1
|
Gentile JE, Heiss CN, Corridon TL, Mortberg MA, Fruhwürth S, Guzman K, Grötschel L, Montoliu-Gaya L, Chan K, Herring NC, Janicki T, Nhass R, Manavala Sarathy J, Erickson B, Kunz R, Erickson A, Braun C, Henry KT, Bry L, Arnold SE, Vallabh Minikel E, Zetterberg H, Vallabh SM. Evidence that minocycline treatment confounds the interpretation of neurofilament as a biomarker. Brain Commun 2025; 7:fcaf175. [PMID: 40417399 PMCID: PMC12100619 DOI: 10.1093/braincomms/fcaf175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/10/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Neurofilament light (NfL) concentration in CSF and blood serves as an important biomarker in neurology drug development. Changes in NfL are generally assumed to reflect changes in neuronal damage, while little is known about the clearance of NfL from biofluids. In a study of asymptomatic individuals at risk for prion disease, both blood and CSF NfL spiked in one participant following a 6-week course of minocycline, absent any other biomarker changes and without subsequent onset of symptoms. We subsequently observed high NfL after minocycline treatment in discarded clinical plasma samples from inpatients, in mouse plasma and in conditioned media from neuron-microglia co-cultures. The specificity and kinetics of NfL response lead us to hypothesize that minocycline does not cause or exacerbate neuronal damage, but instead affects NfL by inhibiting its clearance, posing a potential confounder for the interpretation of this important biomarker.
Collapse
Affiliation(s)
- Juliana E Gentile
- Program in Brain Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina N Heiss
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 80, Sweden
| | - Taylor L Corridon
- Program in Brain Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Meredith A Mortberg
- Program in Brain Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 80, Sweden
| | - Kenia Guzman
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lana Grötschel
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 80, Sweden
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 80, Sweden
| | - Kwan Chan
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Neil C Herring
- Clinical Microbiology Laboratory, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Timothy Janicki
- Clinical Microbiology Laboratory, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Rajaa Nhass
- Clinical Microbiology Laboratory, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Janani Manavala Sarathy
- Clinical Microbiology Laboratory, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | | | - Ryan Kunz
- IQ Proteomics, Framingham, MA 01702, USA
| | | | | | - Katherine T Henry
- In Vivo Drug Metabolism and Pharmacokinetics, Charles River Labs, Worcester, MA 01605, USA
| | - Lynn Bry
- Clinical Microbiology Laboratory, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Massachusetts Host-Microbiome Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Steven E Arnold
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Vallabh Minikel
- Program in Brain Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal 431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 80, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London NW1 3BT, UK
| | - Sonia M Vallabh
- Program in Brain Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
Duy Mac K, Su J. Optical biosensors for diagnosing neurodegenerative diseases. NPJ BIOSENSING 2025; 2:20. [PMID: 40322247 PMCID: PMC12048351 DOI: 10.1038/s44328-025-00040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025]
Abstract
Neurodegenerative diseases involve the progressive loss of neurons in the brain and nervous system, leading to functional decline. Early detection is critical for improving outcomes and advancing therapies. Optical biosensors, some of which offer rapid, label-free, and ultra-sensitive detection, have been applied to early diagnosis and drug screening. This review examines the principles and performance of different optical biosensors used for diagnosing neurodegenerative diseases and discusses potential future advancements.
Collapse
Affiliation(s)
- Khuong Duy Mac
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ USA
| | - Judith Su
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ USA
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ USA
| |
Collapse
|
3
|
Uzgiris AJ, Ladic LA, Pfister SX. Advances in neurofilament light chain analysis. Adv Clin Chem 2025; 126:31-71. [PMID: 40185536 DOI: 10.1016/bs.acc.2025.01.006] [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] [Indexed: 04/07/2025]
Abstract
This chapter provides a comprehensive summary of clinical laboratory testing for neurofilament light chain (NfL) in neurologic disease. A primer on the NfL structure and function is presented with its potential use as a biomarker. The most widely utilized methods for NfL in biologic samples are highlighted and examined. Limitations of current knowledge are considered, as are outstanding questions related to dissemination and standardization of testing. Herein we focus on methods available today and those in development for clinical use. In the final section, a broad vision is presented of how NfL may be utilized in the future to improve diagnosis and treatment of neurologic diseases as well as for maintaining health.
Collapse
Affiliation(s)
- Arejas J Uzgiris
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States.
| | - Lance A Ladic
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| | - Sophia X Pfister
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| |
Collapse
|
4
|
Xu B, Xiao K, Jia X, Cao R, Liang D, A R, Zhang W, Li C, Gao L, Chen C, Shi Q, Dong X. β-synuclein in cerebrospinal fluid as a potential biomarker for distinguishing human prion diseases from Alzheimer's and Parkinson's disease. Alzheimers Res Ther 2025; 17:39. [PMID: 39920821 PMCID: PMC11806663 DOI: 10.1186/s13195-025-01688-9] [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: 10/31/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND β-synuclein (β-syn), mainly expressed in central nerve system, is one of the biomarkers in cerebrospinal fluid (CSF) and blood for synaptic damage, which has been reported to be elevated in CSF and blood of the patients of prion diseases (PrDs). METHODS We analyzed 314 CSF samples from patients in China National Surveillance for CJD. The diagnostic groups of the 223 patients with PrDs included sporadic Creutzfeldt-Jacob disease (sCJD), genetic CJD (gCJD), fatal familial insomnia (FFI) and Gerstmann-Straussler-Scheinker (GSS). 91 patients with non-PrDs comprised Alzheimer's disease (AD), Parkinson's disease (PD), viral encephalitis (VE) or autoimmune encephalitis (AE) were enrolled in the control groups. The CSF β-syn levels were measured by a commercial microfluidic ELISA. The Mann-Whitney U test and Kruskal-Wallis H test were employed to analyze two or more sets of continuous variables. Multiple linear regression was also performed to evaluate the factors for CSF β-syn levels. Receiver operating characteristics (ROC) curves and area under the curve (AUC) values were used to assess the diagnostic performance of β-syn. RESULTS The median of β-syn levels (2074 pg/ml; IQR: 691 to 4332) of all PrDs was significantly higher than that of non-PrDs group (504 pg/ml; IQR: 126 to 3374). The CSF β-syn values in the cohorts of sCJD, T188K-gCJD, E200K-gCJD and P102L-GSS were remarkably higher than that of the group of AD + PD, but similar as that of the group of VE + AE. The elevated CSF β-syn in sCJD and gCJD cases was statistically associated with CSF 14-3-3 positive and appearance of mutism. ROC curve analysis identified satisfied performance for distinguishing from AD + PD, with high AUC values in sCJD (0.7640), T188K-gCJD (0.8489), E200K-gCJD (0.8548), P102L-GSS (0.7689) and D178N-FFI (0.7210), respectively. CONCLUSION Our data here indicate that CSF β-syn is a potential biomarker for distinguishing PrDs (gCJD, sCJD and GSS) from AD and PD, but is much less efficient from VE and AE. These findings have critical implications for early diagnosis and monitoring of synaptic integrity in prion diseases.
Collapse
Affiliation(s)
- Bing Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoxi Jia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rundong Cao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Donglin Liang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruhan A
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weiwei Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chunjie Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liping Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qi Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiaoping Dong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
5
|
Shimamura M, Weijie K, Nonaka T, Kosami K, Ae R, Fujita K, Matsubayashi T, Tsukamoto T, Sanjo N, Satoh K. Neurofilament Light Chain Levels in Serum and Cerebrospinal Fluid Do Not Correlate with Survival Times in Patients with Prion Disease. Biomolecules 2024; 15:8. [PMID: 39858404 PMCID: PMC11760854 DOI: 10.3390/biom15010008] [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: 11/06/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Prion diseases, including Creutzfeldt-Jakob disease (CJD), are deadly neurodegenerative disorders characterized by the buildup of abnormal prion proteins in the brain. This accumulation disrupts neuronal functions, leading to the rapid onset of psychiatric symptoms, ataxia, and cognitive decline. The urgency of timely diagnosis for effective treatment necessitates the identification of strongly correlated biomarkers in bodily fluids, which makes our research crucial. In this study, we employed a fully automated multiplex ELISA (Ella®) to measure the concentrations of 14-3-3 protein, total tau protein, and neurofilament light chain (NF-L) in cerebrospinal fluid (CSF) and serum samples from patients with prion disease and analyzed their link to disease prognosis. However, in North American and European cases, we did not confirm a correlation between NF-L levels and survival time. This discrepancy is believed to stem from differences in treatment policies and measurement methods between Japan and the United States. Nonetheless, our findings suggest that NF-L concentrations could be an early diagnostic marker for CJD patients with further enhancements. The potential impact of our findings on the early diagnosis of CJD patients is significant. Future research should focus on increasing the number of sCJD cases studied in Japan and gathering additional evidence using next-generation measurement techniques.
Collapse
Affiliation(s)
- Mika Shimamura
- Biomedical Research Support Center, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Kong Weijie
- Unit of Medical and Dental Sciences, Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Toshiaki Nonaka
- Division of Cellular and Molecular Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Koki Kosami
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi 329-0498, Japan; (K.K.); (R.A.)
| | - Ryusuke Ae
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi 329-0498, Japan; (K.K.); (R.A.)
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan;
| | - Taiki Matsubayashi
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, 1-5-45 Yushima Bunkyo-ku, Tokyo 113-8510, Japan;
| | - Tadashi Tsukamoto
- Department of Neurology, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan;
| | - Nobuo Sanjo
- Department of Internal Medicine, Division of Neurology, Kudanzaka Hospital, 1-6-12 Kudan-minami, Chiyoda-ku, Tokyo 102-0074, Japan;
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan
| | - Katsuya Satoh
- Unit of Medical and Dental Sciences, Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
- Unit of Brain Research Core Unit, Leading Medical Research Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| |
Collapse
|
6
|
Hermann P, Zerr I. Unmet needs of biochemical biomarkers for human prion diseases. Prion 2024; 18:89-93. [PMID: 38734978 PMCID: PMC11093021 DOI: 10.1080/19336896.2024.2349017] [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: 01/10/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Although the development of aggregation assays has noticeably improved the accuracy of the clinical diagnosis of prion diseases, research on biomarkers remains vital. The major challenges to overcome are non-invasive sampling and the exploration of new biomarkers that may predict the onset or reflect disease progression. This will become extremely important in the near future, when new therapeutics are clinically evaluated and eventually become available for treatment. This article aims to provide an overview of the achievements of biomarker research in human prion diseases, addresses unmet needs in the field, and points out future perspectives.
Collapse
Affiliation(s)
- Peter Hermann
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases, Göttingen, Germany
| |
Collapse
|
7
|
Corriveau-Lecavalier N, Piura YD, Appleby BS, Shir D, Barnard LR, Gogineni V, Jones DT, Day GS. FDG-PET patterns associate with survival in patients with prion disease. Ann Clin Transl Neurol 2024; 11:3227-3237. [PMID: 39470158 DOI: 10.1002/acn3.52230] [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: 07/12/2024] [Revised: 09/05/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVE Prion disease classically presents with rapidly progressive dementia, leading to death within months of diagnosis. Advances in diagnostic testing have improved recognition of patients with atypical presentations and protracted disease courses, raising key questions surrounding the relationship between patterns of neurodegeneration and survival. We assessed the contribution of fluorodeoxyglucose (FDG-PET) imaging for this purpose. METHODS FDG-PET were performed in 40 clinic patients with prion disease. FDG-PET images were projected onto latent factors generated in an external dataset to yield patient-specific eigenvalues. Eigenvalues were input into a clustering algorithm to generate data-driven clusters, which were compared by survival time. RESULTS Median age at FDG-PET was 65.3 years (range 23-85). Median time from FDG-PET to death was 3.7 months (range 0.3-19.0). Four data-driven clusters were generated, termed "Neocortical" (n = 7), "Transitional" (n = 12), "Temporo-parietal" (n = 13), and "Deep nuclei" (n = 6). Deep nuclei and transitional clusters had a shorter survival time than the neocortical cluster. Subsequent analyses suggested that this difference was driven by greater hypometabolism of deep nuclei relative to neocortical areas. FDG-PET-patterns were not associated with demographic (age and sex) or clinical (CSF total-tau, 14-3-3) variables. INTERPRETATION Greater hypometabolism within deep nuclei relative to neocortical areas associated with more rapid decline in patients with prion disease and vice versa. FDG-PET informs large-scale network physiology and may inform the relationship between spreading pathology and survival in patients with prion disease. Future studies should consider whether FDG-PET may enrich multimodal prion disease prognostication models.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yoav D Piura
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Brian S Appleby
- National Prion Disease Pathology Surveillance Center, Case Western Reserve, Cleveland, Ohio, USA
| | - Dror Shir
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, Florida, USA
| |
Collapse
|
8
|
Vallabh SM, Mortberg MA, Allen SW, Kupferschmid AC, Kivisakk P, Hammerschlag BL, Bolling A, Trombetta BA, Devitte-McKee K, Ford AM, Sather LE, Duffy G, Rivera A, Gerber J, McManus AJ, Minikel EV, Arnold SE. Fluid Biomarkers in Individuals at Risk for Genetic Prion Disease up to Disease Conversion. Neurology 2024; 103:e209506. [PMID: 38896810 PMCID: PMC11226308 DOI: 10.1212/wnl.0000000000209506] [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: 01/23/2024] [Accepted: 05/01/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVES To longitudinally characterize disease-relevant CSF and plasma biomarkers in individuals at risk for genetic prion disease up to disease conversion. METHODS This single-center longitudinal cohort study has followed known carriers of PRNP pathogenic variants at risk for prion disease, individuals with a close relative who died of genetic prion disease but who have not undergone predictive genetic testing, and controls. All participants were asymptomatic at first visit and returned roughly annually. We determined PRNP genotypes, measured NfL and GFAP in plasma, and RT-QuIC, total PrP, NfL, T-tau, and beta-synuclein in CSF. RESULTS Among 41 carriers and 21 controls enrolled, 28 (68%) and 15 (71%) were female, and mean ages were 47.5 and 46.1. At baseline, all individuals were asymptomatic. We observed RT-QuIC seeding activity in the CSF of 3 asymptomatic E200K carriers who subsequently converted to symptomatic and died of prion disease. 1 P102L carrier remained RT-QuIC negative through symptom conversion. No other individuals developed symptoms. The prodromal window from detection of RT-QuIC positivity to disease onset was 1 year long in an E200K individual homozygous (V/V) at PRNP codon 129 and 2.5 and 3.1 years in 2 codon 129 heterozygotes (M/V). Changes in neurodegenerative and neuroinflammatory markers were variably observed prior to onset, with increases observed for plasma NfL in 4/4 converters, and plasma GFAP, CSF NfL, CSF T-tau, and CSF beta-synuclein each in 2/4 converters, although values relative to age and fold changes relative to individual baseline were not remarkable for any of these markers. CSF PrP was longitudinally stable with mean coefficient of variation 9.0% across all individuals over up to 6 years, including data from converting individuals at RT-QuIC-positive timepoints. DISCUSSION CSF prion seeding activity may represent the earliest detectable prodromal sign in E200K carriers. Neuronal damage and neuroinflammation markers show limited sensitivity in the prodromal phase. CSF PrP levels remain stable even in the presence of RT-QuIC seeding activity. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT05124392 posted 2017-12-01, updated 2023-01-27.
Collapse
Affiliation(s)
- Sonia M Vallabh
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Meredith A Mortberg
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Shona W Allen
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Ashley C Kupferschmid
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Pia Kivisakk
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Bruno L Hammerschlag
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Anna Bolling
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Bianca A Trombetta
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Kelli Devitte-McKee
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Abaigeal M Ford
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Lauren E Sather
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Griffin Duffy
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Ashley Rivera
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Jessica Gerber
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Alison J McManus
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Eric V Minikel
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| | - Steven E Arnold
- From the McCance Center for Brain Health (S.M.V., M.A.M., E.V.M.) and Department of Neurology (S.M.V., M.A.M., S.W.A., A.C.K., P.K., B.L.H., A.B., B.A.T., K.D.-M., A.M.F., L.E.S., G.D., A.R., J.G., A.J.M., E.V.M., S.E.A.), Massachusetts General Hospital, Boston; Stanley Center for Psychiatric Research (S.M.V., M.A.M., E.V.M.), Broad Institute of MIT and Harvard, Cambridge; and Department of Neurology (S.M.V., P.K., E.V.M., S.E.A.), Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Brown Q, Nicholson E, Wang C, Greenlee J, Seger H, Veneziano S, Cassmann E. Temporal serum neurofilament light chain concentrations in sheep inoculated with the agent of classical scrapie. PLoS One 2024; 19:e0299038. [PMID: 38394122 PMCID: PMC10889644 DOI: 10.1371/journal.pone.0299038] [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: 09/27/2023] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Neurofilament light chain (Nf-L) has been used to detect neuroaxonal damage in the brain caused by physical injury or disease. The purpose of this study was to determine if serum Nf-L could be used as a biomarker for pre-symptomatic detection of scrapie in sheep. METHODS Four sheep with prion protein genotype AVQQ were intranasally inoculated with the classical scrapie strain x124. Blood was collected every 4 weeks until 44 weeks post-inoculation, at which point weekly collection commenced. Serum was analyzed using single molecule array (Quanterix SR-X) to evaluate Nf-L concentrations. RESULTS Scrapie was confirmed in each sheep by testing homogenized brainstem at the level of the obex with a commercially available enzyme immunoassay. Increased serum Nf-L concentrations were identified above the determined cutoff during the last tenth of the respective incubation period for each sheep. Throughout the time course study, PrPSc accumulation was not detected antemortem by immunohistochemistry in rectal tissue at any timepoint for any sheep. RT-QuIC results were inconsistently positive throughout the timepoints tested for each sheep; however, each sheep had at least one timepoint detected positive. When assessing serum Nf-L utility using receiver operator characteristic curves against different clinical parameters, such as asymptomatic and symptomatic (pruritus or neurologic signs), results showed that Nf-L was most useful at being an indicator of disease only late in disease progression when neurologic signs were present. CONCLUSION Serum Nf-L concentrations in the cohort of sheep increased as disease progressed; however, serum Nf-L did not increase during the presymptomatic window. The levels increased substantially throughout the final 10% of the animals' scrapie incubation period when other clinical signs were present. Serum Nf-L is not a reliable biomarker for pre-clinical detection of scrapie.
Collapse
Affiliation(s)
- Quazetta Brown
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Christensen, Ames, United States of America
| | - Eric Nicholson
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Justin Greenlee
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Hannah Seger
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Susan Veneziano
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Eric Cassmann
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| |
Collapse
|
10
|
Smith EN, Lee J, Prilutsky D, Zicha S, Wang Z, Han S, Zach N. Plasma neurofilament light levels show elevation two years prior to diagnosis of amyotrophic lateral sclerosis in the UK Biobank. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:170-176. [PMID: 38013452 DOI: 10.1080/21678421.2023.2285428] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disease with profound unmet need. In patients carrying genetic mutations, elevations in neurofilament light (NfL) have been shown to precede symptom onset, however, the natural history of NfL in general ALS patients is less characterized. METHODS We performed a secondary analysis of the UK Biobank Pharma Proteomics Project (UKB-PPP), a subset of the UK Biobank, a population-based cohort study in the United Kingdom, to examine plasma NfL levels in 237 participants subsequently diagnosed with ALS. We applied logistic and Cox proportional hazards regression to compare cases to 42,752 population-based and 948 age and sex-matched controls. Genetic information was obtained from exome and genotype array data.Results and Conclusions: We observed that NfL was 1.42-fold higher in cases vs population-based controls. At two to three years pre-diagnosis, NfL levels in patients exceeded the 95th percentile of age and sex-matched controls. A time-to-diagnosis analysis showed that a 2-fold increase in NfL levels was associated with a 3.4-fold risk of diagnosis per year, with NfL being most predictive of case status at two years (AUC = 0.96). Participants with genetic variation that might put them at risk for familial disease (N = 46) did not show a different pattern of association than those without (N = 191). DISCUSSION Our findings show that NfL is elevated and discriminative of future ALS diagnosis up to two years prior to diagnosis in patients with and without genetic risk variants.
Collapse
Affiliation(s)
- Erin N Smith
- Human Genetics and Systems Biology, Takeda Development Center Americas, Inc. San Diego, CA, USA
| | - Jonghun Lee
- Human Genetics and Systems Biology, Takeda Development Center Americas, Inc. Cambridge, MA, USA
| | - Daria Prilutsky
- Human Genetics and Systems Biology, Takeda Development Center Americas, Inc. Cambridge, MA, USA
| | - Stephen Zicha
- Neuroscience Translational Medicine, Takeda Development Center Americas, Inc. Cambridge, MA, USA, and
| | - Zemin Wang
- Neuroscience Translational Medicine, Takeda Development Center Americas, Inc. Cambridge, MA, USA, and
| | - Steve Han
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc. Cambridge, MA, USA
| | - Neta Zach
- Neuroscience Translational Medicine, Takeda Development Center Americas, Inc. Cambridge, MA, USA, and
| |
Collapse
|
11
|
Kishida H, Ueda N, Tanaka F. The advances in the early and accurate diagnosis of Creutzfeldt-Jakob disease and other prion diseases: where are we today? Expert Rev Neurother 2023; 23:803-817. [PMID: 37581576 DOI: 10.1080/14737175.2023.2246653] [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: 05/16/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
INTRODUCTION Before the introduction of MRI diffusion-weighted images (DWI), the diagnosis of Creutzfeldt-Jakob disease (CJD) relied upon nonspecific findings including clinical symptoms, EEG abnormalities, and elevated levels of cerebrospinal fluid 14-3-3 protein. Subsequently, the use of DWI has improved diagnostic accuracy, but it sometimes remains difficult to differentiate CJD from encephalitis, epilepsy, and other dementing disorders. The revised diagnostic criteria include real-time quaking-induced conversion (RT-QuIC), detecting small amounts of CJD-specific prion protein, and clinically sensitive DWI. Combining these techniques has further improved diagnostic accuracy, enabling earlier diagnosis. AREAS COVERED Herein, the authors review the recent advances in diagnostic methods and revised diagnostic criteria for sporadic CJD. They also discuss other prion diseases, such as variant CJD and chronic wasting disease, where the emergence of new types is a concern. EXPERT OPINION Despite improvements in diagnostic methods and criteria, some subtypes of prion disease are still difficult to diagnose, and even the diagnosis using the most innovative RT-QuIC test remains a challenge in terms of accuracy and standardization. However, these revised criteria can be adapted to the emergence of new types of prion diseases. It is essential to continue careful surveillance and update information on the latest prion disease phenotypes.
Collapse
Affiliation(s)
- Hitaru Kishida
- Department of Neurology, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Naohisa Ueda
- Department of Neurology, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
12
|
Mok TH, Nihat A, Majbour N, Sequeira D, Holm-Mercer L, Coysh T, Darwent L, Batchelor M, Groveman BR, Orr CD, Hughson AG, Heslegrave A, Laban R, Veleva E, Paterson RW, Keshavan A, Schott JM, Swift IJ, Heller C, Rohrer JD, Gerhard A, Butler C, Rowe JB, Masellis M, Chapman M, Lunn MP, Bieschke J, Jackson GS, Zetterberg H, Caughey B, Rudge P, Collinge J, Mead S. Seed amplification and neurodegeneration marker trajectories in individuals at risk of prion disease. Brain 2023; 146:2570-2583. [PMID: 36975162 PMCID: PMC10232278 DOI: 10.1093/brain/awad101] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Human prion diseases are remarkable for long incubation times followed typically by rapid clinical decline. Seed amplification assays and neurodegeneration biofluid biomarkers are remarkably useful in the clinical phase, but their potential to predict clinical onset in healthy people remains unclear. This is relevant not only to the design of preventive strategies in those at-risk of prion diseases, but more broadly, because prion-like mechanisms are thought to underpin many neurodegenerative disorders. Here, we report the accrual of a longitudinal biofluid resource in patients, controls and healthy people at risk of prion diseases, to which ultrasensitive techniques such as real-time quaking-induced conversion (RT-QuIC) and single molecule array (Simoa) digital immunoassays were applied for preclinical biomarker discovery. We studied 648 CSF and plasma samples, including 16 people who had samples taken when healthy but later developed inherited prion disease (IPD) ('converters'; range from 9.9 prior to, and 7.4 years after onset). Symptomatic IPD CSF samples were screened by RT-QuIC assay variations, before testing the entire collection of at-risk samples using the most sensitive assay. Glial fibrillary acidic protein (GFAP), neurofilament light (NfL), tau and UCH-L1 levels were measured in plasma and CSF. Second generation (IQ-CSF) RT-QuIC proved 100% sensitive and specific for sporadic Creutzfeldt-Jakob disease (CJD), iatrogenic and familial CJD phenotypes, and subsequently detected seeding activity in four presymptomatic CSF samples from three E200K carriers; one converted in under 2 months while two remain asymptomatic after at least 3 years' follow-up. A bespoke HuPrP P102L RT-QuIC showed partial sensitivity for P102L disease. No compatible RT-QuIC assay was discovered for classical 6-OPRI, A117V and D178N, and these at-risk samples tested negative with bank vole RT-QuIC. Plasma GFAP and NfL, and CSF NfL levels emerged as proximity markers of neurodegeneration in the typically slow IPDs (e.g. P102L), with significant differences in mean values segregating healthy control from IPD carriers (within 2 years to onset) and symptomatic IPD cohorts; plasma GFAP appears to change before NfL, and before clinical conversion. In conclusion, we show distinct biomarker trajectories in fast and slow IPDs. Specifically, we identify several years of presymptomatic seeding positivity in E200K, a new proximity marker (plasma GFAP) and sequential neurodegenerative marker evolution (plasma GFAP followed by NfL) in slow IPDs. We suggest a new preclinical staging system featuring clinical, seeding and neurodegeneration aspects, for validation with larger prion at-risk cohorts, and with potential application to other neurodegenerative proteopathies.
Collapse
Affiliation(s)
- Tze How Mok
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Akin Nihat
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Nour Majbour
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Danielle Sequeira
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Leah Holm-Mercer
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Thomas Coysh
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Lee Darwent
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Mark Batchelor
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Christina D Orr
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Andrew G Hughson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Rhiannon Laban
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Elena Veleva
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Ross W Paterson
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Ashvini Keshavan
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Jonathan M Schott
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Imogen J Swift
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Carolin Heller
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Jonathan D Rohrer
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester M13 9PL, UK
- Department of Geriatric Medicine, Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen, 45147 Essen, Germany
- Department of Nuclear Medicine, Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen, 45147 Essen, Germany
| | - Christopher Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge CB2 7EF, UK
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Miles Chapman
- Neuroimmunology and CSF Laboratory, University College London Hospitals NHS Trust National Hospital of Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Michael P Lunn
- Neuroimmunology and CSF Laboratory, University College London Hospitals NHS Trust National Hospital of Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Jan Bieschke
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Graham S Jackson
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- United Kingdom Dementia Research Institute at University College London, London WC1E 6BT, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, S-43180 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792-2420, USA
| | - Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Peter Rudge
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - John Collinge
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| | - Simon Mead
- Medical Research Council Prion Unit at University College London, UCL Institute of Prion Diseases, London W1W 7FF, UK
- NHS National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
13
|
Sun J, Shi Z, Wang L, Zhang X, Luo C, Hua J, Feng M, Chen Z, Wang M, Xu C. Construction of a microcavity-based microfluidic chip with simultaneous SERS quantification of dual biomarkers for early diagnosis of Alzheimer's disease. Talanta 2023; 261:124677. [PMID: 37201340 DOI: 10.1016/j.talanta.2023.124677] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 05/20/2023]
Abstract
Since there is no effective Alzheimer's disease (AD)-modifying therapy available currently, early analysis of AD core biomarkers has become one of great significance and common concern in clinical diagnosis. Herein, we designed an Au-plasmonic shell attached polystyrene (PS) microsphere in a microfluidic chip for simultaneous detection of Aβ1-42 and p-Tau181 protein. The corresponding Raman reporters were identified in femto gram level by ultrasensitive surface enhanced Raman spectroscopy (SERS). Both of Raman experimental data and finite-difference time-domain modeling demonstrates the synergetic coupling between PS microcavity with the optical confinement property and the localized surface plasmon resonance (LSPR) of AuNPs, so leading to highly amplified electromagnetic fields at the 'hot spot'. Moreover, the microfluidic system is designed with multiplex testing and control channels in which the AD-related dual proteins were detected quantitatively with a lower limit of 100 fg mL-1. Thus, the proposed microcavity-based SERS strategy initiates a new way for accurately prediction of AD in human blood samples and provides the potential application for synchronous determination of multiple analytes in general disease assays.
Collapse
Affiliation(s)
- Jianli Sun
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Zengliang Shi
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Li Wang
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Xinyi Zhang
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Chunshan Luo
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Jianyu Hua
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Muyu Feng
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China.
| | - Mingliang Wang
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China.
| | - Chunxiang Xu
- State Key Laboratory of Bioelectronics, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
14
|
Advances in Molecular Psychiatry - March 2023: mitochondrial function, stress, neuroinflammation - bipolar disorder, psychosis, and Alzheimer's disease. Mol Psychiatry 2023; 28:968-971. [PMID: 36899214 DOI: 10.1038/s41380-023-01968-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 03/12/2023]
|
15
|
Vallabh SM, Zou D, Pitstick R, O’Moore J, Peters J, Silvius D, Kriz J, Jackson WS, Carlson GA, Minikel EV, Cabin DE. Therapeutic Trial of anle138b in Mouse Models of Genetic Prion Disease. J Virol 2023; 97:e0167222. [PMID: 36651748 PMCID: PMC9973041 DOI: 10.1128/jvi.01672-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Phenotypic screening has yielded small-molecule inhibitors of prion replication that are effective in vivo against certain prion strains but not others. Here, we sought to test the small molecule anle138b in multiple mouse models of prion disease. In mice inoculated with the RML strain of prions, anle138b doubled survival and durably suppressed astrogliosis measured by live-animal bioluminescence imaging. In knock-in mouse models of the D178N and E200K mutations that cause genetic prion disease, however, we were unable to identify a clear, quantifiable disease endpoint against which to measure therapeutic efficacy. Among untreated animals, the mutations did not impact overall survival, and bioluminescence remained low out to >20 months of age. Vacuolization and PrP deposition were observed in some brain regions in a subset of mutant animals but appeared to be unable to carry the weight of a primary endpoint in a therapeutic study. We conclude that not all animal models of prion disease are suited to well-powered therapeutic efficacy studies, and care should be taken in choosing the models that will support drug development programs. IMPORTANCE There is an urgent need to develop drugs for prion disease, a currently untreatable neurodegenerative disease. In this effort, there is a debate over which animal models can best support a drug development program. While the study of prion disease benefits from excellent animal models because prions naturally afflict many different mammals, different models have different capabilities and limitations. Here, we conducted a therapeutic efficacy study of the drug candidate anle138b in mouse models with two of the most common mutations that cause genetic prion disease. In a more typical model where prions are injected directly into the brain, we found anle138b to be effective. In the genetic models, however, the animals never reached a clear, measurable point of disease onset. We conclude that not all prion disease animal models are ideally suited to drug efficacy studies, and well-defined, quantitative disease metrics should be a priority.
Collapse
Affiliation(s)
- Sonia M. Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Prion Alliance, Cambridge, Massachusetts, USA
| | - Dan Zou
- Montana Veterinary Diagnostic Laboratory, Bozeman, Montana, USA
| | - Rose Pitstick
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Jill O’Moore
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Janet Peters
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Derek Silvius
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Jasna Kriz
- Cervo Brain Research Center, Université Laval, Québec, Québec, Canada
| | - Walker S. Jackson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - George A. Carlson
- Institute for Neurodegenerative Diseases, University of California—San Francisco, San Francisco, California, USA
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Prion Alliance, Cambridge, Massachusetts, USA
| | | |
Collapse
|
16
|
Appleby BS, Shetty S, Elkasaby M. Genetic aspects of human prion diseases. Front Neurol 2022; 13:1003056. [PMID: 36277922 PMCID: PMC9579322 DOI: 10.3389/fneur.2022.1003056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Human prion diseases are rapidly progressive and fatal neurodegenerative conditions caused by a disease-causing isoform of the native prion protein. The prion protein gene (PRNP) encodes for the cellular prion protein, which is the biological substrate for prion disease transmission and neurotoxicity. Human prion diseases have three etiologies: sporadic, genetic, and acquired. PRNP polymorphisms and pathogenic variants play a large role in the frequency, age at onset, and clinicopathologic phenotype of prion diseases. Genetic prion diseases will be covered in detail and information necessary for clinical care, predictive genetic testing, and genetic counseling will be reviewed. Because the prion protein is necessary for transmission and neurotoxicity, many experimental treatments targeting its production are being investigated and hold potential promise as a disease modifying treatment for all forms of prion disease, including asymptomatic mutation carriers. This article will review genetic aspects of human prion disease and their influence on epidemiology, clinicopathologic phenotype, diagnostics, clinical management, and potential treatment approaches.
Collapse
Affiliation(s)
- Brian S. Appleby
- Department of Pathology, National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Neurology, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, United States
- *Correspondence: Brian S. Appleby
| | - Shashirekha Shetty
- Department of Pathology, National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Pathology, Center for Human Genetics Laboratory, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, United States
| | - Mohamed Elkasaby
- Department of Neurology, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
17
|
Halbgebauer S, Steinacker P, Hengge S, Oeckl P, Abu Rumeileh S, Anderl-Straub S, Lombardi J, Von Arnim CAF, Giese A, Ludolph AC, Otto M. CSF levels of SNAP-25 are increased early in Creutzfeldt-Jakob and Alzheimer's disease. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328646. [PMID: 35995553 DOI: 10.1136/jnnp-2021-328646] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Synaptosomal-associated protein 25 (SNAP-25) in cerebrospinal fluid (CSF) is an emerging synaptic biomarker for the early diagnosis of Alzheimer's disease (AD). However, comprehensive studies investigating the marker in Creutzfeldt-Jakob disease (CJD) and in the differential diagnosis of neurodegenerative diseases are still lacking. METHODS We developed a novel, sensitive ELISA for the measurement of SNAP-25 in CSF. In total, we analysed 316 patients from 6 diagnostic groups comprising patients with AD (n=96), CJD (n=55), Parkinson's disease spectrum (n=41), frontotemporal lobar degeneration (n=25) and amyotrophic lateral sclerosis (n=24) and non-neurodegenerative control patients (n=75). Using receiver operating characteristic curve analysis, we analysed the differential diagnostic potential and compared the results with core AD biomarkers. RESULTS SNAP-25 CSF concentrations were elevated in AD and CJD (p<0.0001) but not in the other neurodegenerative diseases. Increased levels were observed already at early AD and CJD stages (p<0.0001). In CJD, SNAP-25 levels correlated negatively with survival time (r=-0.33 (95% CI -0.57 to -0.04, p=0.02). For the discrimination of AD from all other diseases except CJD, we observed a good diagnostic performance for CSF SNAP-25 (area under the curve (AUC) 0.85) which was further improved by applying the ratio with CSF amyloid-β 1-42 (AUC 0.95). For CJD, we could demonstrate a strong differential diagnostic potential against all other groups including AD (AUC 0.97). CONCLUSION Using the novel established CSF SNAP-25 ELISA, we here demonstrate the applicability of SNAP-25 as an early synaptic biomarker for both AD and CJD with a possible prognostic value in patients with CJD.
Collapse
Affiliation(s)
| | - Petra Steinacker
- Department of Neurology, University of Ulm, Ulm, Germany
- Neurology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Sophie Hengge
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases, Ulm, Germany
| | - Samir Abu Rumeileh
- Department of Neurology, University of Ulm, Ulm, Germany
- Neurology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | | | | | - Christine A F Von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Geriatrics, University Medical Center Göttingen, Goettingen, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Albert C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases, Ulm, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
- Neurology, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| |
Collapse
|
18
|
Shir D, Lazar EB, Graff-Radford J, Aksamit AJ, Cutsforth-Gregory JK, Jones DT, Botha H, Ramanan VK, Prusinski C, Porter A, Day GS. Analysis of Clinical Features, Diagnostic Tests, and Biomarkers in Patients With Suspected Creutzfeldt-Jakob Disease, 2014-2021. JAMA Netw Open 2022; 5:e2225098. [PMID: 35921110 PMCID: PMC9350714 DOI: 10.1001/jamanetworkopen.2022.25098] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Detection of prion proteins in cerebrospinal fluid (CSF) using real-time quaking-induced conversion (RT-QuIC) assays has transformed the diagnostic approach to sporadic Creutzfeldt-Jakob disease (CJD), facilitating earlier and more complete recognition of affected patients. It is unclear how expanded recognition of affected patients may affect the diagnostic and prognostic relevance of clinical features and diagnostic tests historically associated with CJD. OBJECTIVE To evaluate clinical features and diagnostic testing in patients presenting with CJD and determine the associations of these features with prognosis. DESIGN, SETTING, AND PARTICIPANTS This cohort study incorporated data from electronic medical records of patients with CJD treated at Mayo Clinic Enterprise tertiary care centers in Rochester, Minnesota; Jacksonville, Florida; and Scottsdale, Arizona. Participants included patients with definite or probable CJD assessed from 2014 to 2021. Data were analyzed October 2021 to January 2022. EXPOSURES Dominant presentation, clinical features, and diagnostic tests associated with CJD. MAIN OUTCOMES AND MEASURES The outcomes of interest were the sensitivity and prognostic value of clinical features and accessible diagnostic tests at presentation with possible CJD. RESULTS A total of 115 patients were identified, including 40 patients (35%) with definite CJD. Mean (SD) age at symptom onset was 64.8 (9.4) years, and 68 patients were women (59%). The sensitivity of clinical markers (myoclonus) and tests historically considered in patients with suspected CJD was poor (eg, stereotyped electroencephalography anomalies: 17 of 105 patients [16%]; elevated CSF protein 14-3-3 levels: 54 of 90 patients [60%]). By comparison, biomarkers with good diagnostic sensitivity at presentation included RT-QuIC (66 of 71 patients [93%]), CSF total tau (T-tau) level greater than 1149 pg/mL (81 of 92 patients [88%]), and characteristic signal anomalies on magnetic resonance imaging (88 of 115 patients [77%]). Multivariable linear regression confirmed shorter survival in patients with myoclonus (difference, -125.9 [95% CI, -236.3 to -15.5] days; P = .03), visual or cerebellar signs (difference, -180.2 [95% CI, -282.2 to -78.2] days; P < .001), elevated CSF protein 14-3-3 levels (difference, -193 [95% CI, -304.9 to -82.9] days; P < .001), and elevated T-tau level (difference for every 1000 pg/mL elevation, -9.1 [95% CI, -17.7 to -1.0] days; P = .04). CONCLUSIONS AND RELEVANCE These findings suggest that CSF RT-QuIC, elevated CSF T-tau level, and stereotyped magnetic resonance imaging anomalies were associated with the diagnosis of CJD, while other clinical findings (eg, myoclonus), stereotyped electroencephalography anomalies, and CSF protein 14-3-3 levels offered less diagnostic value. Visual or cerebellar features, myoclonus, and CSF 14-3-3 and T-tau levels may be associated with disease duration, justifying continued inclusion in the evaluation of patients suspected to have CJD.
Collapse
Affiliation(s)
- Dror Shir
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Amanda Porter
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
19
|
Nihat A, Ranson JM, Harris D, McNiven K, Mok T, Rudge P, Collinge J, Llewellyn DJ, Mead S. Development of prognostic models for survival and care status in sporadic Creutzfeldt-Jakob disease. Brain Commun 2022; 4:fcac201. [PMID: 35974795 PMCID: PMC9374480 DOI: 10.1093/braincomms/fcac201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/04/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
Sporadic Creutzfeldt-Jakob disease, the most common human prion disease, typically presents as a rapidly progressive dementia and has a highly variable prognosis. Despite this heterogeneity, clinicians need to give timely advice on likely prognosis and care needs. No prognostic models have been developed that predict survival or time to increased care status from the point of diagnosis. We aimed to develop clinically useful prognostic models with data from a large prospective observational cohort study. Five hundred and thirty-seven patients were visited by mobile teams of doctors and nurses from the National Health Service National Prion Clinic within 5 days of notification of a suspected diagnosis of sporadic Creutzfeldt-Jakob disease, enrolled to the study between October 2008 and March 2020, and followed up until November 2020. Prediction of survival over 10-, 30- and 100-day periods was the main outcome. Escalation of care status over the same time periods was a secondary outcome for a subsample of 113 patients with low care status at initial assessment. Two hundred and eighty (52.1%) patients were female and the median age was 67.2 (interquartile range 10.5) years. Median survival from initial assessment was 24 days (range 0–1633); 414 patients died within 100 days (77%). Ten variables were included in the final prediction models: sex; days since symptom onset; baseline care status; PRNP codon 129 genotype; Medical Research Council Prion Disease Rating Scale, Motor and Cognitive Examination Scales; count of MRI abnormalities; Mini-Mental State Examination score and categorical disease phenotype. The strongest predictor was PRNP codon 129 genotype (odds ratio 6.65 for methionine homozygous compared with methionine-valine heterozygous; 95% confidence interval 3.02–14.68 for 30-day mortality). Of 113 patients with lower care status at initial assessment, 88 (78%) had escalated care status within 100 days, with a median of 35 days. Area under the curve for models predicting outcomes within 10, 30 and 100 days was 0.94, 0.92 and 0.91 for survival, and 0.87, 0.87 and 0.95 for care status escalation, respectively. Models without PRNP codon 129 genotype, which is not immediately available at initial assessment, were also highly accurate. We have developed a model that can accurately predict survival and care status escalation in sporadic Creutzfeldt-Jakob disease patients using clinical, imaging and genetic data routinely available in a specialist national referral service. The utility and generalizability of these models to other settings could be prospectively evaluated when recruiting to clinical trials and providing clinical care.
Collapse
Affiliation(s)
- Akın Nihat
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust , London WC1N 3BG , UK
| | - Janice M Ranson
- College of Medicine and Health, University of Exeter , Exeter EX1 2HZ , UK
- Deep Dementia Phenotyping Network , Exeter EX1 2LU , UK
| | - Dominique Harris
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
| | - Kirsty McNiven
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust , London WC1N 3BG , UK
| | - TzeHow Mok
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust , London WC1N 3BG , UK
| | - Peter Rudge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust , London WC1N 3BG , UK
| | - David J Llewellyn
- College of Medicine and Health, University of Exeter , Exeter EX1 2HZ , UK
- Deep Dementia Phenotyping Network , Exeter EX1 2LU , UK
- Alan Turing Institute , London NW1 2DB , UK
| | - Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases , Cleveland Street, London W1W 7FF , UK
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust , London WC1N 3BG , UK
| |
Collapse
|
20
|
Coysh T, Mead S. The Future of Seed Amplification Assays and Clinical Trials. Front Aging Neurosci 2022; 14:872629. [PMID: 35813946 PMCID: PMC9257179 DOI: 10.3389/fnagi.2022.872629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Prion-like seeded misfolding of host proteins is the leading hypothesised cause of neurodegenerative diseases. The exploitation of the mechanism in the protein misfolding cyclic amplification (PMCA) and real-time quaking-induced conversion (RT-QuIC) assays have transformed prion disease research and diagnosis and have steadily become more widely used for research into other neurodegenerative disorders. Clinical trials in adult neurodegenerative diseases have been expensive, slow, and disappointing in terms of clinical benefits. There are various possible factors contributing to the failure to identify disease-modifying treatments for adult neurodegenerative diseases, some of which include: limited accuracy of antemortem clinical diagnosis resulting in the inclusion of patients with the “incorrect” pathology for the therapeutic; the role of co-pathologies in neurodegeneration rendering treatments targeting one pathology alone ineffective; treatment of the primary neurodegenerative process too late, after irreversible secondary processes of neurodegeneration have become established or neuronal loss is already extensive; and preclinical models used to develop treatments not accurately representing human disease. The use of seed amplification assays in clinical trials offers an opportunity to tackle these problems by sensitively detecting in vivo the proteopathic seeds thought to be central to the biology of neurodegenerative diseases, enabling improved diagnostic accuracy of the main pathology and co-pathologies, and very early intervention, particularly in patients at risk of monogenic forms of neurodegeneration. The possibility of quantifying proteopathic seed load, and its reduction by treatments, is an attractive pharmacodynamic biomarker in the preclinical and early clinical stages of drug development. Here we review some potential applications of seed amplification assays in clinical trials.
Collapse
Affiliation(s)
- Thomas Coysh
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- National Prion Clinic, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
21
|
Schmitz M, Canaslan S, Espinosa JC, Fernández-Borges N, Villar-Piqué A, Llorens F, Varges D, Maass F, Torres JM, Hermann P, Zerr I. Validation of Plasma and CSF Neurofilament Light Chain as an Early Marker for Sporadic Creutzfeldt-Jakob Disease. Mol Neurobiol 2022; 59:1-9. [PMID: 35716271 DOI: 10.1007/s12035-022-02891-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
Biomarkers are becoming increasingly important for the differential diagnosis of neurodegenerative diseases. Previous observations indicated neurofilament light chain (NfL) as a potential blood-based biomarker for sporadic Creutzfeldt-Jakob disease (sCJD). Here, we investigated the stability, inter-assay/intra-assay variation and the regulation of NfL levels in CSF and plasma in a large cohort of sCJD patients by using a single-molecule array (SIMOA). We defined cutoffs for an accurate diagnosis and measured plasma NfL level in prion-infected mice models at different time points to identify the potential dynamics throughout the disease. Our analyses confirmed CSF and plasma NfL as stable and consistent marker for sCJD. Receiver operating characteristic (ROC) curve analysis showed an AUC of 0.92-0.93 to distinguish sCJD from control groups. Newly defined cutoffs revealed good diagnostic accuracies of CSF and plasma NfL, indicated by a sensitivity of 80-83.5% and a specificity of 87.4-91%. Studies on two humanized prion-infected mice lines (Tg340-PRNP 129MM and Tg361-PRNP 129VV) revealed increased plasma NfL levels in a late pre-clinical or very early clinical stage between 120-150 days post-inoculation. In conclusion, our work supports the potential use of CSF and plasma NfL as a very early biomarker in sCJD diagnostic with good diagnostic accuracies.
Collapse
Affiliation(s)
- Matthias Schmitz
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
| | - Sezgi Canaslan
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Juan Carlos Espinosa
- Centro de Investigación en Sanidad Animal-Instituto Nacional de Investigación Y Tecnología Agraria Y Alimentaria-Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), 28130, Madrid, Spain
| | - Natalia Fernández-Borges
- Centro de Investigación en Sanidad Animal-Instituto Nacional de Investigación Y Tecnología Agraria Y Alimentaria-Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), 28130, Madrid, Spain
| | - Anna Villar-Piqué
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,CIBERNED (Network Center for Biomedical Research of Neurodegenerative Diseases), Institute Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Franc Llorens
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,CIBERNED (Network Center for Biomedical Research of Neurodegenerative Diseases), Institute Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Daniela Varges
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Fabian Maass
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Juan Maria Torres
- Centro de Investigación en Sanidad Animal-Instituto Nacional de Investigación Y Tecnología Agraria Y Alimentaria-Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), 28130, Madrid, Spain
| | - Peter Hermann
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medicine Göttingen, National Reference Center for TSE and the German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
22
|
Blood phospho-tau in Alzheimer disease: analysis, interpretation, and clinical utility. Nat Rev Neurol 2022; 18:400-418. [PMID: 35585226 DOI: 10.1038/s41582-022-00665-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 12/11/2022]
Abstract
Well-authenticated biomarkers can provide critical insights into the biological basis of Alzheimer disease (AD) to enable timely and accurate diagnosis, estimate future burden and support therapeutic trials. Current cerebrospinal fluid and molecular neuroimaging biomarkers fulfil these criteria but lack the scalability and simplicity necessary for widespread application. Blood biomarkers of adequate effectiveness have the potential to act as first-line diagnostic and prognostic tools, and offer the possibility of extensive population screening and use that is not limited to specialized centres. Accelerated progress in our understanding of the biochemistry of brain-derived tau protein and advances in ultrasensitive technologies have enabled the development of AD-specific phosphorylated tau (p-tau) biomarkers in blood. In this Review we discuss how new information on the molecular processing of brain p-tau and secretion of specific fragments into biofluids is informing blood biomarker development, enabling the evaluation of preanalytical factors that affect quantification, and informing harmonized protocols for blood handling. We also review the performance of blood p-tau biomarkers in the context of AD and discuss their potential contexts of use for clinical and research purposes. Finally, we highlight outstanding ethical, clinical and analytical challenges, and outline the steps that need to be taken to standardize inter-laboratory and inter-assay measurements.
Collapse
|
23
|
Affiliation(s)
- Inga Zerr
- From the National Reference Center for Human Prion Diseases, Clinical Dementia Center, Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| |
Collapse
|
24
|
Mead S, Khalili-Shirazi A, Potter C, Mok T, Nihat A, Hyare H, Canning S, Schmidt C, Campbell T, Darwent L, Muirhead N, Ebsworth N, Hextall P, Wakeling M, Linehan J, Libri V, Williams B, Jaunmuktane Z, Brandner S, Rudge P, Collinge J. Prion protein monoclonal antibody (PRN100) therapy for Creutzfeldt-Jakob disease: evaluation of a first-in-human treatment programme. Lancet Neurol 2022; 21:342-354. [PMID: 35305340 DOI: 10.1016/s1474-4422(22)00082-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Human prion diseases, including Creutzfeldt-Jakob disease (CJD), are rapidly progressive, invariably fatal neurodegenerative conditions with no effective therapies. Their pathogenesis involves the obligate recruitment of cellular prion protein (PrPC) into self-propagating multimeric assemblies or prions. Preclinical studies have firmly validated the targeting of PrPC as a therapeutic strategy. We aimed to evaluate a first-in-human treatment programme using an anti-PrPC monoclonal antibody under a Specials Licence. METHODS We generated a fully humanised anti-PrPC monoclonal antibody (an IgG4κ isotype; PRN100) for human use. We offered treatment with PRN100 to six patients with a clinical diagnosis of probable CJD who were not in the terminal disease stages at the point of first assessment and who were able to readily travel to the University College London Hospital (UCLH) Clinical Research Facility, London, UK, for treatment. After titration (1 mg/kg and 10 mg/kg at 48-h intervals), patients were treated with 80-120 mg/kg of intravenous PRN100 every 2 weeks until death or withdrawal from the programme, or until the supply of PRN100 was exhausted, and closely monitored for evidence of adverse effects. Disease progression was assessed by use of the Medical Research Council (MRC) Prion Disease Rating Scale, Motor Scale, and Cognitive Scale, and compared with that of untreated natural history controls (matched for disease severity, subtype, and PRNP codon 129 genotype) recruited between Oct 1, 2008, and July 31, 2018, from the National Prion Monitoring Cohort study. Autopsies were done in two patients and findings were compared with those from untreated natural history controls. FINDINGS We treated six patients (two men; four women) with CJD for 7-260 days at UCLH between Oct 9, 2018, and July 31, 2019. Repeated intravenous dosing of PRN100 was well tolerated and reached the target CSF drug concentration (50 nM) in four patients after 22-70 days; no clinically significant adverse reactions were seen. All patients showed progressive neurological decline on serial assessments with the MRC Scales. Neuropathological examination was done in two patients (patients 2 and 3) and showed no evidence of cytotoxicity. Patient 2, who was treated for 140 days, had the longest clinical duration we have yet documented for iatrogenic CJD and showed patterns of disease-associated PrP that differed from untreated patients with CJD, consistent with drug effects. Patient 3, who had sporadic CJD and only received one therapeutic dose of 80 mg/kg, had weak PrP synaptic labelling in the periventricular regions, which was not a feature of untreated patients with sporadic CJD. Brain tissue-bound drug concentrations across multiple regions in patient 2 ranged from 9·9 μg per g of tissue (SD 0·3) in the thalamus to 27·4 μg per g of tissue (1·5) in the basal ganglia (equivalent to 66-182 nM). INTERPRETATION Our academic-led programme delivered what is, to our knowledge, the first rationally designed experimental treatment for human prion disease to a small number of patients with CJD. The treatment appeared to be safe and reached encouraging CSF and brain tissue concentrations. These findings justify the need for formal efficacy trials in patients with CJD at the earliest possible clinical stages and as prophylaxis in those at risk of prion disease due to PRNP mutations or prion exposure. FUNDING The Cure CJD Campaign, the National Institute for Health Research UCLH Biomedical Research Centre, the Jon Moulton Charitable Trust, and the UK MRC.
Collapse
Affiliation(s)
- Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK
| | - Azadeh Khalili-Shirazi
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK
| | - Caroline Potter
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Tzehow Mok
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK
| | - Akin Nihat
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK
| | - Harpreet Hyare
- Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Stephanie Canning
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Christian Schmidt
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Tracy Campbell
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Lee Darwent
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Nicola Muirhead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Nicolette Ebsworth
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Patrick Hextall
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Madeleine Wakeling
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Jacqueline Linehan
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK
| | - Vincenzo Libri
- NIHR, Biomedical Research Centre, University College London Hospitals, London, UK; Clinical Research Facility, University College London Hospitals, London, UK
| | - Bryan Williams
- NIHR, Biomedical Research Centre, University College London Hospitals, London, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Sebastian Brandner
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Peter Rudge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, University College London, London, UK; National Prion Clinic, National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
25
|
Altuna M, Ruiz I, Zelaya MV, Mendioroz M. Role of Biomarkers for the Diagnosis of Prion Diseases: A Narrative Review. Medicina (B Aires) 2022; 58:medicina58040473. [PMID: 35454316 PMCID: PMC9030755 DOI: 10.3390/medicina58040473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022] Open
Abstract
Prion diseases are progressive and irreversible neurodegenerative disorders with a low incidence (1.5–2 cases per million per year). Genetic (10–15%), acquired (anecdotal) and sporadic (85%) forms of the disease have been described. The clinical spectrum of prion diseases is very varied, although the most common symptoms are rapidly progressive dementia, cerebellar ataxia and myoclonus. Mean life expectancy from the onset of symptoms is 6 months. There are currently diagnostic criteria based on clinical phenotype, as well as neuroimaging biomarkers (magnetic resonance imaging), neurophysiological tests (electroencephalogram and polysomnogram), and cerebrospinal fluid biomarkers (14-3-3 protein and real-time quaking-induced conversion (RT-QuIC)). The sensitivity and specificity of some of these tests (electroencephalogram and 14-3-3 protein) is under debate and the applicability of other tests, such as RT-QuIC, is not universal. However, the usefulness of these biomarkers beyond the most frequent prion disease, sporadic Creutzfeldt–Jakob disease, remains unclear. Therefore, research is being carried out on new, more efficient cerebrospinal fluid biomarkers (total tau, ratio total tau/phosphorylated tau and neurofilament light chain) and potential blood biomarkers (neurofilament light chain, among others) to try to universalize access to early diagnosis in the case of prion diseases.
Collapse
Affiliation(s)
- Miren Altuna
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau—Biomedical Research Institute Sant Pau—Universitat Autònoma de Barcelona, 08041 Barcelona, Spain;
- Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- CITA-Alzheimer Foundation, 20009 Donostia-San Sebastián, Spain
- Correspondence: ; Tel.: +34-935-56-59-86; Fax: +34-935-56-56-02
| | - Iñigo Ruiz
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau—Biomedical Research Institute Sant Pau—Universitat Autònoma de Barcelona, 08041 Barcelona, Spain;
| | - María Victoria Zelaya
- Department of Pathological Anatomy, Hospital Universitario de Navarra, 31008 Pamplona, Spain;
| | - Maite Mendioroz
- Department of Neurology, Hospital Universitario de Navarra, 31008 Pamplona, Spain;
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31006 Pamplona, Spain
| |
Collapse
|
26
|
Mortberg MA, Zhao HT, Reidenbach AG, Gentile JE, Kuhn E, O'Moore J, Dooley PM, Connors TR, Mazur C, Allen SW, Trombetta BA, McManus AJ, Moore MR, Liu J, Cabin DE, Kordasiewicz HB, Mathews J, Arnold SE, Vallabh SM, Minikel EV. PrP concentration in the central nervous system: regional variability, genotypic effects, and pharmacodynamic impact. JCI Insight 2022; 7:156532. [PMID: 35133987 PMCID: PMC8986079 DOI: 10.1172/jci.insight.156532] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/04/2022] [Indexed: 11/17/2022] Open
Abstract
Prion protein (PrP) concentration controls the kinetics of prion replication and is a genetically and pharmacologically validated therapeutic target for prion disease. In order to evaluate PrP concentration as a pharmacodynamic biomarker and assess its contribution to known prion disease risk factors, we developed and validated a plate-based immunoassay reactive for PrP across six species of interest and applicable to brain and cerebrospinal fluid (CSF). PrP concentration varies dramatically between different brain regions in mice, cynomolgus macaques, and humans. PrP expression does not appear to contribute to the known risk factors of age, sex, or common PRNP genetic variants. CSF PrP is lowered in the presence of rare pathogenic PRNP variants, with heterozygous carriers of P102L displaying 55% and of D178N just 31% the CSF PrP concentration of mutation-negative controls. In rodents, pharmacologic reduction of brain Prnp RNA is reflected in brain parenchyma PrP, and in turn in CSF PrP, validating CSF as a sampling compartment for the effect of PrP-lowering therapy. Our findings support the use of CSF PrP as a pharmacodynamic biomarker for PrP-lowering drugs, and suggest that relative reduction from individual baseline CSF PrP concentration may be an appropriate marker for target engagement.
Collapse
Affiliation(s)
- Meredith A Mortberg
- Stanley Center for Psychiatric Research, Broad Institute of Harvard & MIT, Cambridge, United States of America
| | - Hien T Zhao
- Neuroscience, Ionis Pharmaceuticals, Inc., Carlsbad, United States of America
| | - Andrew G Reidenbach
- Stanley Center for Psychiatric Research, Broad Institute of Harvard & MIT, Cambridge, United States of America
| | - Juliana E Gentile
- Stanley Center for Psychiatric Research, Broad Institute of Harvard & MIT, Cambridge, United States of America
| | - Eric Kuhn
- Proteomics Platform, Broad Institute of Harvard & MIT, Cambridge, United States of America
| | - Jill O'Moore
- Comparative Medicine, McLaughlin Research Institute, Great Falls, United States of America
| | - Patrick M Dooley
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, United States of America
| | - Theresa R Connors
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, United States of America
| | - Curt Mazur
- Neuroscience, Ionis Pharmaceuticals, Inc., Carlsbad, United States of America
| | - Shona W Allen
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, United States of America
| | - Bianca A Trombetta
- Department of Neurology, Massachusetts General Hospital, Boston, United States of America
| | - Alison J McManus
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, United States of America
| | | | - Jiewu Liu
- Bioagilytix, Bioagilytix, Boston, United States of America
| | - Deborah E Cabin
- Comparative Medicine, McLaughlin Research Institute, Great Falls, United States of America
| | | | - Joel Mathews
- Neuroscience, Ionis Pharmaceuticals, Inc., Carlsbad, United States of America
| | - Steven E Arnold
- Department of Neurology, Massachusetts General Hospital, Boston, United States of America
| | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard & MIT, Cambridge, United States of America
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of Harvard & MIT, Cambridge, United States of America
| |
Collapse
|
27
|
Halbgebauer S, Abu-Rumeileh S, Oeckl P, Steinacker P, Roselli F, Wiesner D, Mammana A, Beekes M, Kortazar-Zubizarreta I, Perez de Nanclares G, Capellari S, Giese A, Castilla J, Ludolph AC, Žáková D, Parchi P, Otto M. Blood β-Synuclein and Neurofilament Light Chain During the Course of Prion Disease. Neurology 2022; 98:e1434-e1445. [PMID: 35110380 DOI: 10.1212/wnl.0000000000200002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES For early diagnosis and disease monitoring of neurodegenerative diseases (NDs) reliable blood biomarkers are needed. Elevated levels of neurofilament light chain protein (NfL), an axonal damage marker, have been described across different NDs with highest values in prion diseases and amyotrophic lateral sclerosis (ALS). Synaptic degeneration is a common early feature in most NDs and seems to precede neuronal degeneration in prion disease. However, synaptic markers in blood are still missing. Here we investigated if the brain specific protein beta-synuclein might be a suitable blood biomarker for early diagnosis and evaluation of synaptic integrity in prion disease. METHODS We analyzed blood beta-synuclein with a newly established digital ELISA and NfL with single molecule array in samples obtained from human subjects and prion and ALS animal models. Furthermore, beta-synuclein was investigated in brain tissue of Creutzfeldt-Jakob disease (CJD) and control cases. RESULTS We investigated 308 patients including 129 prion disease cases, 8 presymptomatic PRNP mutation carriers, 60 ALS, 68 other ND and 43 control patients. In CJD symptomatic cases beta-synuclein and NfL were markedly increased compared to all other diagnostic groups (p<0.001). In the large majority of pre-symptomatic PRNP mutation carriers beta-synuclein and NfL levels were within normal range. In prion disease animal models, beta-synuclein and NfL displayed normal levels in the pre-symptomatic phase with a sudden elevation at disease onset and a plateau in the symptomatic phase. In contrast to NfL, beta-synuclein was neither elevated in symptomatic ALS patients nor in an ALS animal model. In the discrimination between prion disease and all other groups beta-synuclein (AUC: 0.97, 95% CI: 0.94-0.99, p<0.001) was superior to NfL (AUC: 0.91, 95% CI: 0.88-0.94, p<0.001). Additionally, brain tissue beta-synuclein showed significantly reduced levels in CJD compared to control patients (p<0.001). DISCUSSION Blood beta-synuclein was significantly elevated in CJD patients reflecting ongoing synaptic damage and showed good discriminative characteristics. We therefore propose it as a candidate blood marker for early diagnosis and monitoring of synaptic integrity in prion disease. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that serum beta synuclein concentration accurately distinguishes patients with symptomatic CJD from controls.
Collapse
Affiliation(s)
- Steffen Halbgebauer
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany.,Department of Neurology, Halle University Hospital, Martin Luther University Halle/Wittenberg, Ernst-Grube Strasse 49, 06120 Halle (Saale), Germany
| | - Patrick Oeckl
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Petra Steinacker
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Diana Wiesner
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Michael Beekes
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | | | | | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Armin Giese
- Department of Neuropathology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Albert C Ludolph
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Dana Žáková
- Department of Prion Diseases, Slovak Medical University, Bratislava, Slovakia
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081 Ulm, Germany .,Department of Neurology, Halle University Hospital, Martin Luther University Halle/Wittenberg, Ernst-Grube Strasse 49, 06120 Halle (Saale), Germany
| |
Collapse
|
28
|
NT1-Tau Is Increased in CSF and Plasma of CJD Patients, and Correlates with Disease Progression. Cells 2021; 10:cells10123514. [PMID: 34944022 PMCID: PMC8700417 DOI: 10.3390/cells10123514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 11/21/2022] Open
Abstract
This study investigates the diagnostic and prognostic potential of different forms of tau in biofluids from patients with Creutzfeldt-Jakob disease (CJD). Extracellular tau, which is molecularly heterogeneous, was measured using ultra-sensitive custom-made Simoa assays for N-terminal (NT1), mid-region, and full-length tau. We assessed cross-sectional CSF and plasma from healthy controls, patients with Alzheimer’s disease (AD) and CJD patients. Then, we evaluated the correlation of the best-performing tau assay (NT1-tau) with clinical severity and functional decline (using the MRC Prion Disease Rating Scale) in a longitudinal CJD cohort (n = 145). In a cross-sectional study, tau measured in CSF with the NT1 and mid-region Simoa assays, separated CJD (n = 15) from AD (n = 18) and controls (n = 21) with a diagnostic accuracy (AUCs: 0.98–1.00) comparable to or better than neurofilament light chain (NfL; AUCs: 0.96–0.99). In plasma, NT1-measured tau was elevated in CJD (n = 5) versus AD (n = 15) and controls (n = 15). Moreover, in CJD plasma (n = 145) NT1-tau levels correlated with stage and rate of disease progression, and the effect on clinical progression was modified by the PRNP codon 129. Our findings suggest that plasma NT1-tau shows promise as a minimally invasive diagnostic and prognostic biomarker of CJD, and should be further investigated for its potential to monitor disease progression and response to therapies.
Collapse
|
29
|
Licinio J, Wong ML. Molecular Psychiatry special issue: advances in Alzheimer's disease. Mol Psychiatry 2021; 26:5467-5470. [PMID: 35027660 DOI: 10.1038/s41380-021-01434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Julio Licinio
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Ma-Li Wong
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| |
Collapse
|
30
|
Yuan A, Nixon RA. Neurofilament Proteins as Biomarkers to Monitor Neurological Diseases and the Efficacy of Therapies. Front Neurosci 2021; 15:689938. [PMID: 34646114 PMCID: PMC8503617 DOI: 10.3389/fnins.2021.689938] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Biomarkers of neurodegeneration and neuronal injury have the potential to improve diagnostic accuracy, disease monitoring, prognosis, and measure treatment efficacy. Neurofilament proteins (NfPs) are well suited as biomarkers in these contexts because they are major neuron-specific components that maintain structural integrity and are sensitive to neurodegeneration and neuronal injury across a wide range of neurologic diseases. Low levels of NfPs are constantly released from neurons into the extracellular space and ultimately reach the cerebrospinal fluid (CSF) and blood under physiological conditions throughout normal brain development, maturation, and aging. NfP levels in CSF and blood rise above normal in response to neuronal injury and neurodegeneration independently of cause. NfPs in CSF measured by lumbar puncture are about 40-fold more concentrated than in blood in healthy individuals. New ultra-sensitive methods now allow minimally invasive measurement of these low levels of NfPs in serum or plasma to track disease onset and progression in neurological disorders or nervous system injury and assess responses to therapeutic interventions. Any of the five Nf subunits - neurofilament light chain (NfL), neurofilament medium chain (NfM), neurofilament heavy chain (NfH), alpha-internexin (INA) and peripherin (PRPH) may be altered in a given neuropathological condition. In familial and sporadic Alzheimer's disease (AD), plasma NfL levels may rise as early as 22 years before clinical onset in familial AD and 10 years before sporadic AD. The major determinants of elevated levels of NfPs and degradation fragments in CSF and blood are the magnitude of damaged or degenerating axons of fiber tracks, the affected axon caliber sizes and the rate of release of NfP and fragments at different stages of a given neurological disease or condition directly or indirectly affecting central nervous system (CNS) and/or peripheral nervous system (PNS). NfPs are rapidly emerging as transformative blood biomarkers in neurology providing novel insights into a wide range of neurological diseases and advancing clinical trials. Here we summarize the current understanding of intracellular NfP physiology, pathophysiology and extracellular kinetics of NfPs in biofluids and review the value and limitations of NfPs and degradation fragments as biomarkers of neurodegeneration and neuronal injury.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
- Department of Cell Biology, New York University Grossman School of Medicine, (NYU), Neuroscience Institute, New York, NY, United States
| |
Collapse
|
31
|
Figgie MP, Appleby BS. Clinical Use of Improved Diagnostic Testing for Detection of Prion Disease. Viruses 2021; 13:v13050789. [PMID: 33925126 PMCID: PMC8146465 DOI: 10.3390/v13050789] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Prion diseases are difficult to recognize as many symptoms are shared among other neurologic pathologies and the full spectra of symptoms usually do not appear until late in the disease course. Additionally, many commonly used laboratory markers are non-specific to prion disease. The recent introduction of second-generation real time quaking induced conversion (RT-QuIC) has revolutionized pre-mortem diagnosis of prion disease due to its extremely high sensitivity and specificity. However, RT-QuIC does not provide prognostic data and has decreased diagnostic accuracy in some rarer, atypical prion diseases. The objective of this review is to provide an overview of the current clinical utility of fluid-based biomarkers, neurodiagnostic testing, and brain imaging in the diagnosis of prion disease and to suggest guidelines for their clinical use, with a focus on rarer prion diseases with atypical features. Recent advancements in laboratory-based testing and imaging criteria have shown improved diagnostic accuracy and prognostic potential in prion disease, but because these diagnostic tests are not sensitive in some prion disease subtypes and diagnostic test sensitivities are unknown in the event that CWD transmits to humans, it is important to continue investigations into the clinical utility of various testing modalities.
Collapse
Affiliation(s)
- Mark P. Figgie
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Brian S. Appleby
- Department of Neurology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA;
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence:
| |
Collapse
|
32
|
Zerr I, Villar-Piqué A, Hermann P, Schmitz M, Varges D, Ferrer I, Riggert J, Zetterberg H, Blennow K, Llorens F. Diagnostic and prognostic value of plasma neurofilament light and total-tau in sporadic Creutzfeldt-Jakob disease. Alzheimers Res Ther 2021; 13:86. [PMID: 33883011 PMCID: PMC8059191 DOI: 10.1186/s13195-021-00815-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Blood neurofilament light (Nfl) and total-tau (t-tau) have been described to be increased in several neurological conditions, including prion diseases and other neurodegenerative dementias. Here, we aim to determine the accuracy of plasma Nfl and t-tau in the differential diagnosis of neurodegenerative dementias and their potential value as prognostic markers of disease severity. METHODS Plasma Nfl and t-tau were measured in healthy controls (HC, n = 70), non-neurodegenerative neurological disease with (NND-Dem, n = 17) and without dementia syndrome (NND, n = 26), Alzheimer's disease (AD, n = 44), Creutzfeldt-Jakob disease (CJD, n = 83), dementia with Lewy bodies/Parkinson's disease with dementia (DLB/PDD, n = 35), frontotemporal dementia (FTD, n = 12), and vascular dementia (VaD, n = 22). Biomarker diagnostic accuracies and cutoff points for the diagnosis of CJD were calculated, and associations between Nfl and t-tau concentrations with other fluid biomarkers, demographic, genetic, and clinical data in CJD cases were assessed. Additionally, the value of Nfl and t-tau predicting disease survival in CJD was evaluated. RESULTS Among diagnostic groups, highest plasma Nfl and t-tau concentrations were detected in CJD (fold changes of 38 and 18, respectively, compared to HC). Elevated t-tau was able to differentiate CJD from all other groups, whereas elevated Nfl concentrations were also detected in NND-Dem, AD, DLB/PDD, FTD, and VaD compared to HC. Both biomarkers discriminated CJD from non-CJD dementias with an AUC of 0.93. In CJD, plasma t-tau, but not Nfl, was associated with PRNP codon 129 genotype and CJD subtype. Positive correlations were observed between plasma Nfl and t-tau concentrations, as well as between plasma and CSF concentrations of both biomarkers (p < 0.001). Nfl was increased in rapidly progressive AD (rpAD) compared to slow progressive AD (spAD) and associated to Mini-Mental State Examination results. However, Nfl displayed higher accuracy than t-tau discriminating CJD from rpAD and spAD. Finally, plasma t-tau, but not plasma Nfl, was significantly associated with disease duration, offering a moderate survival prediction capacity. CONCLUSIONS Plasma Nfl and t-tau are useful complementary biomarkers for the differential diagnosis of CJD. Additionally, plasma t-tau emerges as a potential prognostic marker of disease duration.
Collapse
Affiliation(s)
- Inga Zerr
- Department of Neurology, National Reference Center for TSE Surveillance, University Medical Center, Robert-Koch Street 40, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Anna Villar-Piqué
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), L'Hospitalet de Llobregat, Feixa Llarga s/n, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Peter Hermann
- Department of Neurology, National Reference Center for TSE Surveillance, University Medical Center, Robert-Koch Street 40, Göttingen, Germany.
| | - Matthias Schmitz
- Department of Neurology, National Reference Center for TSE Surveillance, University Medical Center, Robert-Koch Street 40, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Daniela Varges
- Department of Neurology, National Reference Center for TSE Surveillance, University Medical Center, Robert-Koch Street 40, Göttingen, Germany
| | - Isidre Ferrer
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), L'Hospitalet de Llobregat, Feixa Llarga s/n, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Joachim Riggert
- Department of Transfusion Medicine, University Medical School, Göttingen, Germany
| | - 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
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute, 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
| | - Franc Llorens
- Department of Neurology, National Reference Center for TSE Surveillance, University Medical Center, Robert-Koch Street 40, Göttingen, Germany.
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), L'Hospitalet de Llobregat, Feixa Llarga s/n, Barcelona, Spain.
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
33
|
Abu-Rumeileh S, Parchi P. Cerebrospinal Fluid and Blood Neurofilament Light Chain Protein in Prion Disease and Other Rapidly Progressive Dementias: Current State of the Art. Front Neurosci 2021; 15:648743. [PMID: 33776643 PMCID: PMC7994519 DOI: 10.3389/fnins.2021.648743] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Rapidly progressive dementia (RPD) is an umbrella term referring to several conditions causing a rapid neurological deterioration associated with cognitive decline and short disease duration. They comprise Creutzfeldt–Jakob disease (CJD), the archetypal RPD, rapidly progressive variants of the most common neurodegenerative dementias (NDs), and potentially treatable conditions such as infectious or autoimmune encephalitis and cerebrovascular disease. Given the significant clinical and, sometimes, neuroradiological overlap between these different disorders, biofluid markers also contribute significantly to the differential diagnosis. Among them, the neurofilament light chain protein (NfL) has attracted growing attention in recent years as a biofluid marker of neurodegeneration due to its sensitivity to axonal damage and the reliability of its measurement in both cerebrospinal fluid (CSF) and blood. Here, we summarize current knowledge regarding biological and clinical implications of NfL evaluation in biofluids across RPDs, emphasizing CJD, and other prion diseases. In the latter, NfL demonstrated a good diagnostic and prognostic accuracy and a potential value as a marker of proximity to clinical onset in pre-symptomatic PRNP mutation carriers. Similarly, in Alzheimer’s disease and other NDs, higher NfL concentrations seem to predict a faster disease progression. While increasing evidence indicates a potential clinical value of NfL in monitoring cerebrovascular disease, the association between NfL and prediction of outcome and/or disease activity in autoimmune encephalitis and infectious diseases has only been investigated in few cohorts and deserves confirmatory studies. In the era of precision medicine and evolving therapeutic options, CSF and blood NfL might aid the diagnostic and prognostic assessment of RPDs and the stratification and management of patients according to disease progression in clinical trials.
Collapse
Affiliation(s)
| | - Piero Parchi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto delle Sciente Neurologiche di Bologna, Bologna, Italy.,Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|