutations that cause early onset AD. Mutations in the genes coding for amyloid precursor protein (APP), presenilin 1 (PSEN-1), and presenilin 2 (PSEN-2) cause over-production of the amyloid-β peptide (Aβ) leading to early deposition of Aβ in the brain, which in turn is hypothesized to initiate a cascade of processes, resulting in neuronal death, cognitive decline, and eventual dementia. Studies of cerebrospinal fluid (CSF) from individuals with the common form of AD, late-onset AD (LOAD), have revealed that low CSF Aβ42 and high CSF tau are associated with AD brain pathology. Herein, we review the literature on CSF biomarkers in autosomal dominant AD (ADAD), which has contributed to a detailed road map of AD pathogenesis, especially during the preclinical period, prior to the appearance of any cognitive symptoms. Current drug trials are also taking advantage of the unique characteristics of ADAD and utilizing CSF biomarkers to accelerate development of effective therapies for AD.
Collapse
Affiliation(s)
- Suzanne Elizabeth Schindler
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine , St. Louis, MO , USA
| | - Anne M Fagan
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
50
|
Babić M, Svob Štrac D, Mück-Šeler D, Pivac N, Stanić G, Hof PR, Simić G. Update on the core and developing cerebrospinal fluid biomarkers for Alzheimer disease. Croat Med J 2015; 55:347-65. [PMID: 25165049 PMCID: PMC4157375 DOI: 10.3325/cmj.2014.55.347] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer disease (AD) is a complex neurodegenerative disorder, whose prevalence will dramatically rise by 2050. Despite numerous clinical trials investigating this disease, there is still no effective treatment. Many trials showed negative or inconclusive results, possibly because they recruited only patients with severe disease, who had not undergone disease-modifying therapies in preclinical stages of AD before severe degeneration occurred. Detection of AD in asymptomatic at risk individuals (and a few presymptomatic individuals who carry an autosomal dominant monogenic AD mutation) remains impractical in many of clinical situations and is possible only with reliable biomarkers. In addition to early diagnosis of AD, biomarkers should serve for monitoring disease progression and response to therapy. To date, the most promising biomarkers are cerebrospinal fluid (CSF) and neuroimaging biomarkers. Core CSF biomarkers (amyloid β1-42, total tau, and phosphorylated tau) showed a high diagnostic accuracy but were still unreliable for preclinical detection of AD. Hence, there is an urgent need for detection and validation of novel CSF biomarkers that would enable early diagnosis of AD in asymptomatic individuals. This article reviews recent research advances on biomarkers for AD, focusing mainly on the CSF biomarkers. In addition to core CSF biomarkers, the potential usefulness of novel CSF biomarkers is discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Goran Simić
- Goran Šimić, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000 Zagreb, Croatia,
| |
Collapse
|