1
|
Yang S, Zhang X, Du X, Yan P, Zhang J, Wang W, Wang J, Zhang L, Sun H, Liu Y, Xu X, Di Y, Zhong J, Wu C, Reinhardt JD, Zheng Y, Wu T. Prediction of cognitive conversion within the Alzheimer's disease continuum using deep learning. Alzheimers Res Ther 2025; 17:41. [PMID: 39948600 PMCID: PMC11823042 DOI: 10.1186/s13195-025-01686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/28/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Early diagnosis and accurate prognosis of cognitive decline in Alzheimer's disease (AD) is important to timely assignment to optimal treatment modes. We aimed to develop a deep learning model to predict cognitive conversion to guide re-assignment decisions to more intensive therapies where needed. METHODS Longitudinal data including five variable sets, i.e. demographics, medical history, neuropsychological outcomes, laboratory and neuroimaging results, from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort were analyzed. We first developed a deep learning model to predicted cognitive conversion using all five variable sets. We then gradually removed variable sets to obtained parsimonious models for four different years of forecasting after baseline within acceptable frames of reduction in overall model fit (AUC remaining > 0.8). RESULTS A total of 607 individuals were included at baseline, of whom 538 participants were followed up at 12 months, 482 at 24 months, 268 at 36 months and 280 at 48 months. Predictive performance was excellent with AUCs ranging from 0.87 to 0.92 when all variable sets were considered. Parsimonious prediction models that still had a good performance with AUC 0.80-0.84 were established, each only including two variable sets. Neuropsychological outcomes were included in all parsimonious models. In addition, biomarker was included at year 1 and year 2, imaging data at year 3 and demographics at year 4. Under our pre-set threshold, the rate of upgrade to more intensive therapies according to predicted cognitive conversion was always higher than according to actual cognitive conversion so as to decrease the false positive rate, indicating the proportion of patients who would have missed upgraded treatment based on prognostic models although they actually needed it. CONCLUSIONS Neurophysiological tests combined with other indicator sets that vary along the AD continuum can improve can provide aid for clinical treatment decisions leading to improved management of the disease. TRAIL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT00106899 (Registration Date: 31 March 2005).
Collapse
Affiliation(s)
- Siyu Yang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of Neurology, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Xintong Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinyu Du
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Peng Yan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jing Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Wei Wang
- Department of Neurology, Nanjing Second Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Jing Wang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shanxi, 710061, China
| | - Lei Zhang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shanxi, 710061, China
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia
- Central Clinical School, Faculty of Medicine, Monash University, Melbourne, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Huaiqing Sun
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yin Liu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xinran Xu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yaxuan Di
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jin Zhong
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Caiyun Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jan D Reinhardt
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, Sichuan, 610207, China.
- Swiss Paraplegic Research, Nottwil, Switzerland.
- Department of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland.
| | - Yu Zheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ting Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
2
|
Panza F, Dibello V, Sardone R, Zupo R, Castellana F, Leccisotti I, Moretti MC, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Resta E, Lozupone M. Successes and failures: the latest advances in the clinical development of amyloid-β-targeting monoclonal antibodies for treating Alzheimer's disease. Expert Opin Biol Ther 2025:1-9. [PMID: 39908579 DOI: 10.1080/14712598.2025.2463963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION The amyloid cascade hypothesis postulated that the accumulation of amyloid-β (Aβ) was the first step of the Alzheimer's disease (AD) pathological process. Effective reduction of Aβ plaque load by numerous drug candidates, among which anti-Aβ monoclonal antibodies, has produced discussible clinical successes and several failures. It was questioned whether Aβ may be the principal AD pathogenic factor and a valid therapeutic target and if targeting Aβ different species could make the difference. AREAS COVERED This review article summarized successes and failures of anti-Aβ monoclonal antibody therapy for AD, delineating the latest advances for their clinical development also according to their target engagement and downstream biomarkers. EXPERT OPINION The preliminary success of the recent Phase III randomized clinical trials (RCTs) of lecanemab, donanemab, and remternetug, and lessons learned from the failure of previous anti-Aβ monoclonal antibodies RCTs, provided critical evidence to support the role of Aβ in AD pathogenesis. The loss of free Aβ instead of an Aβ toxicity may promote AD neuropathology. Cerebrospinal fluid analyses (i.e. increases in Aβ1-42) may indicate a potential benefit of anti-Aβ monoclonal antibodies in AD and downstream biomarkers should be considered for providing comprehension in cognitive and clinical efficacy of future AD RCTs.
Collapse
Affiliation(s)
- Francesco Panza
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Vittorio Dibello
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Health Authority of Taranto, Taranto, Italy
| | - Roberta Zupo
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Fabio Castellana
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Ivana Leccisotti
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Claudia Moretti
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Emanuela Resta
- Translational Medicine and Health System Management, Department of Economy, University of Foggia, Foggia, Italy
| | | |
Collapse
|
3
|
Raji CA, Meysami S, Porter VR, Merrill DA, Mendez MF. Diagnostic utility of brain MRI volumetry in comparing traumatic brain injury, Alzheimer disease and behavioral variant frontotemporal dementia. BMC Neurol 2024; 24:337. [PMID: 39261753 PMCID: PMC11389120 DOI: 10.1186/s12883-024-03844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Brain MRI with volumetric quantification, MRI volumetry, can improve diagnostic delineation of patients with neurocognitive disorders by identifying brain atrophy that may not be evident on visual assessments. OBJECTIVE To investigate diagnostic utility of MRI volumetry in traumatic brain injury (TBI), early-onset Alzheimer disease (EOAD), late-onset Alzheimer disease, and behavioral variant frontotemporal dementia (bvFTD). METHOD We utilized 137 participants of TBI (n = 40), EOAD (n = 45), LOAD (n = 32), and bvFTD (n = 20). Participants had 3D T1 brain MRI imaging amendable to MRI volumetry. Scan volumes were analyzed with Neuroreader. One-way ANOVA compared brain volumes across diagnostic groups. Discriminant analysis was done with leave-one-out cross validation on Neuroreader metrics to determine diagnostic delineation across groups. RESULT LOAD was the oldest compared to other groups (F = 27.5, p < .001). There were no statistically significant differences in sex (p = .58) with women comprising 54.7% of the entire cohort. EOAD and LOAD had the lowest Mini-Mental State Exam (MMSE) scores compared to TBI (p = .04 for EOAD and p = .01 for LOAD). LOAD had lowest hippocampal volumes (Left Hippocampus F = 13.1, Right Hippocampus F = 7.3, p < .001), low white matter volume in TBI (F = 5.9, p < .001), lower left parietal lobe volume in EOAD (F = 9.4, p < .001), and lower total gray matter volume in bvFTD (F = 32.8, p < .001) and caudate atrophy (F = 1737.5, p < .001). Areas under the curve ranged from 92.3 to 100%, sensitivity between 82.2 and 100%, specificity of 78.1-100%. TBI was the most accurately delineated diagnosis. Predictive features included caudate, frontal, parietal, temporal lobar and total white matter volumes. CONCLUSION We identified the diagnostic utility of regional volumetric differences across multiple neurocognitive disorders. Brain MRI volumetry is widely available and can be applied in distinguishing these disorders.
Collapse
Affiliation(s)
- Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA.
- Department of Neurology, Washington University, St. Louis, MO, USA.
| | - Somayeh Meysami
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Verna R Porter
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
- Providence Saint John's Health Center, Santa Monica, CA, USA
| | - David A Merrill
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
- Providence Saint John's Health Center, Santa Monica, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Mario F Mendez
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Greater Los Angeles VA Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
4
|
Abdul Manap AS, Almadodi R, Sultana S, Sebastian MG, Kavani KS, Lyenouq VE, Shankar A. Alzheimer's disease: a review on the current trends of the effective diagnosis and therapeutics. Front Aging Neurosci 2024; 16:1429211. [PMID: 39185459 PMCID: PMC11341404 DOI: 10.3389/fnagi.2024.1429211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
The most prevalent cause of dementia is Alzheimer's disease. Cognitive decline and accelerating memory loss characterize it. Alzheimer's disease advances sequentially, starting with preclinical stages, followed by mild cognitive and/or behavioral impairment, and ultimately leading to Alzheimer's disease dementia. In recent years, healthcare providers have been advised to make an earlier diagnosis of Alzheimer's, prior to individuals developing Alzheimer's disease dementia. Regrettably, the identification of early-stage Alzheimer's disease in clinical settings can be arduous due to the tendency of patients and healthcare providers to disregard symptoms as typical signs of aging. Therefore, accurate and prompt diagnosis of Alzheimer's disease is essential in order to facilitate the development of disease-modifying and secondary preventive therapies prior to the onset of symptoms. There has been a notable shift in the goal of the diagnosis process, transitioning from merely confirming the presence of symptomatic AD to recognizing the illness in its early, asymptomatic phases. Understanding the evolution of disease-modifying therapies and putting effective diagnostic and therapeutic management into practice requires an understanding of this concept. The outcomes of this study will enhance in-depth knowledge of the current status of Alzheimer's disease's diagnosis and treatment, justifying the necessity for the quest for potential novel biomarkers that can contribute to determining the stage of the disease, particularly in its earliest stages. Interestingly, latest clinical trial status on pharmacological agents, the nonpharmacological treatments such as behavior modification, exercise, and cognitive training as well as alternative approach on phytochemicals as neuroprotective agents have been covered in detailed.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- Department of Biomedical Science, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Reema Almadodi
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Shirin Sultana
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | | | | | - Vanessa Elle Lyenouq
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Aravind Shankar
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| |
Collapse
|
5
|
Guo T, Li A, Sun P, He Z, Cai Y, Lan G, Liu L, Li J, Yang J, Zhu Y, Zhao R, Chen X, Shi D, Liu Z, Wang Q, Xu L, Zhou L, Ran P, Wang X, Sun K, Lu J, Han Y. Astrocyte reactivity is associated with tau tangle load and cortical thinning in Alzheimer's disease. Mol Neurodegener 2024; 19:58. [PMID: 39080744 PMCID: PMC11290175 DOI: 10.1186/s13024-024-00750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND It is not fully established whether plasma β-amyloid(Aβ)42/Aβ40 and phosphorylated Tau181 (p-Tau181) can effectively detect Alzheimer's disease (AD) pathophysiology in older Chinese adults and how these biomarkers correlate with astrocyte reactivity, Aβ plaque deposition, tau tangle aggregation, and neurodegeneration. METHODS We recruited 470 older adults and analyzed plasma Aβ42/Aβ40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. Among them, 301, 195, and 70 underwent magnetic resonance imaging, Aβ and tau positron emission tomography imaging. The plasma Aβ42/Aβ40 and p-Tau181 thresholds were defined as ≤0.0609 and ≥2.418 based on the receiver operating characteristic curve analysis using the Youden index by comparing Aβ-PET negative cognitively unimpaired individuals and Aβ-PET positive cognitively impaired patients. To evaluate the feasibility of using plasma Aβ42/Aβ40 (A) and p-Tau181 (T) to detect AD and understand how astrocyte reactivity affects this process, we compared plasma GFAP, Aβ plaque, tau tangle, plasma NfL, hippocampal volume, and temporal-metaROI cortical thickness between different plasma A/T profiles and explored their relations with each other using general linear models, including age, sex, APOE-ε4, and diagnosis as covariates. RESULTS Plasma A+/T + individuals showed the highest levels of astrocyte reactivity, Aβ plaque, tau tangle, and axonal degeneration, and the lowest hippocampal volume and temporal-metaROI cortical thickness. Lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were independently and synergistically correlated with higher plasma GFAP and Aβ plaque. Elevated plasma p-Tau181 and GFAP concentrations were directly and interactively associated with more tau tangle formation. Regarding neurodegeneration, higher plasma p-Tau181 and GFAP concentrations strongly correlated with more axonal degeneration, as measured by plasma NfL, and lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were related to greater hippocampal atrophy. Higher plasma GFAP levels were associated with thinner cortical thickness and significantly interacted with lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 in predicting more temporal-metaROI cortical thinning. Voxel-wise imaging analysis confirmed these findings. DISCUSSION This study provides a valuable reference for using plasma biomarkers to detect AD in the Chinese community population and offers novel insights into how astrocyte reactivity contributes to AD progression, highlighting the importance of targeting reactive astrogliosis to prevent AD.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Pan Sun
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Zhengbo He
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Guoyu Lan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lin Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jieyin Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jie Yang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yalin Zhu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Dai Shi
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Qingyong Wang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, 518107, China
| | - Linsen Xu
- Department of Medical Imaging, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, China
| | - Liemin Zhou
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jie Lu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|
6
|
Lozupone M, Dibello V, Sardone R, Castellana F, Zupo R, Lampignano L, Bortone I, Stallone R, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Panza F. Lessons learned from the failure of solanezumab as a prospective treatment strategy for Alzheimer's disease. Expert Opin Drug Discov 2024; 19:639-647. [PMID: 38685682 DOI: 10.1080/17460441.2024.2348142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION In the last decade, the efforts conducted for discovering Alzheimer's Disease (AD) treatments targeting the best-known pathogenic factors [amyloid-β (Aβ), tau protein, and neuroinflammation] were mostly unsuccessful. Given that a systemic failure of Aβ clearance was supposed to primarily contribute to AD development and progression, disease-modifying therapies with anti-Aβ monoclonal antibodies (e.g. solanezumab, bapineuzumab, gantenerumab, aducanumab, lecanemab and donanemab) are ongoing in randomized clinical trials (RCTs) with contrasting results. AREAS COVERED The present Drug Discovery Case History analyzes the failures of RCTs of solanezumab on AD. Furthermore, the authors review the pharmacokinetics, pharmacodynamics, and tolerability effect of solanezumab from preclinical studies with its analogous m266 in mice. Finally, they describe the RCTs with cognitive, cerebrospinal fluid and neuroimaging findings in mild-to-moderate AD (EXPEDITION studies) and in secondary prevention studies (A4 and DIAN-TU). EXPERT OPINION Solanezumab was one of the first anti-Aβ monoclonal antibodies to be tested in preclinical and clinical AD showing to reduce brain Aβ level by acting on soluble monomeric form of Aβ peptide without significant results on deposits. Unfortunately, this compound showed to accelerate cognitive decline in both asymptomatic and symptomatic trial participants, and this failure of solanezumab further questioned the Aβ cascade hypothesis of AD.
Collapse
Affiliation(s)
- Madia Lozupone
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari Aldo Moro, Bari, Italy
| | - Vittorio Dibello
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Health Authority of Taranto, Taranto, Italy
| | - Fabio Castellana
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Roberta Zupo
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | | | - Ilaria Bortone
- Local Healthcare Authority of Bari, ASL Bari, Bari, Italy
| | - Roberta Stallone
- Neuroscience and Education, Human Resources Excellence in Research, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
7
|
Vos SJB, Delvenne A, Jack CR, Thal DR, Visser PJ. The clinical importance of suspected non-Alzheimer disease pathophysiology. Nat Rev Neurol 2024; 20:337-346. [PMID: 38724589 DOI: 10.1038/s41582-024-00962-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
The development of biomarkers for Alzheimer disease (AD) has led to the origin of suspected non-AD pathophysiology (SNAP) - a heterogeneous biomarker-based concept that describes individuals with normal amyloid and abnormal tau and/or neurodegeneration biomarker status. In this Review, we describe the origins of the SNAP construct, along with its prevalence, diagnostic and prognostic implications, and underlying neuropathology. As we discuss, SNAP can be operationalized using different biomarker modalities, which could affect prevalence estimates and reported characteristics of SNAP in ways that are not yet fully understood. Moreover, the underlying aetiologies that lead to a SNAP biomarker profile, and whether SNAP is the same in people with and without cognitive impairment, remains unclear. Improved insight into the clinical characteristics and pathophysiology of SNAP is of major importance for research and clinical practice, as well as for trial design to optimize care and treatment of individuals with SNAP.
Collapse
Affiliation(s)
- Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | - Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
8
|
De Kort AM, Kaushik K, Kuiperij HB, Jäkel L, Li H, Tuladhar AM, Terwindt GM, Wermer MJH, Claassen JAHR, Klijn CJM, Verbeek MM, Kessels RPC, Schreuder FHBM. The relation of a cerebrospinal fluid profile associated with Alzheimer's disease with cognitive function and neuropsychiatric symptoms in sporadic cerebral amyloid angiopathy. Alzheimers Res Ther 2024; 16:99. [PMID: 38704569 PMCID: PMC11069247 DOI: 10.1186/s13195-024-01454-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/07/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Patients with sporadic cerebral amyloid angiopathy (sCAA) frequently report cognitive or neuropsychiatric symptoms. The aim of this study is to investigate whether in patients with sCAA, cognitive impairment and neuropsychiatric symptoms are associated with a cerebrospinal fluid (CSF) biomarker profile associated with Alzheimer's disease (AD). METHODS In this cross-sectional study, we included participants with sCAA and dementia- and stroke-free, age- and sex-matched controls, who underwent a lumbar puncture, brain MRI, cognitive assessments, and self-administered and informant-based-questionnaires on neuropsychiatric symptoms. CSF phosphorylated tau, total tau and Aβ42 levels were used to divide sCAA patients in two groups: CAA with (CAA-AD+) or without a CSF biomarker profile associated with AD (CAA-AD-). Performance on global cognition, specific cognitive domains (episodic memory, working memory, processing speed, verbal fluency, visuoconstruction, and executive functioning), presence and severity of neuropsychiatric symptoms, were compared between groups. RESULTS sCAA-AD+ (n=31; mean age: 72 ± 6; 42%, 61% female) and sCAA-AD- (n=23; 70 ± 5; 42% female) participants did not differ with respect to global cognition or type of affected cognitive domain(s). The number or severity of neuropsychiatric symptoms also did not differ between sCAA-AD+ and sCAA-AD- participants. These results did not change after exclusion of patients without prior ICH. CONCLUSIONS In participants with sCAA, a CSF biomarker profile associated with AD does not impact global cognition or specific cognitive domains, or the presence of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Anna M De Kort
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kanishk Kaushik
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lieke Jäkel
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hao Li
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marieke J H Wermer
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jurgen A H R Claassen
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Catharina J M Klijn
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Roy P C Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Radboud Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Psychology, Radboud University Medical Center, Nijmegen, The Netherlands
- Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
9
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Janelidze S, Blennow K, Palmqvist S, Stomrud E, Mattsson-Carlgren N, Hansson O. Cerebrospinal fluid reference proteins increase accuracy and interpretability of biomarkers for brain diseases. Nat Commun 2024; 15:3676. [PMID: 38693142 PMCID: PMC11063138 DOI: 10.1038/s41467-024-47971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers reflect brain pathophysiology and are used extensively in translational research as well as in clinical practice for diagnosis of neurological diseases, e.g., Alzheimer's disease (AD). However, CSF biomarker concentrations may be influenced by non-disease related inter-individual variability. Here we use a data-driven approach to demonstrate the existence of inter-individual variability in mean standardized CSF protein levels. We show that these non-disease related differences cause many commonly reported CSF biomarkers to be highly correlated, thereby producing misleading results if not accounted for. To adjust for this inter-individual variability, we identified and evaluated high-performing reference proteins which improved the diagnostic accuracy of key CSF AD biomarkers. Our reference protein method attenuates the risk for false positive findings, and improves the sensitivity and specificity of CSF biomarkers, with broad implications for both research and clinical practice.
Collapse
Affiliation(s)
- Linda Karlsson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
| | - Jacob Vogel
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Clinical Sciences, Clinical Memory Research Unit, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences in Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
10
|
Fujishima M, Kawasaki Y, Mitsuhashi T, Matsuda H. Impact of amyloid and tau positivity on longitudinal brain atrophy in cognitively normal individuals. Alzheimers Res Ther 2024; 16:77. [PMID: 38600602 PMCID: PMC11005141 DOI: 10.1186/s13195-024-01450-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Individuals on the preclinical Alzheimer's continuum, particularly those with both amyloid and tau positivity (A + T +), display a rapid cognitive decline and elevated disease progression risk. However, limited studies exist on brain atrophy trajectories within this continuum over extended periods. METHODS This study involved 367 ADNI participants grouped based on combinations of amyloid and tau statuses determined through cerebrospinal fluid tests. Using longitudinal MRI scans, brain atrophy was determined according to the whole brain, lateral ventricle, and hippocampal volumes and cortical thickness in AD-signature regions. Cognitive performance was evaluated with the Preclinical Alzheimer's Cognitive Composite (PACC). A generalized linear mixed-effects model was used to examine group × time interactions for these measures. In addition, progression risks to mild cognitive impairment (MCI) or dementia were compared among the groups using Cox proportional hazards models. RESULTS A total of 367 participants (48 A + T + , 86 A + T - , 63 A - T + , and 170 A - T - ; mean age 73.8 years, mean follow-up 5.1 years, and 47.4% men) were included. For the lateral ventricle and PACC score, the A + T - and A + T + groups demonstrated statistically significantly greater volume expansion and cognitive decline over time than the A - T - group (lateral ventricle: β = 0.757 cm3/year [95% confidence interval 0.463 to 1.050], P < .001 for A + T - , and β = 0.889 cm3/year [0.523 to 1.255], P < .001 for A + T + ; PACC: β = - 0.19 /year [- 0.36 to - 0.02], P = .029 for A + T - , and β = - 0.59 /year [- 0.80 to - 0.37], P < .001 for A + T +). Notably, the A + T + group exhibited additional brain atrophy including the whole brain (β = - 2.782 cm3/year [- 4.060 to - 1.504], P < .001), hippocampus (β = - 0.057 cm3/year [- 0.085 to - 0.029], P < .001), and AD-signature regions (β = - 0.02 mm/year [- 0.03 to - 0.01], P < .001). Cox proportional hazards models suggested an increased risk of progressing to MCI or dementia in the A + T + group versus the A - T - group (adjusted hazard ratio = 3.35 [1.76 to 6.39]). CONCLUSIONS In cognitively normal individuals, A + T + compounds brain atrophy and cognitive deterioration, amplifying the likelihood of disease progression. Therapeutic interventions targeting A + T + individuals could be pivotal in curbing brain atrophy, cognitive decline, and disease progression.
Collapse
Affiliation(s)
- Motonobu Fujishima
- Department of Radiology, Kumagaya General Hospital, 4-5-1 Nakanishi, Kumagaya, 360-8567, Japan.
| | - Yohei Kawasaki
- Department of Biostatistics, Graduate School of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, 350-0495, Japan
- Biostatistics Section, Clinical Research Center, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikariga-Oka, Fukushima, 960-1295, Japan
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-61-2 Yatsuyamada, Koriyama, 963-8052, Japan
| |
Collapse
|
11
|
Salimi Y, Domingo-Fernández D, Hofmann-Apitius M, Birkenbihl C. Data-Driven Thresholding Statistically Biases ATN Profiling across Cohort Datasets. J Prev Alzheimers Dis 2024; 11:185-195. [PMID: 38230732 PMCID: PMC10995057 DOI: 10.14283/jpad.2023.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/02/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND While the amyloid/tau/neurodegeneration (ATN) framework has found wide application in Alzheimer's disease research, it is unclear if thresholds obtained using distinct thresholding methods are concordant within the same dataset and interchangeable across cohorts. OBJECTIVES To investigate the robustness of data-driven thresholding methods and ATN profiling across cohort datasets. DESIGN AND SETTING We evaluated the impact of thresholding methods on ATN profiles by applying five commonly-used methodologies across cohort datasets. We assessed the generalizability of disease patterns discovered within ATN profiles by clustering individuals from different cohorts who were assigned to the same ATN profile. PARTICIPANTS AND MEASUREMENTS Participants with available CSF amyloid-β 1-42, phosphorylated tau, and total tau measurements were included from eleven AD cohort studies. RESULTS We observed high variability among obtained ATN thresholds, both across methods and datasets that impacted the resulting profile assignments of participants significantly. Clustering participants from different cohorts within the same ATN category indicated that identified disease patterns were comparable across most cohorts and biases introduced through distinct thresholding and data representations remained insignificant in most ATN profiles. CONLUSION Thresholding method selection is a decision of statistical relevance that will inevitably bias the resulting profiling and affect its sensitivity and specificity. Thresholds are likely not directly interchangeable between independent cohorts. To apply the ATN framework as an actionable and robust profiling scheme, a comprehensive understanding of the impact of used thresholding methods, their statistical implications, and a validation of results is crucial.
Collapse
Affiliation(s)
- Y. Salimi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| | - D. Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
| | - M. Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
| | - C. Birkenbihl
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| | - the Alzheimer’s Disease Neuroimaging Initiative
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| | - the Japanese Alzheimer’s Disease Neuroimaging Initiative
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| | - the Alzheimer’s Disease Repository Without Borders Investigators
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| | - the European Prevention of Alzheimer’s Disease (EPAD) Consortium
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53757 Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115 Germany
- Schloß Birlinghoven, Sankt Augustin, 53757 Germany
| |
Collapse
|
12
|
Rani N, Alm KH, Corona-Long CA, Speck CL, Soldan A, Pettigrew C, Zhu Y, Albert M, Bakker A. Tau PET burden in Brodmann areas 35 and 36 is associated with individual differences in cognition in non-demented older adults. Front Aging Neurosci 2023; 15:1272946. [PMID: 38161595 PMCID: PMC10757623 DOI: 10.3389/fnagi.2023.1272946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The accumulation of neurofibrillary tau tangles, a neuropathological hallmark of Alzheimer's disease (AD), occurs in medial temporal lobe (MTL) regions early in the disease process, with some of the earliest deposits localized to subregions of the entorhinal cortex. Although functional specialization of entorhinal cortex subregions has been reported, few studies have considered functional associations with localized tau accumulation. Methods In this study, stepwise linear regressions were used to examine the contributions of regional tau burden in specific MTL subregions, as measured by 18F-MK6240 PET, to individual variability in cognition. Dependent measures of interest included the Clinical Dementia Rating Sum of Boxes (CDR-SB), Mini Mental State Examination (MMSE), and composite scores of delayed episodic memory and language. Other model variables included age, sex, education, APOE4 status, and global amyloid burden, indexed by 11C-PiB. Results Tau burden in right Brodmann area 35 (BA35), left and right Brodmann area 36 (BA36), and age each uniquely contributed to the proportion of explained variance in CDR-SB scores, while right BA36 and age were also significant predictors of MMSE scores, and right BA36 was significantly associated with delayed episodic memory performance. Tau burden in both left and right BA36, along with education, uniquely contributed to the proportion of explained variance in language composite scores. Importantly, the addition of more inclusive ROIs, encompassing less granular segmentation of the entorhinal cortex, did not significantly contribute to explained variance in cognition across any of the models. Discussion These findings suggest that the ability to quantify tau burden in more refined MTL subregions may better account for individual differences in cognition, which may improve the identification of non-demented older adults who are on a trajectory of decline due to AD.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kylie H. Alm
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caitlin A. Corona-Long
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caroline L. Speck
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Tosun D, Thropp P, Southekal S, Spottiswoode B, Fahmi R. Profiling and predicting distinct tau progression patterns: An unsupervised data-driven approach to flortaucipir positron emission tomography. Alzheimers Dement 2023; 19:5605-5619. [PMID: 37288753 PMCID: PMC10704003 DOI: 10.1002/alz.13164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 06/09/2023]
Abstract
INTRODUCTION How to detect patterns of greater tau burden and accumulation is still an open question. METHODS An unsupervised data-driven whole-brain pattern analysis of longitudinal tau positron emission tomography (PET) was used first to identify distinct tau accumulation profiles and then to build baseline models predictive of tau-accumulation type. RESULTS The data-driven analysis of longitudinal flortaucipir PET from studies done by the Alzheimer's Disease Neuroimaging Initiative, Avid Pharmaceuticals, and Harvard Aging Brain Study (N = 348 cognitively unimpaired, N = 188 mild cognitive impairment, N = 77 dementia), yielded three distinct flortaucipir-progression profiles: stable, moderate accumulator, and fast accumulator. Baseline flortaucipir levels, amyloid beta (Aβ) positivity, and clinical variables, identified moderate and fast accumulators with 81% and 95% positive predictive values, respectively. Screening for fast tau accumulation and Aβ positivity in early Alzheimer's disease, compared to Aβ positivity with variable tau progression profiles, required 46% to 77% lower sample size to achieve 80% power for 30% slowing of clinical decline. DISCUSSION Predicting tau progression with baseline imaging and clinical markers could allow screening of high-risk individuals most likely to benefit from a specific treatment regimen.
Collapse
Affiliation(s)
- Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA 94143
- Northern California Institute of Research and Education, San Francisco, CA, USA 94121
| | - Pamela Thropp
- Northern California Institute of Research and Education, San Francisco, CA, USA 94121
| | | | - Bruce Spottiswoode
- Siemens Medical Solutions USA, Inc., Molecular Imaging, Knoxville, TN, USA 37932
| | - Rachid Fahmi
- Siemens Medical Solutions USA, Inc., Molecular Imaging, Knoxville, TN, USA 37932
| |
Collapse
|
14
|
Besser LM, Chrisphonte S, Kleiman MJ, O’Shea D, Rosenfeld A, Tolea M, Galvin JE. The Healthy Brain Initiative (HBI): A prospective cohort study protocol. PLoS One 2023; 18:e0293634. [PMID: 37889891 PMCID: PMC10610524 DOI: 10.1371/journal.pone.0293634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The Health Brain Initiative (HBI), established by University of Miami's Comprehensive Center for Brain Health (CCBH), follows racially/ethnically diverse older adults without dementia living in South Florida. With dementia prevention and brain health promotion as an overarching goal, HBI will advance scientific knowledge by developing novel assessments and non-invasive biomarkers of Alzheimer's disease and related dementias (ADRD), examining additive effects of sociodemographic, lifestyle, neurological and biobehavioral measures, and employing innovative, methodologically advanced modeling methods to characterize ADRD risk and resilience factors and transition of brain aging. METHODS HBI is a longitudinal, observational cohort study that will follow 500 deeply-phenotyped participants annually to collect, analyze, and store clinical, cognitive, behavioral, functional, genetic, and neuroimaging data and biospecimens. Participants are ≥50 years old; have no, subjective, or mild cognitive impairment; have a study partner; and are eligible to undergo magnetic resonance imaging (MRI). Recruitment is community-based including advertisements, word-of-mouth, community events, and physician referrals. At baseline, following informed consent, participants complete detailed web-based surveys (e.g., demographics, health history, risk and resilience factors), followed by two half-day visits which include neurological exams, cognitive and functional assessments, an overnight sleep study, and biospecimen collection. Structural and functional MRI is completed by all participants and a subset also consent to amyloid PET imaging. Annual follow-up visits repeat the same data and biospecimen collection as baseline, except that MRIs are conducted every other year after baseline. ETHICS AND EXPECTED IMPACT HBI has been approved by the University of Miami Miller School of Medicine Institutional Review Board. Participants provide informed consent at baseline and are re-consented as needed with protocol changes. Data collected by HBI will lead to breakthroughs in developing new diagnostics and therapeutics, creating comprehensive diagnostic evaluations, and providing the evidence base for precision medicine approaches to dementia prevention with individualized treatment plans.
Collapse
Affiliation(s)
- Lilah M. Besser
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - Stephanie Chrisphonte
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - Michael J. Kleiman
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - Deirdre O’Shea
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - Amie Rosenfeld
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - Magdalena Tolea
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| | - James E. Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Boca Raton, Florida, United States of America
| |
Collapse
|
15
|
Petersen RC, Graf A, Brady C, De Santi S, Florian H, Landen J, Pontecorvo M, Randolph C, Sink KM, Carrillo MC, Weber CJ. Operationalizing selection criteria for clinical trials in Alzheimer's disease: Biomarker and clinical considerations. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12434. [PMID: 38023620 PMCID: PMC10655199 DOI: 10.1002/trc2.12434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Alzheimer's disease (AD) staging criteria lack standardized, empirical description. Well-defined AD staging criteria are an important consideration in protocol design, influencing a more standardized inclusion/exclusion criteria and defining what constitutes meaningful differentiation among the stages. However, many trials are being designed on the basis of biomarker features and the two need to be coordinated. The Alzheimer's Association Research Roundtable (AARR) Spring 2021 meeting discussed the implementation of preclinical AD staging criteria, and provided recommendations for how they may best be incorporated into clinical trials research. Discussion also included what currently available tools for global clinical trials may best define populations in preclinical AD trials, and if are we able to differentiate preclinical from clinical stages of the disease. Well-defined AD staging criteria are key to improving early detection, diagnostics, clinical trial enrollment, and identifying statistically significant clinical changes, and researchers discussed how emerging blood biomarkers may help with more efficient screening in preclinical stages.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | - Chris Brady
- WCG Clinical Endpoint Solutions, PrincetonNew JerseyUSA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Besser LM, Chrisphonte S, Kleiman MJ, O'Shea D, Rosenfeld A, Tolea M, Galvin JE. The Healthy Brain Initiative (HBI): A prospective cohort study protocol. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.21.23295908. [PMID: 37808766 PMCID: PMC10557773 DOI: 10.1101/2023.09.21.23295908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background The Health Brain Initiative (HBI), established by University of Miami's Comprehensive Center for Brain Health (CCBH), follows racially/ethnically diverse older adults without dementia living in South Florida. With dementia prevention and brain health promotion as an overarching goal, HBI will advance scientific knowledge by developing novel assessments and non-invasive biomarkers of Alzheimer's disease and related dementias (ADRD), examining additive effects of sociodemographic, lifestyle, neurological and biobehavioral measures, and employing innovative, methodologically advanced modeling methods to characterize ADRD risk and resilience factors and transition of brain aging. Methods HBI is a longitudinal, observational cohort study that will follow 500 deeply-phenotyped participants annually to collect, analyze, and store clinical, cognitive, behavioral, functional, genetic, and neuroimaging data and biospecimens. Participants are ≥50 years old; have no, subjective, or mild cognitive impairment; have a study partner; and are eligible to undergo magnetic resonance imaging (MRI). Recruitment is community-based including advertisements, word-of-mouth, community events, and physician referrals. At baseline, following informed consent, participants complete detailed web-based surveys (e.g., demographics, health history, risk and resilience factors), followed by two half-day visits which include neurological exams, cognitive and functional assessments, an overnight sleep study, and biospecimen collection. Structural and functional MRI is completed by all participants and a subset also consent to amyloid PET imaging. Annual follow-up visits repeat the same data and biospecimen collection as baseline, except that MRIs are conducted every other year after baseline. Ethics and expected impact HBI has been approved by the University of Miami Miller School of Medicine Institutional Review Board. Participants provide informed consent at baseline and are re-consented as needed with protocol changes. Data collected by HBI will lead to breakthroughs in developing new diagnostics and therapeutics, create comprehensive diagnostic evaluations, and provide the evidence base for precision medicine approaches to dementia prevention with individualized treatment plans.
Collapse
|
17
|
Erickson P, Simrén J, Brum WS, Ennis GE, Kollmorgen G, Suridjan I, Langhough R, Jonaitis EM, Van Hulle CA, Betthauser TJ, Carlsson CM, Asthana S, Ashton NJ, Johnson SC, Shaw LM, Blennow K, Andreasson U, Bendlin BB, Zetterberg H. Prevalence and Clinical Implications of a β-Amyloid-Negative, Tau-Positive Cerebrospinal Fluid Biomarker Profile in Alzheimer Disease. JAMA Neurol 2023; 80:2807607. [PMID: 37523162 PMCID: PMC10391361 DOI: 10.1001/jamaneurol.2023.2338] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/05/2023] [Indexed: 08/01/2023]
Abstract
Importance Knowledge is lacking on the prevalence and prognosis of individuals with a β-amyloid-negative, tau-positive (A-T+) cerebrospinal fluid (CSF) biomarker profile. Objective To estimate the prevalence of a CSF A-T+ biomarker profile and investigate its clinical implications. Design, Setting, and Participants This was a retrospective cohort study of the cross-sectional multicenter University of Gothenburg (UGOT) cohort (November 2019-January 2021), the longitudinal multicenter Alzheimer Disease Neuroimaging Initiative (ADNI) cohort (individuals with mild cognitive impairment [MCI] and no cognitive impairment; September 2005-May 2022), and 2 Wisconsin cohorts, Wisconsin Alzheimer Disease Research Center and Wisconsin Registry for Alzheimer Prevention (WISC; individuals without cognitive impairment; February 2007-November 2020). This was a multicenter study, with data collected from referral centers in clinical routine (UGOT) and research settings (ADNI and WISC). Eligible individuals had 1 lumbar puncture (all cohorts), 2 or more cognitive assessments (ADNI and WISC), and imaging (ADNI only) performed on 2 separate occasions. Data were analyzed on August 2022 to April 2023. Exposures Baseline CSF Aβ42/40 and phosphorylated tau (p-tau)181; cognitive tests (ADNI: modified preclinical Alzheimer cognitive composite [mPACC]; WISC: modified 3-test PACC [PACC-3]). Exposures in the ADNI cohort included [18F]-florbetapir amyloid positron emission tomography (PET), magnetic resonance imaging (MRI), [18F]-fluorodeoxyglucose PET (FDG-PET), and cross-sectional tau-PET (ADNI: [18F]-flortaucipir, WISC: [18F]-MK6240). Main Outcomes and Measures Primary outcomes were the prevalence of CSF AT biomarker profiles and continuous longitudinal global cognitive outcome and imaging biomarker trajectories in A-T+ vs A-T- groups. Secondary outcomes included cross-sectional tau-PET. Results A total of 7679 individuals (mean [SD] age, 71.0 [8.4] years; 4101 male [53%]) were included in the UGOT cohort, 970 individuals (mean [SD] age, 73 [7.0] years; 526 male [54%]) were included in the ADNI cohort, and 519 individuals (mean [SD] age, 60 [7.3] years; 346 female [67%]) were included in the WISC cohort. The prevalence of an A-T+ profile in the UGOT cohort was 4.1% (95% CI, 3.7%-4.6%), being less common than the other patterns. Longitudinally, no significant differences in rates of worsening were observed between A-T+ and A-T- profiles for cognition or imaging biomarkers. Cross-sectionally, A-T+ had similar tau-PET uptake to individuals with an A-T- biomarker profile. Conclusion and Relevance Results suggest that the CSF A-T+ biomarker profile was found in approximately 5% of lumbar punctures and was not associated with a higher rate of cognitive decline or biomarker signs of disease progression compared with biomarker-negative individuals.
Collapse
Affiliation(s)
- Pontus Erickson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Wagner S. Brum
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilda E. Ennis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | | | | | - Rebecca Langhough
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Erin M. Jonaitis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Carol A. Van Hulle
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Tobey J. Betthauser
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Cynthia M. Carlsson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King’s College London, London, England
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, England
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sterling C. Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulf Andreasson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara B. Bendlin
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Institute of Neurology, Department of Neurodegenerative Disease, University College London, London, England
- UK Dementia Research Institute, University College London, London, England
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
18
|
Yadollahikhales G, Rojas JC. Anti-Amyloid Immunotherapies for Alzheimer's Disease: A 2023 Clinical Update. Neurotherapeutics 2023; 20:914-931. [PMID: 37490245 PMCID: PMC10457266 DOI: 10.1007/s13311-023-01405-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
The amyloid cascade hypothesis is a useful framework for therapeutic development in Alzheimer's disease (AD). Amyloid b1-42 (Aβ) has been the main target of experimental therapies, based on evidence of the neurotoxic effects of Aβ, and of the potential adverse effects of brain Aβ burden detected in humans in vivo by positron emission tomography (PET). Progress on passive anti-amyloid immunotherapy research includes identification of antibodies that facilitate microglial activation, catalytical disaggregation, and increased flow of Aβ from cerebrospinal fluid (CSF) to plasma, thus decreasing the neurotoxic effects of Aβ. Recently completed phase 2 and 3 trials of 3rd generation anti-amyloid immunotherapies are supportive of their clinical efficacy in reducing brain Aβ burden and preventing cognitive decline. Data from recent trials implicate these agents as the first effective disease-modifying therapies against AD and has led to the US Food and Drug Administration (FDA) recent approval of aducanumab and lecanemab, under an accelerated approval pathway. The clinical effects of these agents are modest, however, and associated with amyloid-related imaging abnormalities (ARIA). Testing the effects of anti-Aβ immunotherapies in pre-symptomatic populations and identification of more potent and safer agents is the scope of ongoing and future research. Innovations in clinical trial design will be the key for the efficient and equitable development of novel anti-Aβ immunotherapies. The progress in the field of AD therapeutics will bring new clinical, logistical, and ethical challenges, which pose to revolutionize the practice of neurology, dementia care, and preventive cognitive healthcare.
Collapse
Affiliation(s)
- Golnaz Yadollahikhales
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, 1551 4th Street, 411G, San Francisco, CA, 94158, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, 1551 4th Street, 411G, San Francisco, CA, 94158, USA.
| |
Collapse
|
19
|
Rafii MS, Aisen PS. Detection and treatment of Alzheimer's disease in its preclinical stage. NATURE AGING 2023; 3:520-531. [PMID: 37202518 PMCID: PMC11110912 DOI: 10.1038/s43587-023-00410-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/29/2023] [Indexed: 05/20/2023]
Abstract
Longitudinal multimodal biomarker studies reveal that the continuum of Alzheimer's disease (AD) includes a long latent phase, referred to as preclinical AD, which precedes the onset of symptoms by decades. Treatment during the preclinical AD phase offers an optimal opportunity for slowing the progression of disease. However, trial design in this population is complex. In this Review, we discuss the recent advances in accurate plasma measurements, new recruitment approaches, sensitive cognitive instruments and self-reported outcomes that have facilitated the successful launch of multiple phase 3 trials for preclinical AD. The recent success of anti-amyloid immunotherapy trials in symptomatic AD has increased the enthusiasm for testing this strategy at the earliest feasible stage. We provide an outlook for standard screening of amyloid accumulation at the preclinical stage in clinically normal individuals, during which effective therapy to delay or prevent cognitive decline can be initiated.
Collapse
Affiliation(s)
- Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA.
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
20
|
Heinzinger N, Maass A, Berron D, Yakupov R, Peters O, Fiebach J, Villringer K, Preis L, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Bartels C, Jessen F, Maier F, Glanz W, Buerger K, Janowitz D, Perneczky R, Rauchmann BS, Teipel S, Killimann I, Göerß D, Laske C, Munk MH, Spottke A, Roy N, Heneka MT, Brosseron F, Dobisch L, Ewers M, Dechent P, Haynes JD, Scheffler K, Wolfsgruber S, Kleineidam L, Schmid M, Berger M, Düzel E, Ziegler G. Exploring the ATN classification system using brain morphology. Alzheimers Res Ther 2023; 15:50. [PMID: 36915139 PMCID: PMC10009950 DOI: 10.1186/s13195-023-01185-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 02/08/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND The NIA-AA proposed amyloid-tau-neurodegeneration (ATN) as a classification system for AD biomarkers. The amyloid cascade hypothesis (ACH) implies a sequence across ATN groups that patients might undergo during transition from healthy towards AD: A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+. Here we assess the evidence for monotonic brain volume decline for this particular (amyloid-conversion first, tau-conversion second, N-conversion last) and alternative progressions using voxel-based morphometry (VBM) in a large cross-sectional MRI cohort. METHODS We used baseline data of the DELCODE cohort of 437 subjects (127 controls, 168 SCD, 87 MCI, 55 AD patients) which underwent lumbar puncture, MRI scanning, and neuropsychological assessment. ATN classification was performed using CSF-Aβ42/Aβ40 (A+/-), CSF phospho-tau (T+/-), and adjusted hippocampal volume or CSF total-tau (N+/-). We compared voxel-wise model evidence for monotonic decline of gray matter volume across various sequences over ATN groups using the Bayesian Information Criterion (including also ROIs of Braak stages). First, face validity of the ACH transition sequence A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+ was compared against biologically less plausible (permuted) sequences among AD continuum ATN groups. Second, we evaluated evidence for 6 monotonic brain volume progressions from A-T-N- towards A+T+N+ including also non-AD continuum ATN groups. RESULTS The ACH-based progression A-T-N-➔A+T-N-➔A+T+N-➔A+T+N+ was consistent with cognitive decline and clinical diagnosis. Using hippocampal volume for operationalization of neurodegeneration (N), ACH was most evident in 9% of gray matter predominantly in the medial temporal lobe. Many cortical regions suggested alternative non-monotonic volume progressions over ACH progression groups, which is compatible with an early amyloid-related tissue expansion or sampling effects, e.g., due to brain reserve. Volume decline in 65% of gray matter was consistent with a progression where A status converts before T or N status (i.e., ACH/ANT) when compared to alternative sequences (TAN/TNA/NAT/NTA). Brain regions earlier affected by tau tangle deposition (Braak stage I-IV, MTL, limbic system) present stronger evidence for volume decline than late Braak stage ROIs (V/VI, cortical regions). Similar findings were observed when using CSF total-tau for N instead. CONCLUSION Using the ATN classification system, early amyloid status conversion (before tau and neurodegeneration) is associated with brain volume loss observed during AD progression. The ATN system and the ACH are compatible with monotonic progression of MTL atrophy. TRIAL REGISTRATION DRKS00007966, 04/05/2015, retrospectively registered.
Collapse
Affiliation(s)
- Nils Heinzinger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany. .,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - David Berron
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Jochen Fiebach
- Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
| | - Kersten Villringer
- Center for Stroke Research Berlin, Charité-Universitätsmedizin, Berlin, Germany
| | - Lukas Preis
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany.,Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany.,University of Edinburgh and UK DRI, Edinburgh, UK
| | - Eike Jacob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany.,Department of Medical Sciences, Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Franziska Maier
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Killimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Doreen Göerß
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurology, University of Bonn, Bonn, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neuroscience, Charité-Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Matthias Schmid
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute for Medical Biometry, University Hospital Bonn, Bonn, Germany
| | - Moritz Berger
- Institute for Medical Biometry, University Hospital Bonn, Bonn, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research (IKND), University Hospital Magdeburg, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | |
Collapse
|
21
|
Kasuga K, Tsukie T, Kikuchi M, Tokutake T, Washiyama K, Simizu S, Yoshizawa H, Kuroha Y, Yajima R, Mori H, Arakawa Y, Onda K, Miyashita A, Onodera O, Iwatsubo T, Ikeuchi T. The Clinical Application of Optimized AT(N) Classification in Alzheimer’s Clinical Syndrome (ACS) and non-ACS Conditions. Neurobiol Aging 2023; 127:23-32. [PMID: 37030016 DOI: 10.1016/j.neurobiolaging.2023.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
We aimed to assess the utility of AT(N) classification in clinical practice. We measured the cerebrospinal fluid levels of amyloid-β (Aβ) 42, Aβ40, phosphorylated tau, total tau, and neurofilament light chain (NfL) in samples from 230 patients with Alzheimer's clinical syndrome (ACS) and 328 patients with non-ACS. The concordance of two A-markers (i.e., Aβ42 alone and the Aβ42/Aβ40 ratio) was not significantly different between the ACS (87.4%) and non-ACS (74.1%) groups. However, the frequency of discordant cases with AAβ42-alone+/AAβ-ratio- was significantly higher in the non-ACS (23.8%) than in the ACS group (7.4%). The concordance of two N-markers (i.e., total tau and NfL) was 40.4% in the ACS group and 24.4% in the non-ACS group. In the ACS samples, the frequency of biological Alzheimer's disease (i.e., A+T+) in Ntau+ cases was 95% while that in NNfL+ cases was 65%. Reflecting Aβ deposition and neurodegeneration more accurately, we recommend the use of AT(N) classification defined by cerebrospinal fluid AAβ-ratioTNNfL in clinical practice.
Collapse
|
22
|
Cai Y, Du J, Li A, Zhu Y, Xu L, Sun K, Ma S, Guo T. Initial levels of β-amyloid and tau deposition have distinct effects on longitudinal tau accumulation in Alzheimer's disease. Alzheimers Res Ther 2023; 15:30. [PMID: 36750884 PMCID: PMC9903587 DOI: 10.1186/s13195-023-01178-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND To better assist with the design of future clinical trials for Alzheimer's disease (AD) and aid in our understanding of the disease's symptomatology, it is essential to clarify what roles β-amyloid (Aβ) plaques and tau tangles play in longitudinal tau accumulation inside and outside the medial temporal lobe (MTL) as well as how age, sex, apolipoprotein E (APOE) ε4 (APOE-ε4), and Klotho-VS heterozygosity (KL-VShet) modulate these relationships. METHODS We divided the 325 Aβ PET-positive (A+) participants into two groups, A+/T- (N = 143) and A+/T+ (N = 182), based on the threshold (1.25) of the temporal meta-ROI 18F-flortaucipir (FTP) standardized uptake value ratio (SUVR). We then compared the baseline and slopes of A+/T- and A+/T+ individuals' Aβ plaques and temporal meta-ROI tau tangles with those of A-/T- cognitively unimpaired individuals (N = 162) without neurodegeneration. In addition, we looked into how baseline Aβ and tau may predict longitudinal tau increases and how age, sex, APOE-ε4, and KL-VShet affect these associations. RESULTS In entorhinal, amygdala, and parahippocampal (early tau-deposited regions of temporal meta-ROI), we found that baseline Aβ and tau deposition were positively linked to more rapid tau increases in A+/T- participants. However, in A+/T+ individuals, the longitudinal tau accumulation in fusiform, inferior temporal, and middle temporal cortices (late tau-deposited regions of temporal meta-ROI) was primarily predicted by the level of tau tangles. Furthermore, compared to older participants (age ≥ 65), younger individuals (age < 65) exhibited faster Aβ-dependent but slower tau-related tau accumulations. Additionally, compared to the KL-VShet- group, KL-VShet+ individuals showed a significantly lower rate of tau accumulation associated with baseline entorhinal tau in fusiform and inferior temporal regions. CONCLUSION These findings offer novel perspectives to the design of AD clinical trials and aid in understanding the tau accumulation inside and outside MTL in AD. In particular, decreasing Aβ plaques might be adequate for A+/T- persons but may not be sufficient for A+/T+ individuals in preventing tau propagation and subsequent downstream pathological changes associated with tau.
Collapse
Affiliation(s)
- Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China
| | - Jing Du
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yalin Zhu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Linsen Xu
- Department of Medical Imaging, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518106, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
23
|
Stocks J, Heywood A, Popuri K, Beg MF, Rosen H, Wang L. Longitudinal Spatial Relationships Between Atrophy and Hypometabolism Across the Alzheimer's Disease Continuum. J Alzheimers Dis 2023; 92:513-527. [PMID: 36776061 DOI: 10.3233/jad-220975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND The A/T/N framework allows for the assessment of pathology-specific markers of MRI-derived structural atrophy and hypometabolism on 18FDG-PET. However, how these measures relate to each other locally and distantly across pathology-defined A/T/N groups is currently unclear. OBJECTIVE To determine the regions of association between atrophy and hypometabolism in A/T/N groups both within and across time points. METHODS We examined multivariate multimodal neuroimaging relationships between MRI and 18FDG-PET among suspected non-Alzheimer's disease pathology (SNAP) (A-T/N+; n = 14), Amyloid Only (A+T-N-; n = 24) and Probable AD (A+T+N+; n = 77) groups. Sparse canonical correlation analyses were employed to model spatially disjointed regions of association between MRI and 18FDG-PET data. These relationships were assessed at three combinations of time points -cross-sectionally, between baseline visits and between month 12 (M-12) follow-up visits, as well as longitudinally between baseline and M-12 follow-up. RESULTS In the SNAP group, spatially overlapping relationships between atrophy and hypometabolism were apparent in the bilateral temporal lobes when both modalities were assessed at the M-12 timepoint. Amyloid-Only subjects showed spatially discordant distributed atrophy-hypometabolism relationships at all time points assessed. In Probable AD subjects, local correlations were evident in the bilateral temporal lobes when both modalities were assessed at baseline and at M-12. Across groups, hypometabolism at baseline correlated with non-local, or distant, atrophy at M-12. CONCLUSION These results support the view that local concordance of atrophy and hypometabolism is the result of a tau-mediated process driving neurodegeneration.
Collapse
Affiliation(s)
- Jane Stocks
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ashley Heywood
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karteek Popuri
- School of Engineering Science, Simon Fraser University, Canada.,Memorial University of Newfoundland, Department of Computer Science, St. John's, NL, Canada
| | | | - Howie Rosen
- School of Medicine, University of California, San Francisco, CA, USA
| | - Lei Wang
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department of Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | |
Collapse
|
24
|
Sun Y, Moghekar A, Soldan A, Pettigrew C, Greenberg B, Albert M, Wang MC. Cerebrospinal Fluid Alzheimer's Disease Biomarker Patterns of Change Prior to the Onset of Mild Cognitive Impairment. J Alzheimers Dis 2023; 96:287-300. [PMID: 37742656 PMCID: PMC10793182 DOI: 10.3233/jad-230807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) are altered many years before the onset of clinical symptoms of mild cognitive impairment (MCI). Incorporating clinical symptom onset time into biomarker modeling may enhance our understanding of changes preceding MCI. OBJECTIVE Using a new analytical approach, we examined patterns of biomarker change prior to MCI symptom onset among individuals who progressed from normal cognition to MCI, stratified based on the age of symptom onset. We also analyzed biomarker patterns of change among participants who remained cognitively normal, and examined potential modifiers of biomarker trajectories, including demographics and apolipoprotein E (APOE) status. METHODS Analyses included 93 participants who progressed from normal cognition to MCI and 186 participants who remained cognitively normal, over an average follow-up period of 16.2 years. CSF biomarkers, including Aβ42, Aβ40, total tau (t-tau), and phosphorylated tau181 (p-tau181), were measured using the fully automated Lumipulse assays. RESULTS Among participants who progressed to MCI, Aβ42/Aβ40 decreased, and t-tau and p-tau181 increased. For participants who did not progress to MCI, CSF biomarkers showed relatively stable patterns. In both progressors and non-progressors, APOE4 carriers showed lower Aβ 42/Aβ40 levels (compared to non-carriers) at each point of the mean curves. Among non-progressors, APOE4 carriers had higher levels of p-tau181, p-tau181/(Aβ 42/Aβ40), and t-tau/(Aβ 42/Aβ 40). Additionally, among those who did not progress, female sex was associated with higher levels of t-tau, p-tau181, t-tau/(Aβ 42/Aβ 40), and p-tau181/(Aβ 42/Aβ 40). CONCLUSIONS These findings suggest that this analytic approach may provide additional insights into biomarker changes during early phases of AD.
Collapse
Affiliation(s)
- Yifei Sun
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barry Greenberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mei-Cheng Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | |
Collapse
|
25
|
Utz J, Olm P, Jablonowski J, Siegmann EM, Spitzer P, Lewczuk P, Kornhuber J, Maler JM, Oberstein TJ. Reconceptualization of the Erlangen Score for the Assessment of Dementia Risk: The ERlangen Score. J Alzheimers Dis 2023; 96:265-275. [PMID: 37742651 PMCID: PMC10657695 DOI: 10.3233/jad-230524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND The established Erlangen Score (ES) for the interpretation of cerebrospinal fluid (CSF) biomarkers in the diagnostics of Alzheimer's disease (AD) uses markers of amyloidopathy and tauopathy, equally weighted to form an easy-interpretable ordinal scale. However, these biomarkers are not equally predictive for AD. OBJECTIVE The higher weighting of the Aβ42/Aβ40 ratio, as a reconceptualized ERlangen Score (ERS), was tested for advantages in diagnostic performance. METHODS Non-demented subjects (N = 154) with a mean follow up of 5 years were assigned to a group ranging from 0 to 4 in ES or ERS. Psychometric trajectories and dementia risk were assessed. RESULTS The distribution of subjects between ES and ERS among the groups differed considerably, as grouping allocated 32 subjects to ES group 2, but only 2 to ERS group 2. The discriminative accuracy between the ES (AUC 73.2%, 95% CI [64.2, 82.2]) and ERS (AUC 72.0%, 95% CI [63.1, 81.0]) for dementia risk showed no significant difference. Without consideration of the Aβ42/Aβ40 ratio in ES grouping, the optimal cut-off of the ES shifted to ≥2. CONCLUSIONS The ERS showed advantages over the ES in test interpretation with comparable overall test performance, as fewer cases were allocated to the intermediate risk group. The established cut-off of ≥2 can be maintained for the ERS, whereas it must be adjusted for the ES when determining the Aβ42/Aβ40 ratio.
Collapse
Affiliation(s)
- Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Johannes Jablonowski
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Eva-Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, and Department of Biochemical Diagnostics, University Hospital of Bialystok, Białystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Bayern, Germany
| |
Collapse
|
26
|
He Y, Zhu X, Wang K, Xie J, Zhu Z, Ni M, Wang S, Xie Q. Design, synthesis, and preliminary evaluation of [ 18F]-aryl flurosulfates PET radiotracers via SuFEx methods for β-amyloid plaques in Alzheimer's disease. Bioorg Med Chem 2022; 75:117087. [PMID: 36356533 DOI: 10.1016/j.bmc.2022.117087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
Abstract
[18F]BAY-94-9172, [18F]AV-45, and [18F]GE-067 were FDA approved positron emission tomography (PET) imaging radiotracer of β-amyloid plaques (Aβ) in Alzheimer's disease (AD). However, the radiochemical synthesis requires multi-step reactions and complex procedure. Recently, a protocol for radiochemical synthesis of sulfur fluoride exchange (SuFEx) using ultrafast 19F/18F isotopic exchange had been reported. We developed three pairs of novel 18F-labeled radiotracers by the "SuFEx" method for PET imaging Aβ plaques. The 18F labeling reaction can be completed quickly (30 s) at room temperature and purified using solid-phase extraction (SPE). The radiochemical purity (RCP) of the products was all greater than 95 %. In vitro fluorescent staining using Aβ-transgenesis mice section preliminary verified the affinity of tracers with Aβ. Competitive binding assay displayed high affinity of tracers for towards artificial Aβ1-42 aggregates (Ki values ranging from 3.53 ± 0.39 to 42.0 ± 4.24 nM). In vivo biodistribution and Micro-PET imaging showed that [18F]-Sulfur Fluoride β-Amyloid ([18F]SFA 1-6) could penetration the blood-brain barrier (BBB) in wild-type mice, and [18F]SFA 5-6 had a high initial brain uptake value (3.65 ± 0.9 % and 5.07 ± 0.1 % ID/g, respectively) and a fast washout (Brain uptake2 min/60 min = 4.15 and 4.61, respectively) from the brain. In vitro autoradiography demonstrated the affinity of the [18F]SFA 5-6 to Aβ plaques in AD human brain tissues. Our results suggested that [18F]SFA maybe a potential PET radiotracers for detecting Aβ in Alzheimer's disease.
Collapse
Affiliation(s)
- Yunlin He
- School of Pharmacy, Bengbu Medical College, Anhui, Bengbu 233030, China; Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Xingxing Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Kaixuan Wang
- School of Pharmacy, Bengbu Medical College, Anhui, Bengbu 233030, China; Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Jikui Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Zehua Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Ming Ni
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Shicun Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China
| | - Qiang Xie
- School of Pharmacy, Bengbu Medical College, Anhui, Bengbu 233030, China; Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, Hefei 230001, China.
| |
Collapse
|
27
|
Greenberg BD, Pettigrew C, Soldan A, Wang J, Wang MC, Darrow JA, Albert MS, Moghekar A. CSF Alzheimer Disease Biomarkers: Time-Varying Relationships With MCI Symptom Onset and Associations With Age, Sex, and ApoE4. Neurology 2022; 99:e1640-e1650. [PMID: 36216518 PMCID: PMC9559947 DOI: 10.1212/wnl.0000000000200953] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/24/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES This study aimed to examine whether baseline CSF measures of Alzheimer disease (AD)-related pathology are associated with the time to onset of mild cognitive impairment (MCI) and whether these associations differ by age, sex, Apolipoprotein E (ApoE4) status, and proximal (≤7 years) vs distal (>7 years) time to symptom onset. METHODS Measures of amyloid (Aβ1-42 and Aβ1-40), phospho-tau (ptau181), and total tau (t-tau) were determined from CSF samples obtained at baseline from participants in an ongoing longitudinal project, known as the Biomarkers for Older Controls at Risk for Alzheimer Disease study (BIOCARD) study. The fully automated, Lumipulse G immunoassay was used to analyze the specimens. Cox regression models were used to examine the relationship of baseline biomarker levels with time to symptom onset of MCI and interactions with age, sex, and ApoE allelic status in subjects who progressed from normal cognition to MCI. RESULTS Analyses included 273 participants from the BIOCARD cohort, who were cognitively normal and predominantly middle-aged at baseline, and have been followed for an average of 16 years (max = 23.6). During follow-up, 94 progressed to MCI (median time to symptom onset = 6.9 years). In Cox regression models, elevated ptau181 and t-tau levels were associated with time to MCI symptom onset if it occurred within 7 years of baseline (HR 1.386 and 1.329; p = 0.009 and 0.017, respectively), while a lower Aβ42/Aβ40 ratio was associated with symptom onset if it occurred >7 years from baseline (HR 0.596, p = 0.003). There were also significant 3-way CSF × age × sex interactions for ptau181 and Aβ42/Aβ40, with follow-up analyses indicating that associations between these biomarkers and progression to MCI were stronger among men than among women, but this difference between sexes diminished with increasing age. DISCUSSION The lengthy follow-up of BIOCARD participants permitted an examination of time-varying associations between CSF AD biomarkers with MCI symptom onset and the influence of sex, baseline age, and ApoE4 genotype on these associations. These factors may inform clinical trial enrollment strategies, or trial duration and outcomes, which may use these measures as surrogate markers of treatment response.
Collapse
Affiliation(s)
- Barry D Greenberg
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Corinne Pettigrew
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Anja Soldan
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiangxia Wang
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jacqueline A Darrow
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Marilyn S Albert
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abhay Moghekar
- From the Department of Neurology (B.D.G., C.P., A.S., J.A.D., M.S.A., A.M.), Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
28
|
Baldeiras I, Silva-Spínola A, Lima M, Leitão MJ, Durães J, Vieira D, Tbuas-Pereira M, Cruz VT, Rocha R, Alves L, Machado Á, Milheiro M, Santiago B, Santana I. Alzheimer’s Disease Diagnosis Based on the Amyloid, Tau, and Neurodegeneration Scheme (ATN) in a Real-Life Multicenter Cohort of General Neurological Centers. J Alzheimers Dis 2022; 90:419-432. [DOI: 10.3233/jad-220587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The ATN scheme was proposed as an unbiased biological characterization of the Alzheimer’s disease (AD) spectrum, grouping biomarkers into three categories: brain Amyloidosis-A, Tauopathy-T, Neurodegeneration-N. Although this scheme was mainly recommended for research, it is relevant for diagnosis. Objective: To evaluate the ATN scheme performance in real-life cohorts reflecting the inflow of patients with cognitive complaints and different underlying disorders in general neurological centers. Methods: We included patients (n = 1,128) from six centers with their core cerebrospinal fluid-AD biomarkers analyzed centrally. A was assessed through Aβ 42/Aβ 40, T through pTau-181, and N through tTau. Association between demographic features, clinical diagnosis at baseline/follow-up and ATN profiles was assessed. Results: The prevalence of ATN categories was: A-T-N-: 28.3% ; AD continuum (A + T-/+N-/+): 47.8% ; non-AD (A- plus T or/and N+): 23.9% . ATN profiles prevalence was strongly influenced by age, showing differences according to gender, APOE genotype, and cognitive status. At baseline, 74.6% of patients classified as AD fell in the AD continuum, decreasing to 47.4% in mild cognitive impairment and 42.3% in other neurodegenerative conditions. At follow-up, 41% of patients changed diagnosis, and 92% of patients that changed to AD were classified within the AD continuum. A + was the best individual marker for predicting a final AD diagnosis, and the combinations A + T+(irrespective of N) and A + T+N+had the highest overall accuracy (83%). Conclusion: The ATN scheme is useful to guide AD diagnosis real-life neurological centers settings. However, it shows a lack of accuracy for patients with other types of dementia. In such cases, the inclusion of other markers specific for non-AD proteinopathies could be an important aid to the differential diagnosis.
Collapse
Affiliation(s)
- Inês Baldeiras
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Anuschka Silva-Spínola
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Marisa Lima
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Center for Research in Neuropsychology and Cognitive Behavioral Intervention, Faculty of Psychology and Educational Sciences, University of Coimbra, Coimbra, Portugal
| | - Maria João Leitão
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João Durães
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Daniela Vieira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Tbuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | | | - Raquel Rocha
- ULSM Unidade Local de Sáude de Matosinhos, Matosinhos, Portugal
| | - Luisa Alves
- Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | | | | | | | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
29
|
Kasuga K, Kikuchi M, Tsukie T, Suzuki K, Ihara R, Iwata A, Hara N, Miyashita A, Kuwano R, Iwatsubo T, Ikeuchi T. Different AT(N) profiles and clinical progression classified by two different N markers using total tau and neurofilament light chain in cerebrospinal fluid. BMJ Neurol Open 2022; 4:e000321. [PMID: 36046332 PMCID: PMC9379489 DOI: 10.1136/bmjno-2022-000321] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
Background The AT(N) classification was proposed for categorising individuals according to biomarkers. However, AT(N) profiles may vary depending on the markers chosen and the target population. Methods We stratified 177 individuals who participated in the Japanese Alzheimer's Disease Neuroimaging Initiative by AT(N) classification according to cerebrospinal fluid (CSF) biomarkers. We compared the frequency of AT(N) profiles between the classification using total tau and neurofilament light chain (NfL) as N markers (AT(N)tau and AT(N)NfL). Baseline characteristics, and longitudinal biological and clinical changes were examined between AT(N) profiles. Results We found that 9% of cognitively unimpaired subjects, 49% of subjects with mild cognitive impairment, and 61% of patients with Alzheimer's disease (AD) dementia had the biological AD profile (ie, A+T+) in the cohort. The frequency of AT(N) profiles substantially differed between the AT(N)tau and AT(N)NfL classifications. When we used t-tau as the N marker (AT(N)tau), those who had T- were more frequently assigned to (N)-, whereas those who had T+were more frequently assigned to (N)+ than when we used NfL as the N marker (AT(N)NfL). During a follow-up, the AD continuum group progressed clinically and biologically compared with the normal biomarker group in both the AT(N)tau and AT(N)NfL classifications. More frequent conversion to dementia was observed in the non-AD pathological change group in the AT(N)tau classification, but not in the AT(N)NfL classification. Conclusions AT(N)tau and AT(N)NfL in CSF may capture different aspects of neurodegeneration and provide a different prognostic value. The AT(N) classification aids in understanding the AD continuum biology in various populations.
Collapse
Affiliation(s)
- Kensaku Kasuga
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Masataka Kikuchi
- Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Tamao Tsukie
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Kazushi Suzuki
- Neurology, National Defense Medical College, Tokorozawa, Japan
| | - Ryoko Ihara
- Neurology, Tokyo Metropolitan Geriatric Medical Center Hospital, Tokyo, Japan
| | - Atsushi Iwata
- Neurology, Tokyo Metropolitan Geriatric Medical Center Hospital, Tokyo, Japan
| | - Norikazu Hara
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Akinori Miyashita
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | | | - Takeshi Iwatsubo
- Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Ikeuchi
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | | |
Collapse
|
30
|
Oberstein TJ, Schmidt MA, Florvaag A, Haas AL, Siegmann EM, Olm P, Utz J, Spitzer P, Doerfler A, Lewczuk P, Kornhuber J, Maler JM. Amyloid-β levels and cognitive trajectories in non-demented pTau181-positive subjects without amyloidopathy. Brain 2022; 145:4032-4041. [PMID: 35973034 DOI: 10.1093/brain/awac297] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 11/13/2022] Open
Abstract
Phosphorylated Tau181 (pTau181) in cerebrospinal fluid (CSF) and recently in plasma has been associated with Alzheimer's disease. In the absence of amyloidopathy, individuals with increased total Tau levels and/or temporal lobe atrophy experience no or only mild cognitive decline compared with biomarker-negative controls, leading to the proposal to categorize this constellation as Suspected non-Alzheimer disease pathophysiology (SNAP). We investigated whether the characteristics of SNAP also applied to individuals with increased CSF-pTau181 without amyloidopathy. In this long-term observational study, 285 non-demented individuals, including 76 individuals with subjective cognitive impairment and 209 individuals with mild cognitive impairment, were classified based on their CSF-levels of pTau181 (T), total Tau (N), Amyloid-beta-(Aβ)-42 and Aβ42/Aβ40 ratio (A) into A + T+N±, A + T-N±, A-T + N±, and A-T-N-. The longitudinal analysis included 154 subjects with a follow-up of more than 12 months who were followed to a median of 4.6 years (interquartile range = 4.3 years). We employed linear mixed models on psychometric tests and region of interest analysis of structural MRI data. Cognitive decline and hippocampal atrophy rate were significantly higher in A + T+N ± compared to A-T + N±, whereas there was no difference between A-T + N ± and A-T-N-. Furthermore, there was no significant difference between A-T + N ± and controls in dementia risk (Hazard ratio 0.3, 95% confidence interval [0.1, 1.9]). However, A-T + N ± and A-T-N- could be distinguished based on their Aβ42 and Aβ40 levels. Both Aβ40 and Aβ42 levels were significantly increased in A-T + N ± compared to controls. Long term follow-up of A-T + N ± individuals revealed no evidence that this biomarker constellation was associated with dementia or more severe hippocampal atrophy rates compared to controls. However, because of the positive association of pTau181 with Aβ in the A-T + N ± group, a link to the pathophysiology of Alzheimer´s disease cannot be excluded in this case. We propose to refer to these individuals in the SNAP group as "pTau and Aβ surge with subtle deterioration" (PASSED). The investigation of the circumstances of simultaneous elevation of pTau and Aβ might provide a deeper insight into the process under which Aβ becomes pathological.
Collapse
Affiliation(s)
- Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Manuel Alexander Schmidt
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Florvaag
- Department of Radiology and Nuclear Medicine, Klinikum Nuremberg, Nuremberg, Germany
| | - Anna Lena Haas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva Maria Siegmann
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pauline Olm
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Janine Utz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Doerfler
- Institute of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Neurodegeneration Diagnostics, Medical University of Bialystok, University Hospital of Bialystok, Bialystok, Poland.,Department of Biochemical Diagnostics, University Hospital of Bialystok, Bialystok, Poland
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
31
|
Pemberton HG, Collij LE, Heeman F, Bollack A, Shekari M, Salvadó G, Alves IL, Garcia DV, Battle M, Buckley C, Stephens AW, Bullich S, Garibotto V, Barkhof F, Gispert JD, Farrar G. Quantification of amyloid PET for future clinical use: a state-of-the-art review. Eur J Nucl Med Mol Imaging 2022; 49:3508-3528. [PMID: 35389071 PMCID: PMC9308604 DOI: 10.1007/s00259-022-05784-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Amyloid-β (Aβ) pathology is one of the earliest detectable brain changes in Alzheimer's disease (AD) pathogenesis. The overall load and spatial distribution of brain Aβ can be determined in vivo using positron emission tomography (PET), for which three fluorine-18 labelled radiotracers have been approved for clinical use. In clinical practice, trained readers will categorise scans as either Aβ positive or negative, based on visual inspection. Diagnostic decisions are often based on these reads and patient selection for clinical trials is increasingly guided by amyloid status. However, tracer deposition in the grey matter as a function of amyloid load is an inherently continuous process, which is not sufficiently appreciated through binary cut-offs alone. State-of-the-art methods for amyloid PET quantification can generate tracer-independent measures of Aβ burden. Recent research has shown the ability of these quantitative measures to highlight pathological changes at the earliest stages of the AD continuum and generate more sensitive thresholds, as well as improving diagnostic confidence around established binary cut-offs. With the recent FDA approval of aducanumab and more candidate drugs on the horizon, early identification of amyloid burden using quantitative measures is critical for enrolling appropriate subjects to help establish the optimal window for therapeutic intervention and secondary prevention. In addition, quantitative amyloid measurements are used for treatment response monitoring in clinical trials. In clinical settings, large multi-centre studies have shown that amyloid PET results change both diagnosis and patient management and that quantification can accurately predict rates of cognitive decline. Whether these changes in management reflect an improvement in clinical outcomes is yet to be determined and further validation work is required to establish the utility of quantification for supporting treatment endpoint decisions. In this state-of-the-art review, several tools and measures available for amyloid PET quantification are summarised and discussed. Use of these methods is growing both clinically and in the research domain. Concurrently, there is a duty of care to the wider dementia community to increase visibility and understanding of these methods.
Collapse
Affiliation(s)
- Hugh G Pemberton
- GE Healthcare, Amersham, UK.
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK.
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Fiona Heeman
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ariane Bollack
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - David Vallez Garcia
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mark Battle
- GE Healthcare, Amersham, UK
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | | | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- NIMTLab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
- UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | | |
Collapse
|
32
|
Lokhande VS, Ravi SN, Chakraborty R, Singh V. Equivariance Allows Handling Multiple Nuisance Variables When Analyzing Pooled Neuroimaging Datasets. PROCEEDINGS. IEEE COMPUTER SOCIETY CONFERENCE ON COMPUTER VISION AND PATTERN RECOGNITION 2022; 2022:10422-10431. [PMID: 36268536 PMCID: PMC9581465 DOI: 10.1109/cvpr52688.2022.01018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Pooling multiple neuroimaging datasets across institutions often enables improvements in statistical power when evaluating associations (e.g., between risk factors and disease outcomes) that may otherwise be too weak to detect. When there is only a single source of variability (e.g., different scanners), domain adaptation and matching the distributions of representations may suffice in many scenarios. But in the presence of more than one nuisance variable which concurrently influence the measurements, pooling datasets poses unique challenges, e.g., variations in the data can come from both the acquisition method as well as the demographics of participants (gender, age). Invariant representation learning, by itself, is ill-suited to fully model the data generation process. In this paper, we show how bringing recent results on equivariant representation learning (for studying symmetries in neural networks) instantiated on structured spaces together with simple use of classical results on causal inference provides an effective practical solution. In particular, we demonstrate how our model allows dealing with more than one nuisance variable under some assumptions and can enable analysis of pooled scientific datasets in scenarios that would otherwise entail removing a large portion of the samples. Our code is available on https://github.com/vsingh-group/DatasetPooling.
Collapse
|
33
|
Varesi A, Carrara A, Pires VG, Floris V, Pierella E, Savioli G, Prasad S, Esposito C, Ricevuti G, Chirumbolo S, Pascale A. Blood-Based Biomarkers for Alzheimer's Disease Diagnosis and Progression: An Overview. Cells 2022; 11:1367. [PMID: 35456047 PMCID: PMC9044750 DOI: 10.3390/cells11081367] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease characterized by amyloid-β (Aβ) plaque deposition and neurofibrillary tangle accumulation in the brain. Although several studies have been conducted to unravel the complex and interconnected pathophysiology of AD, clinical trial failure rates have been high, and no disease-modifying therapies are presently available. Fluid biomarker discovery for AD is a rapidly expanding field of research aimed at anticipating disease diagnosis and following disease progression over time. Currently, Aβ1-42, phosphorylated tau, and total tau levels in the cerebrospinal fluid are the best-studied fluid biomarkers for AD, but the need for novel, cheap, less-invasive, easily detectable, and more-accessible markers has recently led to the search for new blood-based molecules. However, despite considerable research activity, a comprehensive and up-to-date overview of the main blood-based biomarker candidates is still lacking. In this narrative review, we discuss the role of proteins, lipids, metabolites, oxidative-stress-related molecules, and cytokines as possible disease biomarkers. Furthermore, we highlight the potential of the emerging miRNAs and long non-coding RNAs (lncRNAs) as diagnostic tools, and we briefly present the role of vitamins and gut-microbiome-related molecules as novel candidates for AD detection and monitoring, thus offering new insights into the diagnosis and progression of this devastating disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
| | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Vitor Gomes Pires
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Valentina Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sakshi Prasad
- Faculty of Medicine, National Pirogov Memorial Medical University, 21018 Vinnytsya, Ukraine;
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy;
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
34
|
Li Q, Lv X, Jin F, Liao K, Gao L, Xu J. Associations of Polygenic Risk Score for Late-Onset Alzheimer's Disease With Biomarkers. Front Aging Neurosci 2022; 14:849443. [PMID: 35493930 PMCID: PMC9047857 DOI: 10.3389/fnagi.2022.849443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is a common irreversible neurodegenerative disease with heterogeneous genetic characteristics. Identifying the biological biomarkers with the potential to predict the conversion from normal controls to LOAD is clinically important for early interventions of LOAD and clinical treatment. The polygenic risk score for LOAD (AD-PRS) has been reported the potential possibility for reliably identifying individuals with risk of developing LOAD recently. To investigate the external phenotype changes resulting from LOAD and the underlying etiology, we summarize the comprehensive associations of AD-PRS with multiple biomarkers, including neuroimaging, cerebrospinal fluid and plasma biomarkers, cardiovascular risk factors, cognitive behavior, and mental health. This systematic review helps improve the understanding of the biomarkers with potential predictive value for LOAD and further optimizing the prediction and accurate treatment of LOAD.
Collapse
Affiliation(s)
- Qiaojun Li
- School of Information Engineering, Tianjin University of Commerce, Tianjin, China
| | - Xingping Lv
- School of Sciences, Tianjin University of Commerce, Tianjin, China
| | - Fei Jin
- Department of Molecular Imaging, Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Kun Liao
- School of Sciences, Tianjin University of Commerce, Tianjin, China
| | - Liyuan Gao
- School of Sciences, Tianjin University of Commerce, Tianjin, China
| | - Jiayuan Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
35
|
Ebenau JL, Pelkmans W, Verberk IMW, Verfaillie SCJ, van den Bosch KA, van Leeuwenstijn M, Collij LE, Scheltens P, Prins ND, Barkhof F, van Berckel BNM, Teunissen CE, van der Flier WM. Association of CSF, Plasma, and Imaging Markers of Neurodegeneration With Clinical Progression in People With Subjective Cognitive Decline. Neurology 2022; 98:e1315-e1326. [PMID: 35110378 PMCID: PMC8967429 DOI: 10.1212/wnl.0000000000200035] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Multiple biomarkers have been suggested to measure neurodegeneration (N) in the AT(N) framework, leading to inconsistencies between studies. We investigated the association of 5 N biomarkers with clinical progression and cognitive decline in individuals with subjective cognitive decline (SCD). METHODS We included individuals with SCD from the Amsterdam Dementia Cohort and SCIENCe project, a longitudinal cohort study (follow-up 4±3 years). We used the following N biomarkers: CSF total tau (t-tau), medial temporal atrophy visual rating on MRI, hippocampal volume (HV), serum neurofilament light (NfL), and serum glial fibrillary acidic protein (GFAP). We determined correlations between biomarkers. We assessed associations between N biomarkers and clinical progression to mild cognitive impairment or dementia (Cox regression) and Mini-Mental State Examination (MMSE) over time (linear mixed models). Models included age, sex, CSF β-amyloid (Aβ) (A), and CSF p-tau (T) as covariates, in addition to the N biomarker. RESULT We included 401 individuals (61±9 years, 42% female, MMSE 28 ± 2, vascular comorbidities 8%-19%). N biomarkers were modestly to moderately correlated (range r -0.28 - 0.58). Serum NfL and GFAP correlated most strongly (r 0.58, p < 0.01). T-tau was strongly correlated with p-tau (r 0.89, p < 0.01), although these biomarkers supposedly represent separate biomarker groups. All N biomarkers individually predicted clinical progression, but only HV, NfL, and GFAP added predictive value beyond Aβ and p-tau (hazard ratio 1.52 [95% CI 1.11-2.09]; 1.51 [1.05-2.17]; 1.50 [1.04-2.15]). T-tau, HV, and GFAP individually predicted MMSE slope (range β -0.17 to -0.11, p < 0.05), but only HV remained associated beyond Aβ and p-tau (β -0.13 [SE 0.04]; p < 0.05). DISCUSSION In cognitively unimpaired older adults, correlations between different N biomarkers were only moderate, indicating they reflect different aspects of neurodegeneration and should not be used interchangeably. T-tau was strongly associated with p-tau (T), which makes it less desirable to use as a measure for N. HV, NfL, and GFAP predicted clinical progression beyond A and T. Our results do not allow to choose one most suitable biomarker for N, but illustrate the added prognostic value of N beyond A and T. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that HV, NfL, and GFAP predicted clinical progression beyond A and T in individuals with SCD.
Collapse
Affiliation(s)
- Jarith L Ebenau
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK.
| | - Wiesje Pelkmans
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Inge M W Verberk
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Sander C J Verfaillie
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Karlijn A van den Bosch
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Mardou van Leeuwenstijn
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Lyduine E Collij
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Philip Scheltens
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Niels D Prins
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Frederik Barkhof
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Bart N M van Berckel
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Charlotte E Teunissen
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Wiesje M van der Flier
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| |
Collapse
|
36
|
Liu Y, Han PR, Hu H, Wang ZT, Guo Y, Ou YN, Cao XP, Tan L, Yu JT. A Multi-Dimensional Comparison of Alzheimer's Disease Neurodegenerative Biomarkers. J Alzheimers Dis 2022; 87:197-209. [PMID: 35275546 DOI: 10.3233/jad-215724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In the 2018 AT(N) framework, neurodegenerative (N) biomarkers plays an essential role in the research and staging of Alzheimer's disease (AD); however, the different choice of N may result in discordances. OBJECTIVE We aimed to compare different potential N biomarkers. METHODS We examined these N biomarkers among 1,238 participants from Alzheimer's Disease Neuroimaging Initiative (ADNI) in their 1) diagnostic utility, 2) cross-sectional and longitudinal correlations between different N biomarkers and clinical variables, and 3) the conversion risk of different N profiles. RESULTS Six neurodegenerative biomarkers changed significantly from preclinical AD, through prodromal AD to AD dementia stage, thus they were chosen as the candidate N biomarkers: hippocampal volume (HV), 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET), cerebrospinal fluid (CSF), total tau (T-tau), plasma neurofilament light chain (NFL), CSF NFL, and CSF neurogranin (Ng). Results indicated that FDG-PET not only had the greatest diagnostic utility in differentiating AD from controls (area under the curve: FDG-PET, 0.922), but also had the strongest association with cognitive scores. Furthermore, FDG-PET positive group showed the fastest memory decline (hazard ratio: FDG-PET, 3.45), which was also true even in the presence of amyloid-β pathology. Moreover, we observed great discordances between three valuable N biomarkers (FDG-PET, HV, and T-tau). CONCLUSION These results underline the importance of using FDG-PET as N in terms of cognitive decline and AD conversion, followed by HV, and could be a great complement to the AT(N) framework.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Pei-Ran Han
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | |
Collapse
|
37
|
Meyer PF, Ashton NJ, Karikari TK, Strikwerda-Brown C, Köbe T, Gonneaud J, Pichet Binette A, Ozlen H, Yakoub Y, Simrén J, Pannee J, Lantero-Rodriguez J, Labonté A, Baker SL, Schöll M, Vanmechelen E, Breitner JCS, Zetterberg H, Blennow K, Poirier J, Villeneuve S. Plasma p-tau231, p-tau181, PET biomarkers and cognitive change in older adults. Ann Neurol 2022; 91:548-560. [PMID: 35084051 DOI: 10.1002/ana.26308] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate novel plasma p-tau231, p-tau181 as well as Aβ40 and Aβ42 assays as indicators of tau and Aβ pathologies measured with positron emission tomography (PET), and their association with cognitive change, in cognitively unimpaired older adults. METHODS In a cohort of 244 older adults at risk of AD owing to a family history of AD dementia, we measured single molecule array (Simoa)-based plasma tau biomarkers (p-tau231, p-tau181), Aβ40 and Aβ42 with immunoprecipitation mass spectrometry, and Simoa NfL. A subset of 129 participants underwent amyloid-β (18 F-NAV4694) and tau (18 F-flortaucipir) PET assessments. We investigated plasma biomarker associations with Aβ and tau PET at the global and voxel level and tested plasma biomarker combinations for improved detection of Aβ-PET positivity. We also investigated associations with 8-year cognitive change. RESULTS Plasma p-tau biomarkers correlated with flortaucipir binding in medial temporal, parietal and inferior temporal regions. P-tau231 showed further associations in lateral parietal and occipital cortices. Plasma Aβ42/40 explained more variance in global Aβ-PET binding than Aβ42 alone. P-tau231 also showed strong and widespread associations with cortical Aβ-PET binding. Combining Aβ42/40 with p-tau231 or p-tau181 allowed for good distinction between Aβ-negative and -positive participants (AUC range 0.81-0.86). Individuals with low plasma Aβ42/40 and high p-tau experienced faster cognitive decline. INTERPRETATION Plasma p-tau231 showed more robust associations with PET biomarkers than p-tau181 in pre-symptomatic individuals. The combination of p-tau and Aβ42/40 biomarkers detected early AD pathology and cognitive decline. Such markers could be used as pre-screening tools to reduce the cost of prevention trials. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Pierre-François Meyer
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Cherie Strikwerda-Brown
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Theresa Köbe
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Julie Gonneaud
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Alexa Pichet Binette
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada.,McGill Centre for Integrative Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Hazal Ozlen
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Yara Yakoub
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Josef Pannee
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anne Labonté
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - John C S Breitner
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada.,McGill Centre for Integrative Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Judes Poirier
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Centre for Studies on Prevention of Alzheimer's Disease (StoP-AD), Montreal, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada.,McGill Centre for Integrative Neuroscience, McGill University, Montreal, Quebec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
38
|
van de Beek M, Ooms FAH, Ebenau JL, Barkhof F, Scheltens P, Teunissen CE, van Harten AC, van der Flier WM, Lemstra AW. Association of the ATN Research Framework With Clinical Profile, Ccognitive Decline, and Mortality in Patients With Dementia With Lewy Bodies. Neurology 2022; 98:e1262-e1272. [PMID: 35074893 DOI: 10.1212/wnl.0000000000200048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/30/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The ATN framework has been developed to categorize biological processes within the Alzheimer's disease (AD) continuum. Since AD pathology often coincides with dementia with Lewy Bodies (DLB), we aimed to investigate the distribution of ATN profiles in DLB and associate ATN-profiles in DLB to prognosis. METHODS We included 202 DLB patients from the Amsterdam Dementia Cohort (68±7yrs, 19%F, MMSE: 24±3, DAT-SPECT abnormal: 105/119). Patients were classified into eight profiles according to the ATN framework, using CSF Aβ42 (A), CSF p-tau (T) and medial temporal atrophy scores (N). We compared presence of clinical symptoms in ATN profiles and used linear mixed models to analyze decline on cognitive tests (follow-up 3±2yrs for n=139). Mortality risk was assessed using Cox proportional hazards analysis. Analyses were performed on both the eight profiles, as well as three clustered categories (normal AD biomarkers, non-AD pathologic change, AD continuum). RESULTS Fifty (25%) DLB patients had normal AD biomarkers (A-T-N-), 37 (18%) had non-AD pathologic change (A-T+N-: 10%/A-T-N+: 6%/A-T+N+: 3%) and 115 (57%) were classified within the AD continuum (A+T-N-: 20%/A+T+N-: 16%/A+T-N+: 10%/A+T+N+: 9%). A+T+N+ patients were older and least often had RBD symptoms. Parkinsonism was more often present in A+T-, compared to A-T+ (independent of N). Compared to patients with normal AD biomarkers, patients in A+ categories showed steeper decline on memory tests and higher mortality risk. Cognitive decline and mortality did not differ between non-AD pathologic change and normal AD biomarkers. DISCUSSION In our DLB cohort, we found clinically relevant associations between ATN categories and disease manifestation. Patients within the AD continuum had steeper cognitive decline and shorter survival. Implementing the ATN framework within DLB patients aids in subtyping patients based on underlying biological processes and could provide targets for future treatment strategies, e.g. AD modifying treatment. Expanding the framework by incorporating markers for alpha-synucleinopathy would improve the use of the framework to characterize dementia patients with mixed pathology, which could enhance proper stratification of patients for therapeutic trials.
Collapse
Affiliation(s)
- Marleen van de Beek
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Floor A H Ooms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jarith L Ebenau
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.,Institutes of Neurology and Healthcare Engineering, UCL, London, England, United Kingdom
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Neurochemistry, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Chan CK, Pettigrew C, Soldan A, Zhu Y, Wang MC, Albert M, Rosenberg PB. Association Between Late-Life Neuropsychiatric Symptoms and Cognitive Decline in Relation to White Matter Hyperintensities and Amyloid Burden. J Alzheimers Dis 2022; 86:1415-1426. [PMID: 35213370 PMCID: PMC9969328 DOI: 10.3233/jad-215267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Neuropsychiatric symptoms (NPS) among cognitively normal older adults are increasingly recognized as risk factors for cognitive decline and impairment. However, the underlying mechanisms remain unclear. OBJECTIVE To examine whether biomarkers of Alzheimer's disease (amyloid burden) and cerebrovascular disease (white matter hyperintensity (WMH) volume) modify the association between NPS and cognitive decline among cognitively unimpaired older adults. METHODS Analyses included 193 cognitively unimpaired participants (M age = 70 years) from the BIOCARD study, including 148 with PET amyloid and WMH biomarker data. NPS were measured with Neuropsychiatric Inventory and Geriatric Depression Scale scores. Linear mixed effects models were used to examine the association between baseline NPS and longitudinal cognitive trajectories (M follow-up = 3.05 years), using separate models for global, episodic memory, and executive function cognitive composite scores. In a subset of individuals with biomarker data, we evaluated whether WMH or cortical amyloid burden modified the relationship between NPS and cognitive change (as indicated by the NPS×biomarker×time interactions). RESULTS Higher baseline NPS were associated with lower executive function scores, but not a faster rate of decline in executive function. NPS symptoms were unrelated to the global or episodic memory composite scores, and there was little evidence of a relationship between NPS symptoms and cognitive change over time. The associations between NPS and cognitive decline did not differ by amyloid or WMH burden, and NPS were unrelated to amyloid and WMH burden. CONCLUSION These results suggest that the effect of neuropsychiatric symptoms on executive dysfunction may occur through mechanisms outside of amyloid and cerebrovascular disease.
Collapse
Affiliation(s)
- Carol K. Chan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Armstrong Institute for Patient Safety and Quality, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mei-Cheng Wang
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Paul B. Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
40
|
Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: the amyloid hypothesis revised. Nat Rev Neurosci 2022; 23:53-66. [PMID: 34815562 PMCID: PMC8840505 DOI: 10.1038/s41583-021-00533-w] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
The current conceptualization of Alzheimer disease (AD) is driven by the amyloid hypothesis, in which a deterministic chain of events leads from amyloid deposition and then tau deposition to neurodegeneration and progressive cognitive impairment. This model fits autosomal dominant AD but is less applicable to sporadic AD. Owing to emerging information regarding the complex biology of AD and the challenges of developing amyloid-targeting drugs, the amyloid hypothesis needs to be reconsidered. Here we propose a probabilistic model of AD in which three variants of AD (autosomal dominant AD, APOE ε4-related sporadic AD and APOE ε4-unrelated sporadic AD) feature decreasing penetrance and decreasing weight of the amyloid pathophysiological cascade, and increasing weight of stochastic factors (environmental exposures and lower-risk genes). Together, these variants account for a large share of the neuropathological and clinical variability observed in people with AD. The implementation of this model in research might lead to a better understanding of disease pathophysiology, a revision of the current clinical taxonomy and accelerated development of strategies to prevent and treat AD.
Collapse
Affiliation(s)
- Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland.
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, and Leuven Brain Institute, University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Cinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences; University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Life Science Partners, Amsterdam, Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer, IM2A, Groupe Hospitalier Pitié-Salpêtrière, Sorbonne Université, Paris, France
- Institut du Cerveau et de la Moelle Épinière, UMR-S975, INSERM, Paris, France
| |
Collapse
|
41
|
Guo T, Landau SM, Jagust WJ. Age, vascular disease, and Alzheimer's disease pathologies in amyloid negative elderly adults. ALZHEIMERS RESEARCH & THERAPY 2021; 13:174. [PMID: 34654465 PMCID: PMC8520216 DOI: 10.1186/s13195-021-00913-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/03/2021] [Indexed: 02/04/2023]
Abstract
Background We recently reported that CSF phosphorylated tau (p-Tau181) relative to Aβ40 (CSF p-Tau/Aβ40 ratio) was less noisy and increased associations with Alzheimer’s disease (AD) biomarkers compared to CSF p-Tau181 alone. While elevations of CSF p-Tau/Aβ40 can occur in amyloid-β (Aβ) negative (Aβ-) individuals, the factors associated with these elevations and their role in neurodegeneration and cognitive decline are unknown. We aim to explore factors associated with elevated tau in CSF, and how these elevated tau are related to neurodegeneration and cognitive decline in the absence of Aβ positivity. Methods We examined relationships between CSF p-Tau/Aβ40, and CSF Aβ42/Aβ40, Aβ PET, and white matter hyperintensities (WMH) as well as vascular risk factors in 149 cognitively unimpaired and 52 impaired individuals who were presumably not on the Alzheimer’s disease (AD) pathway due to negative Aβ status on both CSF and PET. Subgroups had 18F-fluorodeoxyglucose (FDG) PET and adjusted hippocampal volume (aHCV), and longitudinal measures of CSF, aHCV, FDG PET, and cognition data, so we examined CSF p-Tau/Aβ40 associations with these measures as well. Results Elevated CSF p-Tau/Aβ40 was associated with older age, male sex, greater WMH, and hypertension as well as a pattern of hippocampal atrophy and temporoparietal hypometabolism characteristic of AD. Lower CSF Aβ42/Aβ40, higher WMH, and hypertension but not age, sex, Aβ PET, APOE-ε4 status, body mass index, smoking, and hyperlipidemia at baseline predicted CSF p-Tau/Aβ40 increases over approximately 5 years of follow-up. The relationship between CSF p-Tau/Aβ40 and subsequent cognitive decline was partially or fully explained by neurodegenerative measurements. Conclusions These data provide surprising clues as to the etiology and significance of tau pathology in the absence of Aβ. It seems likely that, in addition to age, both cerebrovascular disease and subthreshold levels of Aβ are related to this tau accumulation. Crucially, this phenotype of CSF tau elevation in amyloid-negative individuals share features with AD such as a pattern of metabolic decline and regional brain atrophy. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00913-5.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | | |
Collapse
|
42
|
Yoon B, Guo T, Provost K, Korman D, Ward TJ, Landau SM, Jagust WJ. Abnormal tau in amyloid PET negative individuals. Neurobiol Aging 2021; 109:125-134. [PMID: 34715443 DOI: 10.1016/j.neurobiolaging.2021.09.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
We examined the characteristics of individuals with biomarker evidence of tauopathy but without β-amyloid (Aβ) (A-T+) in relation to individuals with (A+T+) and without (A-T-) evidence of Alzheimer's disease (AD). We included 561 participants with Aβ and tau PET from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We compared A-T- (n = 316), A-T+ (n = 63), and A+T+ (n = 182) individuals on demographics, amyloid, tau, hippocampal volumes, and cognition. A-T+ individuals were low on apolipoprotein E ɛ4 prevalence (17%) and had no evidence of subtly elevated brain Aβ within the negative range. The severity of tau deposition, hippocampal atrophy, and cognitive dysfunction in the A-T+ group was intermediate between A-T- and A+T+ (all p < 0.001). Tau uptake patterns in A-T+ individuals were heterogeneous, but approximately 29% showed tau deposition in the medial temporal lobe only, consistent with primary age-related tauopathy and an additional 32% showed a pattern consistent with AD. A-T+ individuals also share other features that are characteristic of AD such as cognitive impairment and neurodegeneration, but this group is heterogeneous and likely reflects more than one disorder.
Collapse
Affiliation(s)
- Bora Yoon
- Department of Neurology, Konyang University Hospital, Konyang University, College of Medicine, Daejeon, Korea.
| | - Tengfei Guo
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Karine Provost
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Deniz Korman
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Tyler J Ward
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
43
|
Wang YL, Chen J, Du ZL, Weng H, Zhang Y, Li R, Jia Z, Sun M, Jiang J, Wang FZ, Xu J. Plasma p-tau181 Level Predicts Neurodegeneration and Progression to Alzheimer's Dementia: A Longitudinal Study. Front Neurol 2021; 12:695696. [PMID: 34557143 PMCID: PMC8452983 DOI: 10.3389/fneur.2021.695696] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/09/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Plasma-based biomarkers would be potential biomarkers for early diagnosis of Alzheimer's disease (AD) because they are more available and cost-effective than cerebrospinal fluid (CSF) or neuroimaging. Therefore, we aimed to evaluate whether phosphorylated tau181 (p-tau181) in plasma could be an accurate AD predictor. Methods: Participants from the ADNI database included 185 cognitively unimpaired subjects with negative Aβ (CU–), 66 subjects with pre-clinical AD (CU with positive Aβ), 164 subjects with mild cognitive impairment with negative Aβ (MCI–), 254 subjects with prodromal AD (MCI with positive Aβ), and 98 subjects with dementia. Multiple linear regression models, linear mixed-effects models, and local regression were used to explore cross-sectional and longitudinal associations of plasma p-tau181 with cognition, neuroimaging, or CSF biomarkers adjusted for age, sex, education, and APOE genotype. Besides, Kaplan–Meier and adjusted Cox-regression model were performed to predict the risk of progression to dementia. Receiver operating characteristic analyses were performed to evaluate the predictive value of p-tau181. Results: Plasma p-tau181 level was highest in AD dementia, followed by prodromal AD and pre-clinical AD. In pre-clinical AD, plasma p-tau181 was negatively associated with hippocampal volume (β = −0.031, p-value = 0.017). In prodromal AD, plasma p-tau181 was associated with decreased global cognition, executive function, memory, language, and visuospatial functioning (β range −0.119 to −0.273, p-value < 0.05) and correlated with hippocampal volume (β = −0.028, p-value < 0.005) and white matter hyperintensity volume (WMH) volume (β = 0.02, p-value = 0.01). In AD dementia, increased plasma p-tau181 was associated with worse memory. In the whole group, baseline plasma p-tau181 was significantly associated with longitudinal increases in multiple neuropsychological test z-scores and correlated with AD-related CSF biomarkers and hippocampal volume (p-value < 0.05). Meanwhile, CU or MCI with high plasma p-tau181 carried a higher risk of progression to dementia. The area under the curve (AUC) of the adjusted model (age, sex, education, APOE genotype, and plasma p-tau181) was 0.78; that of additionally included CSF biomarkers was 0.84. Conclusions: Plasma p-tau181 level is related to multiple AD-associated cognitive domains and AD-related CSF biomarkers at the clinical stages of AD. Moreover, plasma p-tau181 level is related to the change rates of cognitive decline and hippocampal atrophy. Thus, this study confirms the utility of plasma p-tau181 as a non-invasive biomarker for early detection and prediction of AD.
Collapse
Affiliation(s)
- Yan-Li Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinglong Chen
- Department of Geriatric Medicine, China National Clinical Key Specialty, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhong-Li Du
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China.,Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Haoyi Weng
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China.,WeGene, Shenzhen Zaozhidao Technology Co. Ltd., Shenzhen, China.,Hunan Provincial Key Lab on Bioinformatics, School of Science and Engineering, Central South University, Shenzhen, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Runzhi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fang-Ze Wang
- Department of Cardiology, Weifang People's Hospital, Weifang, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
44
|
Bollinger RM, Keleman A, Thompson R, Westerhaus E, Fagan AM, Benzinger TL, Schindler SE, Xiong C, Balota D, Morris JC, Ances BM, Stark SL. Falls: a marker of preclinical Alzheimer disease: a cohort study protocol. BMJ Open 2021; 11:e050820. [PMID: 34526343 PMCID: PMC8444237 DOI: 10.1136/bmjopen-2021-050820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Progression to symptomatic Alzheimer disease (AD) occurs slowly over a series of preclinical stages. Declining functional mobility may be an early indicator of loss of brain network integration and may lead to an increased risk of experiencing falls. It is unknown whether measures of functional mobility and falls are preclinical markers of AD. The purpose of this study is to examine (1) the relationship between falls and functional mobility with AD biomarkers to determine when falls occur within the temporal progression to symptomatic Alzheimer disease, and (2) the attentional compared with perceptual/motor systems that underlie falls and functional mobility changes seen with AD. METHODS AND ANALYSIS This longitudinal cohort study will be conducted at the Knight Alzheimer Disease Research Center. Approximately 350 cognitively normal participants (with and without preclinical AD) will complete an in-home visit every year for 4 years. During each yearly assessment, functional mobility will be assessed using the Performance Oriented Mobility Assessment, Timed Up and Go, and Timed Up and Go dual task. Data regarding falls (including number and severity) will be collected monthly by self-report and confirmed through interviews. This study will leverage ongoing neuropsychological assessments and neuroimaging (including molecular imaging using positron emission tomography and MRI) performed by the Knight Alzheimer Disease Research Center. Relationships between falls and biomarkers of amyloid, tau and neurodegeneration will be evaluated. ETHICS AND DISSEMINATION This study was approved by the Washington University in St. Louis Institutional Review Board (reference number 201807135). Written informed consent will be obtained in the home prior to the collection of any study data. Results will be published in peer-reviewed publications and presented at national and international conferences. TRIAL REGISTRATION NUMBER NCT04949529; Pre-results.
Collapse
Affiliation(s)
- Rebecca M Bollinger
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Audrey Keleman
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Regina Thompson
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth Westerhaus
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tammie Ls Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David Balota
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, Missouri, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Susan L Stark
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
45
|
Ebenau JL, van der Lee SJ, Hulsman M, Tesi N, Jansen IE, Verberk IM, van Leeuwenstijn M, Teunissen CE, Barkhof F, Prins ND, Scheltens P, Holstege H, van Berckel BN, van der Flier WM. Risk of dementia in APOE ε4 carriers is mitigated by a polygenic risk score. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12229. [PMID: 34541285 PMCID: PMC8438688 DOI: 10.1002/dad2.12229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/09/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022]
Abstract
INTRODUCTION We investigated relationships among genetic determinants of Alzheimer's disease (AD), amyloid/tau/neurodegenaration (ATN) biomarkers, and risk of dementia. METHODS We studied cognitively normal individuals with subjective cognitive decline (SCD) from the Amsterdam Dementia Cohort and SCIENCe project. We examined associations between genetic variants and ATN biomarkers, and evaluated their predictive value for incident dementia. A polygenic risk score (PRS) was calculated based on 39 genetic variants. The APOE gene was not included in the PRS and was analyzed separately. RESULTS The PRS and APOE ε4 were associated with amyloid-positive ATN profiles, and APOE ε4 additionally with isolated increased tau (A-T+N-). A high PRS and APOE ε4 separately predicted AD dementia. Combined, a high PRS increased while a low PRS attenuated the risk associated with ε4 carriers. DISCUSSION Genetic variants beyond APOE are clinically relevant and contribute to the pathophysiology of AD. In the future, a PRS might be used in individualized risk profiling.
Collapse
Affiliation(s)
- Jarith L. Ebenau
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Sven J. van der Lee
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Clinical GeneticsAmsterdam UMCAmsterdamthe Netherlands
| | - Marc Hulsman
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Clinical GeneticsAmsterdam UMCAmsterdamthe Netherlands
- Delft Bioinformatics LabDelft University of TechnologyDelftthe Netherlands
| | - Niccolò Tesi
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Clinical GeneticsAmsterdam UMCAmsterdamthe Netherlands
- Delft Bioinformatics LabDelft University of TechnologyDelftthe Netherlands
| | - Iris E. Jansen
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Complex Trait GeneticsCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVU UniversityAmsterdamthe Netherlands
| | - Inge M.W. Verberk
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Neurochemistry LaboratoryDepartment of Clinical ChemistryVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Mardou van Leeuwenstijn
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Clinical ChemistryVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Niels D. Prins
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Philip Scheltens
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Henne Holstege
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Clinical GeneticsAmsterdam UMCAmsterdamthe Netherlands
- Delft Bioinformatics LabDelft University of TechnologyDelftthe Netherlands
| | - Bart N.M. van Berckel
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Radiology & Nuclear MedicineAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Epidemiology and BiostatisticsAmsterdam UMCAmsterdamthe Netherlands
| |
Collapse
|
46
|
Developing the ATX(N) classification for use across the Alzheimer disease continuum. Nat Rev Neurol 2021; 17:580-589. [PMID: 34239130 DOI: 10.1038/s41582-021-00520-w] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in the development of highly accurate fluid and neuroimaging biomarkers have catalysed the conceptual transformation of Alzheimer disease (AD) from the traditional clinical symptom-based definition to a clinical-biological construct along a temporal continuum. The AT(N) system is a symptom-agnostic classification scheme that categorizes individuals using biomarkers that chart core AD pathophysiological features, namely the amyloid-β (Aβ) pathway (A), tau-mediated pathophysiology (T) and neurodegeneration (N). This biomarker matrix is now expanding towards an ATX(N) system, where X represents novel candidate biomarkers for additional pathophysiological mechanisms such as neuroimmune dysregulation, synaptic dysfunction and blood-brain barrier alterations. In this Perspective, we describe the conceptual framework and clinical importance of the existing AT(N) system and the evolving ATX(N) system. We provide a state-of-the-art summary of the potential contexts of use of these systems in AD clinical trials and future clinical practice. We also discuss current challenges related to the validation, standardization and qualification process and provide an outlook on the real-world application of the AT(N) system.
Collapse
|
47
|
Rubega M, Formaggio E, Di Marco R, Bertuccelli M, Tortora S, Menegatti E, Cattelan M, Bonato P, Masiero S, Del Felice A. Cortical correlates in upright dynamic and static balance in the elderly. Sci Rep 2021; 11:14132. [PMID: 34238987 PMCID: PMC8266885 DOI: 10.1038/s41598-021-93556-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Falls are the second most frequent cause of injury in the elderly. Physiological processes associated with aging affect the elderly's ability to respond to unexpected balance perturbations, leading to increased fall risk. Every year, approximately 30% of adults, 65 years and older, experiences at least one fall. Investigating the neurophysiological mechanisms underlying the control of static and dynamic balance in the elderly is an emerging research area. The study aimed to identify cortical and muscular correlates during static and dynamic balance tests in a cohort of young and old healthy adults. We recorded cortical and muscular activity in nine elderly and eight younger healthy participants during an upright stance task in static and dynamic (core board) conditions. To simulate real-life dual-task postural control conditions, the second set of experiments incorporated an oddball visual task. We observed higher electroencephalographic (EEG) delta rhythm over the anterior cortex in the elderly and more diffused fast rhythms (i.e., alpha, beta, gamma) in younger participants during the static balance tests. When adding a visual oddball, the elderly displayed an increase in theta activation over the sensorimotor and occipital cortices. During the dynamic balance tests, the elderly showed the recruitment of sensorimotor areas and increased muscle activity level, suggesting a preferential motor strategy for postural control. This strategy was even more prominent during the oddball task. Younger participants showed reduced cortical and muscular activity compared to the elderly, with the noteworthy difference of a preferential activation of occipital areas that increased during the oddball task. These results support the hypothesis that different strategies are used by the elderly compared to younger adults during postural tasks, particularly when postural and cognitive tasks are combined. The knowledge gained in this study could inform the development of age-specific rehabilitative and assistive interventions.
Collapse
Affiliation(s)
- Maria Rubega
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy
| | - Emanuela Formaggio
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy
| | - Roberto Di Marco
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy
| | - Margherita Bertuccelli
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy
| | - Stefano Tortora
- Department of Information Engineering, University of Padua, Padova, Italy, 35131
| | - Emanuele Menegatti
- Department of Information Engineering, University of Padua, Padova, Italy, 35131
| | - Manuela Cattelan
- Department of Statistical Sciences, University of Padua, Padova, 35121, Italy
| | - Paolo Bonato
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Charlestown, Boston, MA, 02129, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Stefano Masiero
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy
- Padova Neuroscience Center, Padova, 35128, Italy
| | - Alessandra Del Felice
- Department of Neuroscience, Section of Rehabilitation, University of Padua, Padova, 35128, Italy.
- Padova Neuroscience Center, Padova, 35128, Italy.
| |
Collapse
|
48
|
Allison SL, Jonaitis EM, Koscik RL, Hermann BP, Mueller KD, Cary RP, Ma Y, Rowley HA, Carlsson CM, Asthana S, Zetterberg H, Blennow K, Bendlin BB, Johnson SC. Neurodegeneration, Alzheimer's disease biomarkers, and longitudinal verbal learning and memory performance in late middle age. Neurobiol Aging 2021; 102:151-160. [PMID: 33765428 PMCID: PMC8286465 DOI: 10.1016/j.neurobiolaging.2021.01.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 12/29/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022]
Abstract
This study examined the effect of neurodegeneration, and its interaction with Alzheimer's disease (AD) cerebrospinal fluid biomarkers, on longitudinal verbal learning and memory performance in cognitively unimpaired (CU) late middle-aged adults. Three hundred and forty-two CU adults (cognitive baseline mean age = 58.4), with cerebrospinal fluid and structural MRI, completed 2-10 (median = 5) cognitive assessments. Learning and memory were assessed using the Rey Auditory Verbal Learning Test (RAVLT). We used sequential comparison of nested linear mixed effects models to analyze the data. Model selection preserved a significant ptau181/Aβ42 × global atrophy × age interaction; individuals with less global atrophy and lower ptau181/Aβ42 levels had less learning and delayed recall decline than individuals with more global atrophy and/or higher levels of ptau181/Aβ42. The hippocampal volume × age × ptau181/Aβ42 interaction was not significant. Findings suggest that in a sample of CU late middle-aged adults, individuals with AD biomarkers, global atrophy, or both evidence greater verbal learning and memory decline than individuals without either risk factor.
Collapse
Affiliation(s)
- Samantha L Allison
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca L Koscik
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bruce P Hermann
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kimberly D Mueller
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin, Madison, WI, USA
| | - Robert P Cary
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Yue Ma
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Howard A Rowley
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M Carlsson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sanjay Asthana
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology, University College London, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara B Bendlin
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
49
|
Delmotte K, Schaeverbeke J, Poesen K, Vandenberghe R. Prognostic value of amyloid/tau/neurodegeneration (ATN) classification based on diagnostic cerebrospinal fluid samples for Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2021; 13:84. [PMID: 33879243 PMCID: PMC8059197 DOI: 10.1186/s13195-021-00817-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/23/2021] [Indexed: 11/12/2022]
Abstract
Objective The primary study objective of this retrospective academic memory clinic-based observational longitudinal study was to investigate the prognostic value of a cerebrospinal fluid (CSF)-based ATN classification for subsequent cognitive decline during the 3 years following lumbar puncture in a clinical, real-life setting. The secondary objective was to investigate the prognostic value of CSF biomarkers as continuous variables. Methods Data from 228 patients (median age 67 (47–85) years), who presented at the Neurology Memory Clinic UZ/KU Leuven between September 2011 and December 2016, were included with a follow-up period of up to 36 months. Patients underwent a CSF AD biomarker test for amyloid-beta 1–42 (Aβ42), hyperphosphorylated tau (p181-tau) and total tau (t-tau) in the clinical work-up for diagnostic reasons. Patients were divided into ATN classes based on CSF biomarkers: Aβ42 for amyloid (A), p181-tau for tau (T), and t-tau as a measure for neurodegeneration (N). Based on retrospective data analysis, cognitive performance was evaluated by Mini Mental State Examination (MMSE) scores every 6 months over a period up to 36 months following the lumbar puncture. The statistical analysis was based on linear mixed-effects modeling (LME). Results The distribution in the current clinical sample was as follows: A−/T−/N− 32.02%, A+/T−/N− 33.33%, A+/T+/N+ 17.11%, A+/T−/N+ 11.84%, A−/T−/N+ 4.39%, A−/T+/N+ 1.32% (3 cases), with no cases in the A−/T+/N− and A+/T+/N− class. Hence, the latter 3 classes were excluded from further analyses. The change of MMSE relative to A−/T−/N− over a 36-month period was significant in all four ATN classes: A+/T+/N+ = − 4.78 points on the MMSE; A−/T−/N+ = − 4.76; A+/T−/N+ = − 2.83; A+/T−/N− = − 1.96. The earliest significant difference was seen in the A+/T+/N+ class at 12 months after baseline. The effect of ATN class on future cognitive decline was confirmed for a different set of CSF thresholds. All individual baseline CSF biomarkers including the Aβ42/t-tau ratio showed a significant correlation with subsequent cognitive decline, with the highest correlation seen for Aβ42/t-tau. Conclusion ATN classification based on CSF biomarkers has a statistically significant and clinically relevant prognostic value for the course of cognitive decline in a 3-year period in a clinical practice setting. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00817-4.
Collapse
Affiliation(s)
- Koen Delmotte
- Department of Neurology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium. .,Department of Neurology, Jessa Hospital, Hasselt, Belgium.
| | - Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Laboratory of Neuropathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Koen Poesen
- Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.,Laboratory for Molecular Neurobiomarker Research, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium.,Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Guo T, Korman D, Baker SL, Landau SM, Jagust WJ. Longitudinal Cognitive and Biomarker Measurements Support a Unidirectional Pathway in Alzheimer's Disease Pathophysiology. Biol Psychiatry 2021; 89:786-794. [PMID: 32919611 PMCID: PMC9682985 DOI: 10.1016/j.biopsych.2020.06.029] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Amyloid-β (Aβ) likely plays a primary role in Alzheimer's disease pathogenesis, but longitudinal Aβ, tau, and neurodegeneration (A/T/N) measurements in the same individuals have rarely been examined to verify the temporal dynamics of these biomarkers. METHODS In this study, we investigated the temporal ordering of Aβ, tau, and neurodegeneration using longitudinal biomarkers in nondemented elderly individuals. A total of 395 cognitively unimpaired individuals and 204 individuals with mild cognitive impairment (320 [53%] were female) were classified into 8 A±/T±/N± categories according to the abnormal (+)/normal (-) status of Aβ (18F-florbetapir or 18F-florbetaben) positron emission tomography (PET), 18F-flortaucipir PET, and adjusted hippocampal volume (aHCV). Follow-up Aβ PET, tau PET, and aHCV measurements at 0.6 to 4.1 years were available for 35% to 63% of the sample. Baseline Aβ, tau, and aHCV were compared between different A/T/N profiles. We investigated the associations of baseline and longitudinal Aβ, tau, and neurodegeneration in relation to one another continuously. RESULTS Among T- participants, tau was higher for A+/T-/N- individuals compared with the A-/T-/N- group (p = .02). Among N- participants, neurodegeneration was worse among A+/T+/N- individuals compared with the A-/T-/N- group (p = .001). High baseline Aβ was associated (p < .001) with subsequent tau increase and high baseline tau was associated (p = .002) with subsequent aHCV decrease, whereas high tau and low aHCV at baseline were not associated with subsequent Aβ increase. CONCLUSIONS These findings define a sequence of pathological events in Alzheimer's disease that support a current model of Alzheimer's disease pathogenesis in which Aβ appears early, followed by deposition of abnormal tau aggregates and subsequent neurodegeneration.
Collapse
Affiliation(s)
- Tengfei Guo
- Helen Wills Neuroscience Institute, University of California, Berkeley, California; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California.
| | - Deniz Korman
- Helen Wills Neuroscience Institute, University of California, Berkeley, California
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, California; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, California; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California
| |
Collapse
|