1
|
Chisholm TS, Hunter CA. Ligands for Protein Fibrils of Amyloid-β, α-Synuclein, and Tau. Chem Rev 2025; 125:5282-5348. [PMID: 40327808 PMCID: PMC12164286 DOI: 10.1021/acs.chemrev.4c00838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025]
Abstract
Amyloid fibrils are characteristic features of many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. The use of small molecule ligands that bind to amyloid fibrils underpins both fundamental research aiming to better understand the pathology of neurodegenerative disease, and clinical research aiming to develop diagnostic tools for these diseases. To date, a large number of amyloid-binding ligands have been reported in the literature, predominantly targeting protein fibrils composed of amyloid-β (Aβ), tau, and α-synuclein (αSyn) fibrils. Fibrils formed by a particular protein can adopt a range of possible morphologies, but protein fibrils formed in vivo possess disease-specific morphologies, highlighting the need for morphology-specific amyloid-binding ligands. This review details the morphologies of Aβ, tau, and αSyn fibril polymorphs that have been reported as a result of structural work and describes a database of amyloid-binding ligands containing 4,288 binding measurements for 2,404 unique compounds targeting Aβ, tau, or αSyn fibrils.
Collapse
Affiliation(s)
- Timothy S. Chisholm
- Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.
| | - Christopher A. Hunter
- Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.
| |
Collapse
|
2
|
Mao M, Zhou Y, Zhang H, Deng P, Yang J, Zhong J, Li N, Liu Q, Li X, Wu X, Cheng Y. Synthesis and identification of azocoumarin derivatives toward imaging of α-synuclein aggregates in the brain. Eur J Med Chem 2025; 290:117587. [PMID: 40168910 DOI: 10.1016/j.ejmech.2025.117587] [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: 01/25/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
To identify α-synuclein aggregation in synucleinopathies is still challenging, due to the lack of specific probes for α-synuclein aggregates with efficient brain uptake. In this work, compact molecules based on coumarin scaffold were synthesized and evaluated for detection and bioimaging of α-synuclein aggregates in the brain. Among the developed compounds, azocoumarin 5 containing push-pull electronic architecture featured selective fluorescence enhancement towards α-synuclein aggregates in comparison to other β-sheet protein species (β-amyloid, tau). In addition, azocoumarin [18F]Cou-NNF was succesfully developed, and demonstrated its potential as radiotracer for imaging brain α-synuclein aggregates, owing to its favorable affinity for α-synuclein aggregates accompanied with efficient brain uptake and little defluorination in vivo. Overall, compact azocoumarin provides an effective lead structure for developing α-synuclein probes, and N=N bond shows promise in enhancing selective affinity for α-synuclein aggregates.
Collapse
Affiliation(s)
- Meiting Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Huihui Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Pengxin Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jie Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jing Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Na Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qiangqiang Liu
- Chengdu New Radiomedicine Technology Co., Ltd., Chengdu, 610200, China
| | - Xianghui Li
- Chengdu New Radiomedicine Technology Co., Ltd., Chengdu, 610200, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yan Cheng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Schuler M, Gerwert G, Mann M, Woitzik N, Langenhoff L, Hubert D, Duman D, Höveler A, Galkowski S, Simon J, Denz R, Weber S, Kwon EH, Wanka R, Kötting C, Güldenhaupt J, Beyer L, Tönges L, Mollenhauer B, Gerwert K. Alpha-synuclein misfolding as fluid biomarker for Parkinson's disease measured with the iRS platform. EMBO Mol Med 2025; 17:1203-1221. [PMID: 40281305 PMCID: PMC12162852 DOI: 10.1038/s44321-025-00229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Misfolding and aggregation of alpha-synuclein (αSyn) play a key role in the pathophysiology of Parkinson's disease (PD). Despite considerable advances in diagnostics, an early and differential diagnosis of PD still represents a major challenge. We innovated the immuno-infrared sensor (iRS) platform for measuring αSyn misfolding. We analyzed cerebrospinal fluid (CSF) from two cohorts comprising PD cases, atypical Parkinsonian disorders, and disease controls. We obtained an AUC of 0.90 (n = 134, 95% CI 0.85-0.96) for separating PD/MSA from controls by determination of the αSyn misfolding by iRS. Using two thresholds divided individuals as unaffected/affected by misfolding with an intermediate area in between. Comparing the affected/unaffected cases, controls versus PD/MSA cases were classified with 97% sensitivity and 92% specificity. The spectral data revealed misfolding from an α-helical/random-coil αSyn in controls to β-sheet enriched αSyn in PD and MSA cases. Moreover, a first subgroup analysis implied the potential for patient stratification in clinically overlapping cases. The iRS, directly measuring all αSyn conformers, is complementary to the αSyn seed-amplification assays (SAAs), which however only amplify seeding competent conformers.
Collapse
Affiliation(s)
- Martin Schuler
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Grischa Gerwert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Marvin Mann
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Nathalie Woitzik
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Lennart Langenhoff
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Diana Hubert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Deniz Duman
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Adrian Höveler
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Sandy Galkowski
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Jonas Simon
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Robin Denz
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - Sandrina Weber
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Eun-Hae Kwon
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Robin Wanka
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Kötting
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Jörn Güldenhaupt
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Léon Beyer
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Brit Mollenhauer
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
- Paracelsus-Elena-Klinik, Klinikstraße 16, 34128, Kassel, Germany
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany.
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
4
|
Carrazana E, Montalbán-Gutiérrez L, Chana-Cuevas P, Salvadores N. Advancing Parkinson's diagnosis: seed amplification assay for α-synuclein detection in minimally invasive samples. Mol Cell Biochem 2025; 480:3297-3314. [PMID: 39760833 DOI: 10.1007/s11010-024-05190-y] [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: 09/09/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by tremor, rigidity, and bradykinesia, beginning with early loss of dopaminergic neurons in the ventrolateral substantia nigra and advancing to broader neurodegeneration in the midbrain. The clinical heterogeneity of PD and the lack of specific diagnostic tests present significant challenges, highlighting the need for reliable biomarkers for early diagnosis. Alpha-synuclein (α-Syn), a protein aggregating into Lewy bodies and neurites in PD patients, has emerged as a key biomarker due to its central role in PD pathophysiology and potential to reflect pathological processes. Additionally, α-Syn allows earlier differentiation between PD and other neurodegenerative disorders with similar symptoms. Currently, detection of α-Syn pathology in post-mortem brain tissue remains the primary means of achieving a conclusive diagnosis, often revealing significant misdiagnoses. Seed amplification assay (SAA), initially developed for prion diseases, has been adapted to detect α-Syn aggregates in cerebrospinal fluid, showing promise for early diagnosis. Recent studies have demonstrated that SAA can also detect α-Syn aggregates in peripheral samples collected via minimally invasive procedures, such as skin, olfactory mucosa, saliva, and blood. However, the lack of standardized protocols limits clinical application. Standardizing protocols is essential to improve assay reliability and enable accurate patient identification for emerging therapies. This review examines studies on SAA for detecting α-Syn aggregates in minimally invasive samples, focusing on sample collection, processing, and reaction conditions.
Collapse
Affiliation(s)
- Elizabeth Carrazana
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile
| | - Leonardo Montalbán-Gutiérrez
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile
- Escuela de Medicina, Facultad de Medicina y Ciencias de La Salud, Universidad Mayor, Temuco, Chile
| | - Pedro Chana-Cuevas
- Centro de Trastornos del Movimiento (CETRAM), Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Natalia Salvadores
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile.
- Escuela de Medicina, Facultad de Medicina y Ciencias de La Salud, Universidad Mayor, Temuco, Chile.
| |
Collapse
|
5
|
Brooker SM, Gonzalez-Latapi P. Biomarkers in Parkinson's Disease. Neurol Clin 2025; 43:229-248. [PMID: 40185520 DOI: 10.1016/j.ncl.2024.12.005] [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
Parkinson's disease (PD) is a leading cause of disability worldwide, and there is a pressing need to develop therapeutics to slow or halt disease progression. The identification of reliable biomarkers of PD at all stages of disease will be a critical step toward optimizing diagnosis and therapeutic development. For PD, biomarkers could serve multiple important functions. There have been significant advances in biomarker development in PD in recent years, and in this review, the authors summarize the current state of the PD biomarker field covering major advances in fluid, tissue, and neuroimaging biomarkers.
Collapse
Affiliation(s)
- Sarah M Brooker
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paulina Gonzalez-Latapi
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
6
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
7
|
Alam MM, Lee SH, Wasim S, Lee SY. PET tracer development for imaging α-synucleinopathies. Arch Pharm Res 2025; 48:333-350. [PMID: 40227383 DOI: 10.1007/s12272-025-01538-0] [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: 10/04/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025]
Abstract
Abnormal α-synuclein aggregation is a key neuropathological hallmark of α-synucleinopathies, such as Parkinson's disease (PD), multiple system atrophy (MSA), and several other neurological disorders, and closely contributes to pathogenesis. The primary characteristics of α-synucleinopathies are selective targeted neurodegeneration and the accumulation of Lewy pathologies. Specifically, α-synuclein positron emission tomography (PET) radiotracers target the fibrillar forms of the protein, thus enhancing early diagnosis and the evaluation of treatment effectiveness for various α-synucleinopathies. Therefore, in vivo detection of α-synuclein aggregates using targeted radiolabeled probes would aid in drug development, early diagnosis, and ongoing disease monitoring. As such, no promising α-synuclein biomarkers suitable for clinical applications have been reported. PET is a valuable non-invasive technique for imaging drug distribution in tissues and receptor occupancy at target sites in living animals and humans. Advances in PET biomarkers have significantly enhanced our understanding of the mechanisms underlying PD. This review summarizes recent ongoing efforts in the development of selective PET tracers for α-synuclein and discusses future perspectives.
Collapse
Affiliation(s)
| | - Sun Hak Lee
- Neuroscience Research Institute, Gachon University, Incheon, 20565, Republic of Korea
| | - Sobia Wasim
- Neuroscience Research Institute, Gachon University, Incheon, 20565, Republic of Korea
| | - Sang-Yoon Lee
- Neuroscience Research Institute, Gachon University, Incheon, 20565, Republic of Korea.
- Department of Neuroscience, College of Medicine, Gachon University, Incheon, 21936, Republic of Korea.
| |
Collapse
|
8
|
Soto C, Mollenhauer B, Hansson O, Kang UJ, Alcalay RN, Standaert D, Trenkwalder C, Marek K, Galasko D, Poston K. Toward a biological definition of neuronal and glial synucleinopathies. Nat Med 2025; 31:396-408. [PMID: 39885358 DOI: 10.1038/s41591-024-03469-7] [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: 05/28/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025]
Abstract
Cerebral accumulation of alpha-synuclein (αSyn) aggregates is the hallmark event in a group of neurodegenerative diseases-collectively called synucleinopathies-which include Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. Currently, these are diagnosed by their clinical symptoms and definitively confirmed postmortem by the presence of αSyn deposits in the brain. Here, we summarize the drawbacks of the current clinical definition of synucleinopathies and outline the rationale for moving toward an earlier, biology-anchored definition of these disorders, with or without the presence of clinical symptoms. We underscore the utility of the αSyn seed amplification assay to detect aggregated αSyn in living patients and to differentiate between neuronal or glial αSyn pathology. We anticipate that a biological definition of synucleinopathies, if well-integrated with the current clinical classifications, will enable further understanding of the disease pathogenesis and contribute to the development of effective, disease-modifying therapies.
Collapse
Affiliation(s)
- Claudio Soto
- Department of Neurology, Mitchell Center for Alzheimer's disease and related brain disorders, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA.
| | - Brit Mollenhauer
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
- Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Un Jung Kang
- Departments of Neurology and Neuroscience & Physiology, Neuroscience Institute, Fresco Institute for Parkinson's Disease and Movement Disorders, Parekh Center for Interdisciplinary Neurology, Grossman School of Medicine, New York University, New York, NY, USA
| | - Roy N Alcalay
- Columbia University Irving Medical Center, New York, NY, USA
- Tel Aviv Sourasky Medical Center, Tel Aviv University School of Medicine, Tel Aviv, Israel
| | - David Standaert
- Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Claudia Trenkwalder
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | - Kenneth Marek
- Institute for Neurodegerative Disorders, New Haven, CT, USA
| | - Douglas Galasko
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, UC San Diego, La Jolla, CA, USA
| | - Kathleen Poston
- Department of Neurology & Neurological Sciences, Stanford Movement Disorders Center, Stanford University, Stanford, CA, USA
| |
Collapse
|
9
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [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: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
10
|
Bruno MK, Dhall R, Duquette A, Haq IU, Honig LS, Lamotte G, Mari Z, McFarland NR, Montaser-Kouhsari L, Rodriguez-Porcel F, Shurer J, Siddiqui J, Spears CC, Wills AMA, Diaz K, Golbe LI. A General Neurologist's Practical Diagnostic Algorithm for Atypical Parkinsonian Disorders: A Consensus Statement. Neurol Clin Pract 2024; 14:e200345. [PMID: 39185098 PMCID: PMC11341009 DOI: 10.1212/cpj.0000000000200345] [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: 11/06/2023] [Accepted: 04/16/2024] [Indexed: 08/27/2024]
Abstract
Purpose of Review The most common four neurodegenerative atypical parkinsonian disorders (APDs) are progressive supranuclear palsy (PSP), multiple system atrophy (MSA), corticobasal syndrome (CBS), and dementia with Lewy bodies (DLB). Their formal diagnostic criteria often require subspecialty experience to implement as designed and all require excluding competing diagnoses without clearly specifying how to do that. Validated diagnostic criteria are not available at all for many of the other common APDs, including normal pressure hydrocephalus (NPH), vascular parkinsonism (VP), or drug-induced parkinsonism (DIP). APDs also include conditions of structural, genetic, vascular, toxic/metabolic, infectious, and autoimmune origin. Their differential diagnosis can be challenging early in the course, if the presentation is atypical, or if a rare or non-neurodegenerative condition is present. This review equips community general neurologists to make an early provisional diagnosis before, or in place of, referral to a tertiary center. Early diagnosis would allay diagnostic uncertainty, allow prompt symptomatic management, provide disease-specific information and support resources, avoid further pointless testing and treatments, and create the possibility of trial referral. Recent Findings We address 64 APDs using one over-arching flow diagram and a series of detailed tables. Most instances of APDs can be diagnosed with a careful history and neurological exam, along with a non-contrast brain MRI. Additional diagnostic tests are rarely needed but are delineated where applicable. Our diagnostic algorithm encourages referral to a tertiary center whenever the general neurologist feels it would be in the patient's best interest. Our algorithm emphasizes that the diagnosis of APDs is an iterative process, refined with the appearance of new diagnostic features, availability of new technology, and advances in scientific understanding of the disorders. Clinicians' proposals for all diagnostic tests for the APDs, including repeat visits, should be discussed with patients and their families to ensure that the potential information to be gained aligns with their larger clinical goals. Summary We designed this differential diagnostic algorithm for the APDs to enhance general neurologists' diagnostic skills and confidence and to help them address the less common or more ambiguous cases.
Collapse
Affiliation(s)
- Michiko K Bruno
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Rohit Dhall
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Antoine Duquette
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Ihtsham U Haq
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Lawrence S Honig
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Guillaume Lamotte
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Zoltan Mari
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Nikolaus R McFarland
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Leila Montaser-Kouhsari
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Federico Rodriguez-Porcel
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Jessica Shurer
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Junaid Siddiqui
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Christopher C Spears
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Anne-Marie A Wills
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Kristophe Diaz
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Lawrence I Golbe
- Neuroscience Institute (MKB), The Queen's Medical Center; Medicine (MKB), University of Hawaii, John A Burns School of Medicine, Honolulu; Neurology (RD), University of Arkansas for Medical Sciences, Little Rock; Service de Neurologie (AD), Département de Médecine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada; Neurology (IUH), University of Miami, FL; Neurology (LSH), Columbia University Irving Medical Center, New York; Neurology (GL), The University of Utah; Neurology (GL), George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Neurology (NRM), University of Florida, Gainesville; Neurology (LM-K), Brigham and Women Hospital and Harvard Medical School, Boston, MA; Neurology (ZM), Johns Hopkins University, Baltimore, MD; Cleveland Clinic Lou Ruvo Center for Brain Health (ZM), Las Vegas, NV; Neurology (FR-P), Medical University of South Carolina, Charleston; CurePSP (J. Shurer, KD, LIG), New York; Neurological Institute (J. Siddiqui), Cleveland Clinic, OH; Neurology (CCS), University of Michigan, Ann Arbor; Neurology (AMW), Massachusetts General Hospital and Harvard Medical School, Boston; and Neurology (LIG), Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
11
|
Mekala S, Wu Y, Li YM. Strategies of positron emission tomography (PET) tracer development for imaging of tau and α-synuclein in neurodegenerative disorders. RSC Med Chem 2024:d4md00576g. [PMID: 39678127 PMCID: PMC11638850 DOI: 10.1039/d4md00576g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, characterized by the presence of extracellular amyloid plaques consisting of β-amyloid peptides (Aβ) and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau (pTau) protein in the brain. Genetic and animal studies strongly indicate that Aβ, tau and neuroinflammation play important roles in the pathogenesis of AD. Several staging models showed that NFTs correlated well with the disease progression. Positron emission tomography (PET) imaging has become a widely used non-invasive technique to image NFTs for early diagnosis of AD. Despite the remarkable progress made over the past few years, tau PET imaging is still challenging due to the nature of tau pathology and the technical aspects of PET imaging. Tau pathology often coexists with other proteinopathies, such as Aβ plaques and α-synuclein aggregates. Distinguishing tau-specific signals from other overlapping pathologies is difficult, especially in the context of AD, where multiple protein aggregates are present, as well as the spectrum of different tau isoforms (3R and 4R) and conformations. Moreover, tracers should ideally have optimal pharmacokinetic properties to penetrate the blood-brain barrier (BBB) while maintaining specificity, low toxicity, low non-specific binding, rapid uptake and clearance from the brain, and formation of no radiolabeled metabolites in the brain. On the other hand, Parkinson's disease (PD) is a progressive neurodegenerative movement disorder characterized by the abnormal accumulations of α-synuclein in neurons. Heterogeneity and the unclear pathogenesis of PD hinder early and accurate diagnosis of the disease for therapeutic development in clinical use. In this review, while referring to existing reviews, we focus on the design strategies and current progress in tau (NFTs) targeting new PET tracers for AD; evolution of non-AD tau targeting PET tracers for applications including progressive supranuclear paralysis (PSP) and corticobasal degeneration (CBD); new PET tracer development for α-synuclein aggregate imaging in PD and giving an outlook for future PET tracer development.
Collapse
Affiliation(s)
- Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| |
Collapse
|
12
|
Barba L, Abu-Rumeileh S, Barthel H, Massa F, Foschi M, Bellomo G, Gaetani L, Thal DR, Parnetti L, Otto M. Clinical and diagnostic implications of Alzheimer's disease copathology in Lewy body disease. Brain 2024; 147:3325-3343. [PMID: 38991041 DOI: 10.1093/brain/awae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024] Open
Abstract
Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with a higher risk of dementia and, consequently, be not rarely misdiagnosed. In this review, we summarize the current understanding of LBD-AD by discussing the synergistic effects of AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment for use in LBD-AD and their possible diagnostic and prognostic values. AD pathology can be predicted in vivo by means of CSF, MRI and PET markers, whereas the most promising technique to date for identifying Lewy pathology in different biological tissues is the α-synuclein seed amplification assay. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity as in pure AD. Implementing the use of blood-based AD biomarkers might allow faster screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account when considering differential diagnoses of dementia syndromes, to allow prognostic evaluation on an individual level, and to guide symptomatic and disease-modifying therapies.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig 04103, Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Matteo Foschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna 48121, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Dietmar R Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Leuven 3001, Belgium
- Department of Pathology, UZ Leuven, Leuven 3000, Belgium
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| |
Collapse
|
13
|
Maddocks GM, Eisenstein M, Soh HT. Biosensors for Parkinson's Disease: Where Are We Now, and Where Do We Need to Go? ACS Sens 2024; 9:4307-4327. [PMID: 39189973 DOI: 10.1021/acssensors.4c00790] [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: 08/28/2024]
Abstract
Parkinson's Disease is the second most common neurological disease in the United States, yet there is no cure, no pinpointed cause, and no definitive diagnostic procedure. Parkinson's is typically diagnosed when patients present with motor symptoms such as slowness of movement and tremors. However, none of these are specific to Parkinson's, and a confident diagnosis of Parkinson's is typically only achieved when 60-80% of dopaminergic neurons are no longer functioning, at which point much of the damage to the brain is irreversible. This Perspective details ongoing efforts and accomplishments in biosensor research with the goal of overcoming these issues for Parkinson's diagnosis and care, with a focus on the potential impact of early diagnosis and associated opportunities to pinpoint a cause and a cure. We critically analyze the strengths and shortcomings of current technologies and discuss the ideal characteristics of a diagnostic technology toolbox to guide future research decisions in this space. Finally, we assess what role biosensors can play in facilitating precision medicine for Parkinson's patients.
Collapse
Affiliation(s)
- Grace M Maddocks
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - M Eisenstein
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - H Tom Soh
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
14
|
Chassé M, Vasdev N. Emerging targets for positron emission tomography imaging in proteinopathies. NPJ IMAGING 2024; 2:30. [PMID: 39185440 PMCID: PMC11338821 DOI: 10.1038/s44303-024-00032-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
Positron emission tomography (PET) imaging of neurodegenerative disease has historically focused on a small number of established targets. The development of selective PET radiotracers for novel biological targets enables new ways to interrogate the neuropathology of proteinopathies and will advance our understanding of neurodegeneration. This perspective aims to highlight recent PET radiotracers developed for five emerging targets in proteinopathies (i.e., mHTT, BACE1, TDP-43, OGA, and CH24H).
Collapse
Affiliation(s)
- Melissa Chassé
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
15
|
Jack CR, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, Molinuevo JL, Okonkwo OC, Pani L, Rafii MS, Scheltens P, Siemers E, Snyder HM, Sperling R, Teunissen CE, Carrillo MC. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement 2024; 20:5143-5169. [PMID: 38934362 PMCID: PMC11350039 DOI: 10.1002/alz.13859] [Citation(s) in RCA: 511] [Impact Index Per Article: 511.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/28/2024]
Abstract
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step-by-step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. HIGHLIGHTS: We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early-changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later-changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Collapse
Affiliation(s)
| | - J. Scott Andrews
- Global Evidence & OutcomesTakeda Pharmaceuticals Company LimitedCambridgeMassachusettsUSA
| | - Thomas G. Beach
- Civin Laboratory for NeuropathologyBanner Sun Health Research InstituteSun CityArizonaUSA
| | - Teresa Buracchio
- Office of NeuroscienceU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Ana Graf
- NovartisNeuroscience Global Drug DevelopmentBaselSwitzerland
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Faculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University Hospital, MalmöLundSweden
| | - Carole Ho
- DevelopmentDenali TherapeuticsSouth San FranciscoCaliforniaUSA
| | - William Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Eric McDade
- Department of NeurologyWashington University St. Louis School of MedicineSt. LouisMissouriUSA
| | - Jose Luis Molinuevo
- Department of Global Clinical Development H. Lundbeck A/SExperimental MedicineCopenhagenDenmark
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin School of MedicineMadisonWisconsinUSA
| | - Luca Pani
- University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of Medicine at the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Philip Scheltens
- Amsterdam University Medical Center (Emeritus)NeurologyAmsterdamthe Netherlands
| | - Eric Siemers
- Clinical ResearchAcumen PharmaceuticalsZionsvilleIndianaUSA
| | - Heather M. Snyder
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| | - Reisa Sperling
- Department of Neurology, Brigham and Women's HospitalMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Department of Laboratory MedicineAmsterdam UMC, Neurochemistry LaboratoryAmsterdamthe Netherlands
| | - Maria C. Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
16
|
Sun X, Badachhape A, Bhandari P, Chin J, Annapragada A, Tanifum E. A dual target molecular magnetic resonance imaging probe for noninvasive profiling of pathologic alpha-synuclein and microgliosis in a mouse model of Parkinson's disease. Front Neurosci 2024; 18:1428736. [PMID: 39114484 PMCID: PMC11303179 DOI: 10.3389/fnins.2024.1428736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is characterized by progressive deposition of alpha-synuclein (α-syn) aggregates in dopaminergic neurons and neuroinflammation. Noninvasive in vivo imaging of α-syn aggregate accumulation and neuroinflammation can elicit the underlying mechanisms involved in disease progression and facilitate the development of effective treatment as well as disease diagnosis and prognosis. Here we present a novel approach to simultaneously profile α-syn aggregation and reactive microgliosis in vivo, by targeting oligomeric α-syn in cerebrospinal fluid with nanoparticle bearing a magnetic resonance imaging (MRI), contrast payload. In this proof-of-concept report we demonstrate, in vitro, that microglia and neuroblastoma cell lines internalize agglomerates formed by cross-linking the nanoparticles with oligomeric α-syn. Delayed in vivo MRI scans following intravenous administration of the nanoparticles in the M83 α-syn transgenic mouse line show statistically significant MR signal enhancement in test mice versus controls. The in vivo data were validated by ex-vivo immunohistochemical analysis which show strong correlation between in vivo MRI signal enhancement, Lewy pathology distribution, and microglia activity in the treated brain tissue. Furthermore, neuronal and microglial cells in brain tissue from treated mice display strong cytosolic signal originating from the nanoparticles, attributed to in vivo cell uptake of nanoparticle/oligomeric α-syn agglomerates.
Collapse
Affiliation(s)
- Xianwei Sun
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Andrew Badachhape
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Prajwal Bhandari
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Ananth Annapragada
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
- Department of Radiology, Texas Children’s Hospital, Houston, TX, United States
| | - Eric Tanifum
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
- Department of Radiology, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
17
|
Suri K, Ramesh M, Bhandari M, Gupta V, Kumar V, Govindaraju T, Murugan NA. Role of Amyloidogenic and Non-Amyloidogenic Protein Spaces in Neurodegenerative Diseases and their Mitigation Using Theranostic Agents. Chembiochem 2024; 25:e202400224. [PMID: 38668376 DOI: 10.1002/cbic.202400224] [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: 03/11/2024] [Revised: 04/23/2024] [Indexed: 06/15/2024]
Abstract
Neurodegenerative diseases (NDDs) refer to a complex heterogeneous group of diseases which are associated with the accumulation of amyloid fibrils or plaques in the brain leading to progressive loss of neuronal functions. Alzheimer's disease is one of the major NDD responsible for 60-80 % of all dementia cases. Currently, there are no curative or disease-reversing/modifying molecules for many of the NDDs except a few such as donepezil, rivastigmine, galantamine, carbidopa and levodopa which treat the disease-associated symptoms. Similarly, there are very few FDA-approved tracers such as flortaucipir (Tauvid) for tau fibril imaging and florbetaben (Neuraceq), flutemetamol (Vizamyl), and florbetapir (Amyvid) for amyloid imaging available for diagnosis. Recent advances in the cryogenic electron microscopy reported distinctly different microstructures for tau fibrils associated with different tauopathies highlighting the possibility to develop tauopathy-specific imaging agents and therapeutics. In addition, it is important to identify the proteins that are associated with disease development and progression to know about their 3D structure to develop various diagnostics, therapeutics and theranostic agents. The current article discusses in detail the disease-associated amyloid and non-amyloid proteins along with their structural insights. We comprehensively discussed various novel proteins associated with NDDs and their implications in disease pathology. In addition, we document various emerging chemical compounds developed for diagnosis and therapy of different NDDs with special emphasis on theranostic agents for better management of NDDs.
Collapse
Affiliation(s)
- Kapali Suri
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Mansi Bhandari
- Department of computer science and engineering, Jamia Hamdard University, Hamdard Nagar, New Delhi, Delhi, 110062
| | - Vishakha Gupta
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Virendra Kumar
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - N Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| |
Collapse
|
18
|
Rose C, Tomas-Grau RH, Zabala B, Michel PP, Brunel JM, Chehín R, Raisman-Vozari R, Ferrié L, Figadère B. C9-Functionalized Doxycycline Analogs as Drug Candidates to Prevent Pathological α-Synuclein Aggregation and Neuroinflammation in Parkinson's Disease Degeneration. ChemMedChem 2024; 19:e202300597. [PMID: 38526011 DOI: 10.1002/cmdc.202300597] [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: 10/31/2023] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Doxycycline, a semi-synthetic tetracycline, is a widely used antibiotic for treating mild-to-moderate infections, including skin problems. However, its anti-inflammatory and antioxidant properties, combined with its ability to interfere with α-synuclein aggregation, make it an attractive candidate for repositioning in Parkinson's disease. Nevertheless, the antibiotic activity of doxycycline restricts its potential use for long-term treatment of Parkinsonian patients. In the search for non-antibiotic tetracyclines that could operate against Parkinson's disease pathomechanisms, eighteen novel doxycycline derivatives were designed. Specifically, the dimethyl-amino group at C4 was reduced, resulting in limited antimicrobial activity, and several coupling reactions were performed at position C9 of the aromatic D ring, this position being one of the most reactive for introducing substituents. Using the Thioflavin-T assay, we found seven compounds were more effective than doxycycline in inhibiting α-synuclein aggregation. Furthermore, two of these derivatives exhibited better anti-inflammatory effects than doxycycline in a culture system of microglial cells used to model Parkinson's disease neuroinflammatory processes. Overall, through structure-activity relationship studies, we identified two newly designed tetracyclines as promising drug candidates for Parkinson's disease treatment.
Collapse
Affiliation(s)
- Clémence Rose
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| | | | - Brenda Zabala
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Patrick Pierre Michel
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Jean-Michel Brunel
- UMR_MD1 Membranes et Cibles Thérapeutiques, U1261 INSERM, Aix-Marseille Université, 13385, Marseille, France
| | - Rosana Chehín
- IMMCA, CONICET-UNT-SIPROSA, Tucumán, 4000, Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Laurent Ferrié
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| |
Collapse
|
19
|
Bonanno F, Saw RS, Bleher D, Papadopoulos I, Bowden GD, Bjerregaard-Andersen K, Windhorst AD, Pichler BJ, Herfert K, Maurer A. Advancing Parkinson's Disease Diagnostics: The Potential of Arylpyrazolethiazole Derivatives for Imaging α-Synuclein Aggregates. ACS OMEGA 2024; 9:24774-24788. [PMID: 38882134 PMCID: PMC11170759 DOI: 10.1021/acsomega.4c01301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024]
Abstract
The development of positron emission tomography (PET) tracers capable of detecting α-synuclein (α-syn) aggregates in vivo would represent a breakthrough for advancing the understanding and enabling the early diagnosis of Parkinson's disease and related disorders. It also holds the potential to assess the efficacy of therapeutic interventions. However, this remains challenging due to different structures of α-syn aggregates, the need for selectivity over other structurally similar amyloid proteins, like amyloid-β (Aβ), which frequently coexist with α-syn pathology, and the low abundance of the target in the brain that requires the development of a high-affinity ligand. To develop a successful PET tracer for the central nervous system (CNS), stringent criteria in terms of polarity and molecular size must also be considered, as the tracer must penetrate the blood-brain barrier and have low nonspecific binding to brain tissue. Here, we report a series of arylpyrazolethiazole (APT) derivatives, rationally designed from a structure-activity relationship study centered on existing ligands for α-syn fibrils, with a particular focus on the selectivity toward α-syn fibrils and control of physicochemical properties suitable for a CNS PET tracer. In vitro competition binding assays performed against [3H]MODAG-001 using recombinant α-syn and Aβ1-42 fibrils revealed APT-13 with an inhibition constant of 27.8 ± 9.7 nM and a selectivity of more than 3.3 fold over Aβ. Radiolabeled [11C]APT-13 demonstrated excellent brain penetration in healthy mice with a peak standardized uptake value of 1.94 ± 0.29 and fast washout from the brain (t 1/2 = 9 ± 1 min). This study highlights the potential of APT-13 as a lead compound for developing PET tracers to detect α-syn aggregates in vivo.
Collapse
Affiliation(s)
- Federica Bonanno
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
| | - Ran Sing Saw
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
| | - Daniel Bleher
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
| | - Ioannis Papadopoulos
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
| | - Gregory D Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| | | | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1085c, 1081 HV Amsterdam, The Netherlands
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, Tübingen 72076, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| |
Collapse
|
20
|
Dickmann CGF, Milicevic Sephton S, Barker RA, Aigbirhio FI. PET Ligands for Imaging Mutant Huntingtin Aggregates: A Case Study in Non-For-Profit Scientific Management. Chembiochem 2024; 25:e202400152. [PMID: 38695673 DOI: 10.1002/cbic.202400152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/02/2024] [Indexed: 06/13/2024]
Abstract
Positron emission tomography imaging of misfolded proteins with high-affinity and selective radioligands has played a vital role in expanding our knowledge of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. The pathogenesis of Huntington's disease, a CAG trinucleotide repeat disorder, is similarly linked to the presence of protein fibrils formed from mutant huntingtin (mHTT) protein. Development of mHTT fibril-specific radioligands has been limited by the lack of structural knowledge around mHTT and a dearth of available hit compounds for medicinal chemistry refinement. Over the past decade, the CHDI Foundation, a non-for-profit scientific management organisation has orchestrated a large-scale screen of small molecules to identify high affinity ligands of mHTT, with lead compounds now reaching clinical maturity. Here we describe the mHTT radioligands developed to date and opportunities for further improvement of this radiotracer class.
Collapse
Affiliation(s)
- Catherine G F Dickmann
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Selena Milicevic Sephton
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Toyohara J, Vugts D, Kiss OC, Todde S, Li XG, Liu Z, Yang Z, Gillings N, Cazzola E, Szymanski W, Meulen NVD, Reilly R, Taddei C, Schirrmacher R, Li Z, Lagebo YJ, Bentaleb N, Souza Albernaz MD, Lapi S, Ramogida C, Mukherjee A, Ajenjo J, Deuther-Conrad W, Bourdeau C. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2024; 9:42. [PMID: 38753262 PMCID: PMC11098975 DOI: 10.1186/s41181-024-00268-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development. MAIN BODY This selection of highlights provides commentary on 24 different topics selected by each coauthoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field in many aspects.
Collapse
Affiliation(s)
- Jun Toyohara
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Danielle Vugts
- Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Oliver C Kiss
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
| | - Sergio Todde
- University of Milano-Bicoccia, Tecnomed Foundation, Monza, Italy
| | - Xiang-Guo Li
- Turku PET Centre and Department of Chemistry, and InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | | | - Zhi Yang
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Nic Gillings
- Copenhagen University Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | - Naoual Bentaleb
- National Center for Nuclear Energy, Science and Technology-CNESTEN, Rabat, Morocco
| | - Marta de Souza Albernaz
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suzanne Lapi
- University of Alabama at Birmingham, Birmingham, USA
| | - Caterina Ramogida
- Simon Fraser University, Burnaby, Vancouver, Canada
- TRIUMF, Burnaby, Vancouver, Canada
| | - Archana Mukherjee
- Bhabha Atomic Research Center and Homi Bhabha National Institute, Mumbai, India
| | - Javier Ajenjo
- Molecular Imaging Program at Stanford (MIPS), Dept of Radiology, School of Medicine, Stanford University, Stanford, CA), USA
| | - Winnie Deuther-Conrad
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Leipzig, Germany
| | | |
Collapse
|
22
|
Liu M, Wang Z, Shang H. Multiple system atrophy: an update and emerging directions of biomarkers and clinical trials. J Neurol 2024; 271:2324-2344. [PMID: 38483626 PMCID: PMC11055738 DOI: 10.1007/s00415-024-12269-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 04/28/2024]
Abstract
Multiple system atrophy is a rare, debilitating, adult-onset neurodegenerative disorder that manifests clinically as a diverse combination of parkinsonism, cerebellar ataxia, and autonomic dysfunction. It is pathologically characterized by oligodendroglial cytoplasmic inclusions containing abnormally aggregated α-synuclein. According to the updated Movement Disorder Society diagnostic criteria for multiple system atrophy, the diagnosis of clinically established multiple system atrophy requires the manifestation of autonomic dysfunction in combination with poorly levo-dopa responsive parkinsonism and/or cerebellar syndrome. Although symptomatic management of multiple system atrophy can substantially improve quality of life, therapeutic benefits are often limited, ephemeral, and they fail to modify the disease progression and eradicate underlying causes. Consequently, effective breakthrough treatments that target the causes of disease are needed. Numerous preclinical and clinical studies are currently focusing on a set of hallmarks of neurodegenerative diseases to slow or halt the progression of multiple system atrophy: pathological protein aggregation, synaptic dysfunction, aberrant proteostasis, neuronal inflammation, and neuronal cell death. Meanwhile, specific biomarkers and measurements with higher specificity and sensitivity are being developed for the diagnosis of multiple system atrophy, particularly for early detection of the disease. More intriguingly, a growing number of new disease-modifying candidates, which can be used to design multi-targeted, personalized treatment in patients, are being investigated, notwithstanding the failure of most previous attempts.
Collapse
Affiliation(s)
- Min Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Zhiyao Wang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
23
|
Pees A, Tong J, Birudaraju S, Munot YS, Liang SH, Saturnino Guarino D, Mach RH, Mathis CA, Vasdev N. Development of Pyridothiophene Compounds for PET Imaging of α-Synuclein. Chemistry 2024; 30:e202303921. [PMID: 38354298 PMCID: PMC12189351 DOI: 10.1002/chem.202303921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/16/2024]
Abstract
Aggregated α-synuclein (α-syn) protein is a pathological hallmark of Parkinson's disease (PD) and Lewy body dementia (LBD). Development of positron emission tomography (PET) radiotracers to image α-syn aggregates has been a longstanding goal. This work explores the suitability of a pyridothiophene scaffold for α-syn PET radiotracers, where 47 derivatives of a potent pyridothiophene (asyn-44; Kd=1.85 nM) were synthesized and screened against [3H]asyn-44 in competitive binding assays using post-mortem PD brain homogenates. Equilibrium inhibition constant (Ki) values of the most potent compounds were determined, of which three had Ki's in the lower nanomolar range (12-15 nM). An autoradiography study confirmed that [3H]asyn-44 is promising for imaging brain sections from multiple system atrophy and PD donors. Fluorine-18 labelled asyn-44 was synthesized in 6±2 % radiochemical yield (decay-corrected, n=5) with a molar activity of 263±121 GBq/μmol. Preliminary PET imaging of [18F]asyn-44 in rats showed high initial brain uptake (>1.5 standardized uptake value (SUV)), moderate washout (~0.4 SUV at 60 min), and low variability. Radiometabolite analysis showed 60-80 % parent tracer in the brain after 30 and 60 mins. While [18F]asyn-44 displayed good in vitro properties and acceptable brain uptake, troublesome radiometabolites precluded further PET imaging studies. The synthesis and in vitro evaluation of additional pyridothiophene derivatives are underway, with the goal of attaining improved affinity and metabolic stability.
Collapse
Affiliation(s)
- Anna Pees
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
| | | | | | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30322, United States
| | - Dinahlee Saturnino Guarino
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 1012, 231 S. 34th Street, Philadelphia, Pennsylvania, 19104-6323, United States
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 1012, 231 S. 34th Street, Philadelphia, Pennsylvania, 19104-6323, United States
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213, United States
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T-1R8, Canada
| |
Collapse
|
24
|
Bisi N, Pinzi L, Rastelli G, Tonali N. Early Diagnosis of Neurodegenerative Diseases: What Has Been Undertaken to Promote the Transition from PET to Fluorescence Tracers. Molecules 2024; 29:722. [PMID: 38338465 PMCID: PMC10856728 DOI: 10.3390/molecules29030722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's Disease (AD) and Parkinson's Disease (PD) represent two among the most frequent neurodegenerative diseases worldwide. A common hallmark of these pathologies is the misfolding and consequent aggregation of amyloid proteins into soluble oligomers and insoluble β-sheet-rich fibrils, which ultimately lead to neurotoxicity and cell death. After a hundred years of research on the subject, this is the only reliable histopathological feature in our hands. Since AD and PD are diagnosed only once neuronal death and the first symptoms have appeared, the early detection of these diseases is currently impossible. At present, there is no effective drug available, and patients are left with symptomatic and inconclusive therapies. Several reasons could be associated with the lack of effective therapeutic treatments. One of the most important factors is the lack of selective probes capable of detecting, as early as possible, the most toxic amyloid species involved in the onset of these pathologies. In this regard, chemical probes able to detect and distinguish among different amyloid aggregates are urgently needed. In this article, we will review and put into perspective results from ex vivo and in vivo studies performed on compounds specifically interacting with such early species. Following a general overview on the three different amyloid proteins leading to insoluble β-sheet-rich amyloid deposits (amyloid β1-42 peptide, Tau, and α-synuclein), a list of the advantages and disadvantages of the approaches employed to date is discussed, with particular attention paid to the translation of fluorescence imaging into clinical applications. Furthermore, we also discuss how the progress achieved in detecting the amyloids of one neurodegenerative disease could be leveraged for research into another amyloidosis. As evidenced by a critical analysis of the state of the art, substantial work still needs to be conducted. Indeed, the early diagnosis of neurodegenerative diseases is a priority, and we believe that this review could be a useful tool for better investigating this field.
Collapse
Affiliation(s)
- Nicolò Bisi
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Nicolò Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| |
Collapse
|
25
|
Ndayisaba A, Pitaro AT, Willett AS, Jones KA, de Gusmao CM, Olsen AL, Kim J, Rissanen E, Woods JK, Srinivasan SR, Nagy A, Nagy A, Mesidor M, Cicero S, Patel V, Oakley DH, Tuncali I, Taglieri-Noble K, Clark EC, Paulson J, Krolewski RC, Ho GP, Hung AY, Wills AM, Hayes MT, Macmore JP, Warren L, Bower PG, Langer CB, Kellerman LR, Humphreys CW, Glanz BI, Dielubanza EJ, Frosch MP, Freeman RL, Gibbons CH, Stefanova N, Chitnis T, Weiner HL, Scherzer CR, Scholz SW, Vuzman D, Cox LM, Wenning G, Schmahmann JD, Gupta AS, Novak P, Young GS, Feany MB, Singhal T, Khurana V. Clinical Trial-Ready Patient Cohorts for Multiple System Atrophy: Coupling Biospecimen and iPSC Banking to Longitudinal Deep-Phenotyping. CEREBELLUM (LONDON, ENGLAND) 2024; 23:31-51. [PMID: 36190676 PMCID: PMC9527378 DOI: 10.1007/s12311-022-01471-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease of unknown etiology characterized by widespread aggregation of the protein alpha-synuclein in neurons and glia. Its orphan status, biological relationship to Parkinson's disease (PD), and rapid progression have sparked interest in drug development. One significant obstacle to therapeutics is disease heterogeneity. Here, we share our process of developing a clinical trial-ready cohort of MSA patients (69 patients in 2 years) within an outpatient clinical setting, and recruiting 20 of these patients into a longitudinal "n-of-few" clinical trial paradigm. First, we deeply phenotype our patients with clinical scales (UMSARS, BARS, MoCA, NMSS, and UPSIT) and tests designed to establish early differential diagnosis (including volumetric MRI, FDG-PET, MIBG scan, polysomnography, genetic testing, autonomic function tests, skin biopsy) or disease activity (PBR06-TSPO). Second, we longitudinally collect biospecimens (blood, CSF, stool) and clinical, biometric, and imaging data to generate antecedent disease-progression scores. Third, in our Mass General Brigham SCiN study (stem cells in neurodegeneration), we generate induced pluripotent stem cell (iPSC) models from our patients, matched to biospecimens, including postmortem brain. We present 38 iPSC lines derived from MSA patients and relevant disease controls (spinocerebellar ataxia and PD, including alpha-synuclein triplication cases), 22 matched to whole-genome sequenced postmortem brain. iPSC models may facilitate matching patients to appropriate therapies, particularly in heterogeneous diseases for which patient-specific biology may elude animal models. We anticipate that deeply phenotyped and genotyped patient cohorts matched to cellular models will increase the likelihood of success in clinical trials for MSA.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Ariana T Pitaro
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Andrew S Willett
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Kristie A Jones
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Claudio Melo de Gusmao
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Abby L Olsen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jisoo Kim
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Eero Rissanen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jared K Woods
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sharan R Srinivasan
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI , 48103, USA
| | - Anna Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Amanda Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Merlyne Mesidor
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Steven Cicero
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Viharkumar Patel
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Idil Tuncali
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Katherine Taglieri-Noble
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Emily C Clark
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jordan Paulson
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Richard C Krolewski
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gary P Ho
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Albert Y Hung
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anne-Marie Wills
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael T Hayes
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jason P Macmore
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | | | - Pamela G Bower
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Carol B Langer
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Lawrence R Kellerman
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Christopher W Humphreys
- Department of Pulmonary, Sleep and Critical Care Medicine, Salem Hospital, MassGeneral Brigham, Salem, MA, 01970, USA
| | - Bonnie I Glanz
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Elodi J Dielubanza
- Department of Urology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Roy L Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Christopher H Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Nadia Stefanova
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Tanuja Chitnis
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Howard L Weiner
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Clemens R Scherzer
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Sonja W Scholz
- Laboratory of Neurogenetics, Disorders and Stroke, National Institute of Neurological, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, 21287, USA
| | - Dana Vuzman
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Laura M Cox
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gregor Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anoopum S Gupta
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Peter Novak
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Geoffrey S Young
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tarun Singhal
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Vikram Khurana
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA.
| |
Collapse
|
26
|
Pang Z, Cravatt BF, Ye L. Deciphering Drug Targets and Actions with Single-Cell and Spatial Resolution. Annu Rev Pharmacol Toxicol 2024; 64:507-526. [PMID: 37722721 DOI: 10.1146/annurev-pharmtox-033123-123610] [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] [Indexed: 09/20/2023]
Abstract
Recent advances in chemical, molecular, and genetic approaches have provided us with an unprecedented capacity to identify drug-target interactions across the whole proteome and genome. Meanwhile, rapid developments of single-cell and spatial omics technologies are revolutionizing our understanding of the molecular architecture of biological systems. However, a significant gap remains in how we align our understanding of drug actions, traditionally based on molecular affinities, with the in vivo cellular and spatial tissue heterogeneity revealed by these newer techniques. Here, we review state-of-the-art methods for profiling drug-target interactions and emerging multiomics tools to delineate the tissue heterogeneity at single-cell resolution. Highlighting the recent technical advances enabling high-resolution, multiplexable in situ small-molecule drug imaging (clearing-assisted tissue click chemistry, or CATCH), we foresee the integration of single-cell and spatial omics platforms, data, and concepts into the future framework of defining and understanding in vivo drug-target interactions and mechanisms of actions.
Collapse
Affiliation(s)
- Zhengyuan Pang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA;
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA;
| | - Li Ye
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
27
|
Taha A, Alassi A, Gjedde A, Wong DF. Transforming Neurology and Psychiatry: Organ-specific PET Instrumentation and Clinical Applications. PET Clin 2024; 19:95-103. [PMID: 37813719 DOI: 10.1016/j.cpet.2023.06.002] [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: 10/11/2023]
Abstract
PET technology has immense potential for furthering understanding of the brain and associated disorders, including advancements in high-resolution tomographs and hybrid imaging modalities. Novel radiotracers targeting specific neurotransmitter systems and molecular markers provide opportunities to unveil intricate mechanisms underlying neurologic and psychiatric conditions. As PET imaging techniques and analysis methods continue to be refined, the field is poised to make significant contributions to personalized medicine for more targeted and effective interventions. PET instrumentation has advanced the fields of neurology and psychiatry, providing insights into pathophysiology and development of effective treatments.
Collapse
Affiliation(s)
- Ahmed Taha
- Mallinckrodt Institute of Radiology, Washington University in St Louis, Saint Louis, MO, USA
| | - Amer Alassi
- Mallinckrodt Institute of Radiology, Washington University in St Louis, Saint Louis, MO, USA
| | - Albert Gjedde
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Denmark; Department of Neuroscience, University of Copenhagen, Denmark
| | - Dean F Wong
- Mallinckrodt Institute of Radiology, Departments of Radiology, Psychiatry, Neurology, Neuroscience, Washington University in St Louis, Saint Louis, MO, USA.
| |
Collapse
|
28
|
Savoie FA, Arpin DJ, Vaillancourt DE. Magnetic Resonance Imaging and Nuclear Imaging of Parkinsonian Disorders: Where do we go from here? Curr Neuropharmacol 2024; 22:1583-1605. [PMID: 37533246 PMCID: PMC11284713 DOI: 10.2174/1570159x21666230801140648] [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: 08/10/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 08/04/2023] Open
Abstract
Parkinsonian disorders are a heterogeneous group of incurable neurodegenerative diseases that significantly reduce quality of life and constitute a substantial economic burden. Nuclear imaging (NI) and magnetic resonance imaging (MRI) have played and continue to play a key role in research aimed at understanding and monitoring these disorders. MRI is cheaper, more accessible, nonirradiating, and better at measuring biological structures and hemodynamics than NI. NI, on the other hand, can track molecular processes, which may be crucial for the development of efficient diseasemodifying therapies. Given the strengths and weaknesses of NI and MRI, how can they best be applied to Parkinsonism research going forward? This review aims to examine the effectiveness of NI and MRI in three areas of Parkinsonism research (differential diagnosis, prodromal disease identification, and disease monitoring) to highlight where they can be most impactful. Based on the available literature, MRI can assist with differential diagnosis, prodromal disease identification, and disease monitoring as well as NI. However, more work is needed, to confirm the value of MRI for monitoring prodromal disease and predicting phenoconversion. Although NI can complement or be a substitute for MRI in all the areas covered in this review, we believe that its most meaningful impact will emerge once reliable Parkinsonian proteinopathy tracers become available. Future work in tracer development and high-field imaging will continue to influence the landscape for NI and MRI.
Collapse
Affiliation(s)
- Félix-Antoine Savoie
- Department of Applied Physiology and Kinesiology, Laboratory for Rehabilitation Neuroscience, University of Florida, Gainesville, FL, USA
| | - David J. Arpin
- Department of Applied Physiology and Kinesiology, Laboratory for Rehabilitation Neuroscience, University of Florida, Gainesville, FL, USA
| | - David E. Vaillancourt
- Department of Applied Physiology and Kinesiology, Laboratory for Rehabilitation Neuroscience, University of Florida, Gainesville, FL, USA
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
Uzuegbunam BC, Li J, Paslawski W, Weber W, Svenningsson P, Ågren H, Hooshyar Yousefi B. In Silico and In Vitro Study towards the Rational Design of 4,4'-Disarylbisthiazoles as a Selective α-Synucleinopathy Biomarker. Int J Mol Sci 2023; 24:16445. [PMID: 38003637 PMCID: PMC10671360 DOI: 10.3390/ijms242216445] [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: 10/17/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
The α-synucleinopathies are a group of neurodegenerative diseases characterized by the deposition of α-synuclein aggregates (α-syn) in the brain. Currently, there is no suitable tracer to enable a definitive early diagnosis of these diseases. We reported candidates based on 4,4'-disarylbisthiazole (DABTA) scaffold with a high affinity towards α-syn and excellent selectivity over Aβ and tau fibrils. Based on prior in silico studies, a focused library of 23 halogen-containing and O-methylated DABTAs was prepared. The DABTAs were synthesized via a modified two-step Hantzsch thiazole synthesis, characterized, and used in competitive binding assays against [3H]PiB and [3H]DCVJ. The DABTAs were obtained with an overall chemical yield of 15-71%, and showed a calculated lipophilicity of 2.5-5.7. The ligands demonstrated an excellent affinity to α-syn with both [3H]PiB and [3H]DCVJ: Ki 0.1-4.9 nM and up to 20-3900-fold selectivity over Aβ and tau fibrils. It could be concluded that in silico simulation is useful for the rational design of a new generation of DABTAs. Further investigation of the leads in the next step is encouraged: radiolabeling of the ligands with radioisotopes such as fluorine-18 or carbon-11 for in vivo, ex vivo, and translational research and for further in vitro experiments on human-derived protein aggregates.
Collapse
Affiliation(s)
- Bright C. Uzuegbunam
- Department of Nuclear Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Junhao Li
- Department of Physics and Astronomy, Uppsala University, 751 20 Uppsala, Sweden
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Hans Ågren
- Department of Physics and Astronomy, Uppsala University, 751 20 Uppsala, Sweden
| | | |
Collapse
|
30
|
Tao Y, Xia W, Zhao Q, Xiang H, Han C, Zhang S, Gu W, Tang W, Li Y, Tan L, Li D, Liu C. Structural mechanism for specific binding of chemical compounds to amyloid fibrils. Nat Chem Biol 2023; 19:1235-1245. [PMID: 37400537 DOI: 10.1038/s41589-023-01370-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/26/2023] [Indexed: 07/05/2023]
Abstract
Amyloid fibril is an important pharmaceutical target for diagnostic and therapeutic treatment of neurodegenerative diseases. However, rational design of chemical compounds that interact with amyloid fibrils is unachievable due to the lack of mechanistic understanding of the ligand-fibril interaction. Here we used cryoelectron microscopy to survey the amyloid fibril-binding mechanism of a series of compounds including classic dyes, (pre)clinical imaging tracers and newly identified binders from high-throughput screening. We obtained clear densities of several compounds in complex with an α-synuclein fibril. These structures unveil the basic mechanism of the ligand-fibril interaction, which exhibits remarkable difference from the canonical ligand-protein interaction. In addition, we discovered a druggable pocket that is also conserved in the ex vivo α-synuclein fibrils from multiple system atrophy. Collectively, these findings expand our knowledge of protein-ligand interaction in the amyloid fibril state, which will enable rational design of amyloid binders in a medicinally beneficial way.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Huaijiang Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chao Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Gu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenjun Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
31
|
Kim HY, Chia WK, Hsieh CJ, Guarino DS, Graham TJA, Lengyel-Zhand Z, Schneider M, Tomita C, Lougee MG, Kim HJ, Pagar VV, Lee H, Hou C, Garcia BA, Petersson EJ, O’Shea J, Kotzbauer PT, Mathis CA, Lee VMY, Luk KC, Mach RH. A Novel Brain PET Radiotracer for Imaging Alpha Synuclein Fibrils in Multiple System Atrophy. J Med Chem 2023; 66:12185-12202. [PMID: 37651366 PMCID: PMC10617560 DOI: 10.1021/acs.jmedchem.3c00779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Abnormal α-synuclein (α-syn) aggregation characterizes α-synucleinopathies, including Parkinson's disease (PD) and multiple system atrophy (MSA). However, no suitable positron emission tomography (PET) radiotracer for imaging α-syn in PD and MSA exists currently. Our structure-activity relationship studies identified 4-methoxy-N-(4-(3-(pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octan-8-yl)phenyl)benzamide (4i) as a PET radiotracer candidate for imaging α-syn. In vitro assays revealed high binding of 4i to recombinant α-syn fibrils (inhibition constant (Ki) = 6.1 nM) and low affinity for amyloid beta (Aβ) fibrils in Alzheimer's disease (AD) homogenates. However, [3H]4i also exhibited high specific binding to AD, progressive supranuclear palsy, and corticobasal degeneration tissues as well as PD and MSA tissues, suggesting notable affinity to tau. Nevertheless, the specific binding to pathologic α-syn aggregates in MSA post-mortem brain tissues was significantly higher than in PD tissues. This finding demonstrated the potential use of [11C]4i as a PET tracer for imaging α-syn in MSA patients. Nonhuman primate PET studies confirmed good brain uptake and rapid washout for [11C]4i.
Collapse
Affiliation(s)
- Ho Young Kim
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Wai Kit Chia
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Chia-Ju Hsieh
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Dinahlee Saturnino Guarino
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Thomas J. A. Graham
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Zsofia Lengyel-Zhand
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Mark Schneider
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Cosette Tomita
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Marshall G. Lougee
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Hee Jong Kim
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6303, USA
| | - Vinayak V. Pagar
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Catherine Hou
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6303, USA
| | - E. James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Jennifer O’Shea
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110-1010, USA
| | - Paul T. Kotzbauer
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110-1010, USA
| | - Chester A. Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research, Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-2676, USA
| | - Kelvin C. Luk
- Center for Neurodegenerative Disease Research, Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-2676, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| |
Collapse
|
32
|
Di Nanni A, Saw RS, Battisti UM, Bowden GD, Boeckermann A, Bjerregaard-Andersen K, Pichler BJ, Herfert K, Herth MM, Maurer A. A Fluorescent Probe as a Lead Compound for a Selective α-Synuclein PET Tracer: Development of a Library of 2-Styrylbenzothiazoles and Biological Evaluation of [ 18F]PFSB and [ 18F]MFSB. ACS OMEGA 2023; 8:31450-31467. [PMID: 37663501 PMCID: PMC10468942 DOI: 10.1021/acsomega.3c04292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023]
Abstract
A method to detect and quantify aggregated α-synuclein (αSYN) fibrils in vivo would drastically impact the current understanding of multiple neurodegenerative diseases, revolutionizing their diagnosis and treatment. Several efforts have produced promising scaffolds, but a notable challenge has hampered the establishment of a clinically successful αSYN positron emission tomography (PET) tracer: the requirement of high selectivity over the other misfolded proteins amyloid β (Aβ) and tau. By designing and screening a library of 2-styrylbenzothiazoles based on the selective fluorescent probe RB1, this study aimed at developing a selective αSYN PET tracer. [3H]PiB competition binding assays identified PFSB (Ki = 25.4 ± 2.3 nM) and its less lipophilic analogue MFSB, which exhibited enhanced affinity to αSYN (Ki = 10.3 ± 4.7 nM) and preserved selectivity over Aβ. The two lead compounds were labeled with fluorine-18 and evaluated using in vitro autoradiography on human brain slices, where they demonstrated up to 4-fold increased specific binding in MSA cases compared to the corresponding control, reasonably reflecting selective binding to αSYN pathology. In vivo PET imaging showed [18F]MFSB successfully crosses the blood-brain barrier (BBB) and is taken up in the brain (SUV = 1.79 ± 0.02). Although its pharmacokinetic profile raises the need for additional structural optimization, [18F]MFSB represents a critical step forward in the development of a successful αSYN PET tracer by overcoming the major challenge of αSYN/Aβ selectivity.
Collapse
Affiliation(s)
- Adriana Di Nanni
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
| | - Ran Sing Saw
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
| | - Umberto M. Battisti
- Department
of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Gregory D. Bowden
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University Tübingen, 72076 Tübingen, Germany
| | - Adam Boeckermann
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
| | | | - Bernd J. Pichler
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University Tübingen, 72076 Tübingen, Germany
| | - Kristina Herfert
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
| | - Matthias M. Herth
- Department
of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
| | - Andreas Maurer
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 72076 Tübingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
33
|
Houssein NJ, Henriksen AC, Hejl AM, Marner L. Diagnostic accuracy of cerebral [ 18F]FDG PET in atypical parkinsonism. EJNMMI Res 2023; 13:74. [PMID: 37572162 PMCID: PMC10423182 DOI: 10.1186/s13550-023-01025-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/03/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Atypical parkinsonism (AP) often presents with Parkinson's symptoms but has a much worse long-term prognosis. The diagnosis is presently based on clinical criteria, but a cerebral positron emission tomography (PET) scan with [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG) may assist in the diagnosis of AP such as multiple system atrophy (MSA), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Lewy body dementia (DLB). Only few studies have evaluated the sensitivity and specificity of [18F]FDG PET for separating the diseases in a mixed patient population, which we aim to assess in a retrospective material. RESULTS We identified 156 patients referred for a cerebral [18F]FDG PET for suspicion of AP during 2017-2019. The [18F]FDG PET was analysed by a nuclear medicine specialist blinded to clinical information but with access to dopamine transporter imaging. The reference standard was the follow-up clinical diagnosis (follow-up: 6-72 months). The overall accuracy for correct classification was 74%. Classification sensitivity (95% confidence interval, CI) and specificity (95% CI) for MSA (n = 20) were 1.00 (0.83-1.00) and 0.91 (0.85-0.95), for DLB/Parkinson with dementia (PDD) (n = 26) were 0.81 (0.61-0.93) and 0.97 (0.92-0.99) and for CBD/PSP (n = 68) were 0.62 (0.49-0.73) and 0.97 (0.90-0.99). CONCLUSIONS Our results support the additional use of [18F]FDG PET for the clinical diagnosis of AP with moderate to high sensitivity and specificity. Use of [18F]FDG PET may be beneficial for prognosis and supportive treatment of the patients and useful for future clinical treatment trials.
Collapse
Affiliation(s)
- Naba Jawad Houssein
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark.
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Alexander Cuculiza Henriksen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
| | - Anne-Mette Hejl
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Lisbeth Marner
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Xiang J, Tao Y, Xia Y, Luo S, Zhao Q, Li B, Zhang X, Sun Y, Xia W, Zhang M, Kang SS, Ahn EH, Liu X, Xie F, Guan Y, Yang JJ, Bu L, Wu S, Wang X, Cao X, Liu C, Zhang Z, Li D, Ye K. Development of an α-synuclein positron emission tomography tracer for imaging synucleinopathies. Cell 2023; 186:3350-3367.e19. [PMID: 37421950 PMCID: PMC10527432 DOI: 10.1016/j.cell.2023.06.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 07/10/2023]
Abstract
Synucleinopathies are characterized by the accumulation of α-synuclein (α-Syn) aggregates in the brain. Positron emission tomography (PET) imaging of synucleinopathies requires radiopharmaceuticals that selectively bind α-Syn deposits. We report the identification of a brain permeable and rapid washout PET tracer [18F]-F0502B, which shows high binding affinity for α-Syn, but not for Aβ or Tau fibrils, and preferential binding to α-Syn aggregates in the brain sections. Employing several cycles of counter screenings with in vitro fibrils, intraneuronal aggregates, and neurodegenerative disease brain sections from several mice models and human subjects, [18F]-F0502B images α-Syn deposits in the brains of mouse and non-human primate PD models. We further determined the atomic structure of the α-Syn fibril-F0502B complex by cryo-EM and revealed parallel diagonal stacking of F0502B on the fibril surface through an intense noncovalent bonding network via inter-ligand interactions. Therefore, [18F]-F0502B is a promising lead compound for imaging aggregated α-Syn in synucleinopathies.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Biomedical Sciences, School of Medicine, JiangHan University, #8, Sanjiaohu Rd., Wuhan 430056, China
| | - Shilin Luo
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Yunpeng Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Mingming Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eun-Hee Ahn
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Lihong Bu
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shengxi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
35
|
Wan L, Zhu S, Chen Z, Qiu R, Tang B, Jiang H. Multidimensional biomarkers for multiple system atrophy: an update and future directions. Transl Neurodegener 2023; 12:38. [PMID: 37501056 PMCID: PMC10375766 DOI: 10.1186/s40035-023-00370-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple system atrophy (MSA) is a fatal progressive neurodegenerative disease. Biomarkers are urgently required for MSA to improve the diagnostic and prognostic accuracy in clinic and facilitate the development and monitoring of disease-modifying therapies. In recent years, significant research efforts have been made in exploring multidimensional biomarkers for MSA. However, currently few biomarkers are available in clinic. In this review, we systematically summarize the latest advances in multidimensional biomarkers for MSA, including biomarkers in fluids, tissues and gut microbiota as well as imaging biomarkers. Future directions for exploration of novel biomarkers and promotion of implementation in clinic are also discussed.
Collapse
Affiliation(s)
- Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China.
| |
Collapse
|
36
|
Zhang S, Dong H, Bian J, Li D, Liu C. Targeting amyloid proteins for clinical diagnosis of neurodegenerative diseases. FUNDAMENTAL RESEARCH 2023; 3:505-519. [PMID: 38933553 PMCID: PMC11197785 DOI: 10.1016/j.fmre.2022.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Abnormal aggregation and accumulation of pathological amyloid proteins such as amyloid-β, Tau, and α-synuclein play key pathological roles and serve as histological hallmarks in different neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, various post-translational modifications (PTMs) have been identified on pathological amyloid proteins and are subjected to change during disease progression. Given the central role of amyloid proteins in NDs, tremendous efforts have been made to develop amyloid-targeting strategies for clinical diagnosis and molecular classification of NDs. In this review, we summarize two major strategies for targeting amyloid aggregates, with a focus on the trials in AD diagnosis. The first strategy is a positron emission tomography (PET) scan of protein aggregation in the brain. We mainly focus on introducing the development of small-molecule PET tracers for specifically recognizing pathological amyloid fibrils. The second strategy is the detection of PTM biomarkers on amyloid proteins in cerebrospinal fluid and plasma. We discuss the pathological roles of different PTMs in diseases and how we can use the PTM profile of amyloid proteins for clinical diagnosis. Finally, we point out the potential technical challenges of these two strategies, and outline other potential strategies, as well as a combination of multiple strategies, for molecular diagnosis of NDs.
Collapse
Affiliation(s)
- Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Dong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Bian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
- Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
37
|
Pakula RJ, Scott PJH. Applications of radiolabeled antibodies in neuroscience and neuro-oncology. J Labelled Comp Radiopharm 2023; 66:269-285. [PMID: 37322805 DOI: 10.1002/jlcr.4049] [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: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Positron emission tomography (PET) is a powerful tool in medicine and drug development, allowing for non-invasive imaging and quantitation of biological processes in live organisms. Targets are often probed with small molecules, but antibody-based PET is expanding because of many benefits, including ease of design of new antibodies toward targets, as well as the very strong affinities that can be expected. Application of antibodies to PET imaging of targets in the central nervous system (CNS) is a particularly nascent field, but one with tremendous potential. In this review, we discuss the growth of PET in imaging of CNS targets, present the promises and progress in antibody-based CNS PET, explore challenges faced by the field, and discuss questions that this promising approach will need to answer moving forward for imaging and perhaps even radiotherapy.
Collapse
Affiliation(s)
- Ryan J Pakula
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Noguchi-Shinohara M, Ono K. The Mechanisms of the Roles of α-Synuclein, Amyloid-β, and Tau Protein in the Lewy Body Diseases: Pathogenesis, Early Detection, and Therapeutics. Int J Mol Sci 2023; 24:10215. [PMID: 37373401 DOI: 10.3390/ijms241210215] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Lewy body diseases (LBD) are pathologically defined as the accumulation of Lewy bodies composed of an aggregation of α-synuclein (αSyn). In LBD, not only the sole aggregation of αSyn but also the co-aggregation of amyloidogenic proteins, such as amyloid-β (Aβ) and tau, has been reported. In this review, the pathophysiology of co-aggregation of αSyn, Aβ, and tau protein and the advancement in imaging and fluid biomarkers that can detect αSyn and co-occurring Aβ and/or tau pathologies are discussed. Additionally, the αSyn-targeted disease-modifying therapies in clinical trials are summarized.
Collapse
Affiliation(s)
- Moeko Noguchi-Shinohara
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| |
Collapse
|
39
|
Björk L, Klingstedt T, Nilsson KPR. Thiophene-Based Ligands: Design, Synthesis and Their Utilization for Optical Assignment of Polymorphic-Disease-Associated Protein Aggregates. Chembiochem 2023; 24:e202300044. [PMID: 36891883 PMCID: PMC10404026 DOI: 10.1002/cbic.202300044] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/10/2023]
Abstract
The development of ligands for detecting protein aggregates is of great interest, as these aggregated proteinaceous species are the pathological hallmarks of several devastating diseases, including Alzheimer's disease. In this regard, thiophene-based ligands have emerged as powerful tools for fluorescent assessment of these pathological entities. The intrinsic conformationally sensitive photophysical properties of poly- and oligothiophenes have allowed optical assignment of disease-associated protein aggregates in tissue sections, as well as real-time in vivo imaging of protein deposits. Herein, we recount the chemical evolution of different generations of thiophene-based ligands, and exemplify their use for the optical distinction of polymorphic protein aggregates. Furthermore, the chemical determinants for achieving a superior fluorescent thiophene-based ligand, as well as the next generation of thiophene-based ligands targeting distinct aggregated species are described. Finally, the directions for future research into the chemical design of thiophene-based ligands that can aid in resolving the scientific challenges around protein aggregation diseases are discussed.
Collapse
Affiliation(s)
- Linnea Björk
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
40
|
Di Nanni A, Saw RS, Bowden GD, Bidesi NSR, Bjerregaard-Andersen K, Korat Š, Herth MM, Pichler BJ, Herfert K, Maurer A. The Structural Combination of SIL and MODAG Scaffolds Fails to Enhance Binding to α-Synuclein but Reveals Promising Affinity to Amyloid β. Molecules 2023; 28:molecules28104001. [PMID: 37241742 DOI: 10.3390/molecules28104001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
A technique to image α-synuclein (αSYN) fibrils in vivo is an unmet scientific and clinical need that would represent a transformative tool in the understanding, diagnosis, and treatment of various neurodegenerative diseases. Several classes of compounds have shown promising results as potential PET tracers, but no candidate has yet exhibited the affinity and selectivity required to reach clinical application. We hypothesized that the application of the rational drug design technique of molecular hybridization to two promising lead scaffolds could enhance the binding to αSYN up to the fulfillment of those requirements. By combining the structures of SIL and MODAG tracers, we developed a library of diarylpyrazoles (DAPs). In vitro evaluation through competition assays against [3H]SIL26 and [3H]MODAG-001 showed the novel hybrid scaffold to have preferential binding affinity for amyloid β (Aβ) over αSYN fibrils. A ring-opening modification on the phenothiazine building block to produce analogs with increased three-dimensional flexibility did not result in an improved αSYN binding but a complete loss of competition, as well as a significant reduction in Aβ affinity. The combination of the phenothiazine and the 3,5-diphenylpyrazole scaffolds into DAP hybrids did not generate an enhanced αSYN PET tracer lead compound. Instead, these efforts identified a scaffold for promising Aβ ligands that may be relevant to the treatment and monitoring of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Adriana Di Nanni
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Ran Sing Saw
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Gregory D Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Natasha S R Bidesi
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Kaare Bjerregaard-Andersen
- Department of Antibody Engineering and Biochemistry, H. Lundbeck A/S, Ottiliavej 9, 2500 Copenhagen, Denmark
| | - Špela Korat
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117 Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1117 Amsterdam, The Netherlands
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
41
|
Jiang Y, Lin Y, Krishnaswamy S, Pan R, Wu Q, Sandusky-Beltran LA, Liu M, Kuo MH, Kong XP, Congdon EE, Sigurdsson EM. Single-domain antibody-based noninvasive in vivo imaging of α-synuclein or tau pathology. SCIENCE ADVANCES 2023; 9:eadf3775. [PMID: 37163602 PMCID: PMC10171817 DOI: 10.1126/sciadv.adf3775] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/11/2023] [Indexed: 05/12/2023]
Abstract
Intracellular deposition of α-synuclein and tau are hallmarks of synucleinopathies and tauopathies, respectively. Recently, several dye-based imaging probes with selectivity for tau aggregates have been developed, but suitable imaging biomarkers for synucleinopathies are still unavailable. Detection of both of these aggregates early in the disease process may allow for prophylactic therapies before functional impairments have manifested, highlighting the importance of developing specific imaging probes for these lesions. In contrast to the β sheet dyes, single-domain antibodies, found in camelids and a few other species, are highly specific, and their small size allows better brain entry and distribution than whole antibodies. Here, we have developed such imaging ligands via phage display libraries derived from llamas immunized with α-synuclein and tau preparations, respectively. These probes allow noninvasive and specific in vivo imaging of α-synuclein versus tau pathology in mice, with the brain signal correlating strongly with lesion burden. These small antibody derivatives have great potential for in vivo diagnosis of these diseases.
Collapse
Affiliation(s)
- Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Yan Lin
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Senthilkumar Krishnaswamy
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Qian Wu
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Leslie A. Sandusky-Beltran
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Erin E. Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Einar M. Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| |
Collapse
|
42
|
Cory MB, Jones CM, Shaffer KD, Venkatesh Y, Giannakoulias S, Perez RM, Lougee MG, Hummingbird E, Pagar VV, Hurley CM, Li A, Mach RH, Kohli RM, Petersson EJ. FRETing about the details: Case studies in the use of a genetically encoded fluorescent amino acid for distance-dependent energy transfer. Protein Sci 2023; 32:e4633. [PMID: 36974585 PMCID: PMC10108435 DOI: 10.1002/pro.4633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Förster resonance energy transfer (FRET) is a valuable method for monitoring protein conformation and biomolecular interactions. Intrinsically fluorescent amino acids that can be genetically encoded, such as acridonylalanine (Acd), are particularly useful for FRET studies. However, quantitative interpretation of FRET data to derive distance information requires careful use of controls and consideration of photophysical effects. Here we present two case studies illustrating how Acd can be used in FRET experiments to study small molecule induced conformational changes and multicomponent biomolecular complexes.
Collapse
Affiliation(s)
- Michael B. Cory
- Graduate Group in Biochemistry and BiophysicsPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Chloe M. Jones
- Graduate Group in Biochemistry and BiophysicsPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Kyle D. Shaffer
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Yarra Venkatesh
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Sam Giannakoulias
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Ryann M. Perez
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Marshall G. Lougee
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Eshe Hummingbird
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Vinayak V. Pagar
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Christina M. Hurley
- Graduate Group in Biochemistry and BiophysicsPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Allen Li
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Robert H. Mach
- Department of RadiologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - Rahul M. Kohli
- Department of Biochemistry and BiophysicsPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
- Department of MedicinePerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| | - E. James Petersson
- Department of ChemistrySchool of Arts and Sciences, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
- Department of Biochemistry and BiophysicsPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania19104USA
| |
Collapse
|
43
|
Orlovskaya VV, Fedorova OS, Viktorov NB, Vaulina DD, Krasikova RN. One-Pot Radiosynthesis of [18F]Anle138b—5-(3-Bromophenyl)-3-(6-[18F]fluorobenzo[d][1,3]dioxol-5-yl)-1H-pyrazole—A Potential PET Radiotracer Targeting α-Synuclein Aggregates. Molecules 2023; 28:molecules28062732. [PMID: 36985703 PMCID: PMC10052605 DOI: 10.3390/molecules28062732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
Availability of PET imaging radiotracers targeting α-synuclein aggregates is important for early diagnosis of Parkinson’s disease and related α-synucleinopathies, as well as for the development of new therapeutics. Derived from a pyrazole backbone, 11C-labelled derivatives of anle138b (3-(1,3-benzodioxol-5-yl)-5-(3-bromophenyl)-1H-pyrazole)—an inhibitor of α-synuclein and prion protein oligomerization—are currently in active development as the candidates for PET imaging α-syn aggregates. This work outlines the synthesis of a radiotracer based on the original structure of anle138b, labelled with fluorine-18 isotope, eminently suitable for PET imaging due to half-life and decay energy characteristics (97% β+ decay, 109.7 min half-life, and 635 keV positron energy). A three-step radiosynthesis was developed starting from 6-[18F]fluoropiperonal (6-[18F]FP) that was prepared using (piperonyl)(phenyl)iodonium bromide as a labelling precursor. The obtained 6-[18F]FP was used directly in the condensation reaction with tosylhydrazide followed by 1,3-cycloaddition of the intermediate with 3′-bromophenylacetylene eliminating any midway without any intermediate purifications. This one-pot approach allowed the complete synthesis of [18F]anle138b within 105 min with RCY of 15 ± 3% (n = 3) and Am in the range of 32–78 GBq/µmol. The [18F]fluoride processing and synthesis were performed in a custom-built semi-automated module, but the method can be implemented in all the modern automated platforms. While there is definitely space for further optimization, the procedure developed is well suited for preclinical studies of this novel radiotracer in animal models and/or cell cultures.
Collapse
Affiliation(s)
- Viktoriya V. Orlovskaya
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Science, 197376 St. Petersburg, Russia
| | - Olga S. Fedorova
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Science, 197376 St. Petersburg, Russia
| | - Nikolai B. Viktorov
- St. Petersburg State Technological Institute, Technical University, 190013 St. Petersburg, Russia
| | - Daria D. Vaulina
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Science, 197376 St. Petersburg, Russia
| | - Raisa N. Krasikova
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Science, 197376 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
44
|
Kanasuwan A, Deuther-Conrad W, Chongruchiroj S, Sarasamkan J, Chotipanich C, Vajragupta O, Arunrungvichian K. Selective α 3β 4 Nicotinic Acetylcholine Receptor Ligand as a Potential Tracer for Drug Addiction. Int J Mol Sci 2023; 24:ijms24043614. [PMID: 36835028 PMCID: PMC9959096 DOI: 10.3390/ijms24043614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
α3β4 Nicotinic acetylcholine receptor (nAChR) has been recognized as an emerging biomarker for the early detection of drug addiction. Herein, α3β4 nAChR ligands were designed and synthesized to improve the binding affinity and selectivity of two lead compounds, (S)-QND8 and (S)-T2, for the development of an α3β4 nAChR tracer. The structural modification was achieved by retaining the key features and expanding the molecular structure with a benzyloxy group to increase the lipophilicity for blood-brain barrier penetration and to extend the ligand-receptor interaction. The preserved key features are a fluorine atom for radiotracer development and a p-hydroxyl motif for ligand-receptor binding affinity. Four (R)- and (S)-quinuclidine-triazole (AK1-AK4) were synthesized and the binding affinity, together with selectivity to α3β4 nAChR subtype, were determined by competitive radioligand binding assay using [3H]epibatidine as a radioligand. Among all modified compounds, AK3 showed the highest binding affinity and selectivity to α3β4 nAChR with a Ki value of 3.18 nM, comparable to (S)-QND8 and (S)-T2 and 3069-fold higher affinity to α3β4 nAChR in comparison to α7 nAChR. The α3β4 nAChR selectivity of AK3 was considerably higher than those of (S)-QND8 (11.8-fold) and (S)-T2 (294-fold). AK3 was shown to be a promising α3β4 nAChR tracer for further development as a radiotracer for drug addiction.
Collapse
Affiliation(s)
- Apinan Kanasuwan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Bangkok 10400, Thailand
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaengphet 6 Rd., Bangkok 10210, Thailand
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany
| | - Sumet Chongruchiroj
- Department of Microbiology, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Bangkok 10400, Thailand
| | - Jiradanai Sarasamkan
- Department of Radiology, Faculty of Medicine, Khon Kaen University, 123 Mittraphap Rd., Khon Kaen 40002, Thailand
| | - Chanisa Chotipanich
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, 906 Kamphaengphet 6 Rd., Bangkok 10210, Thailand
| | - Opa Vajragupta
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Rd., Bangkok 10330, Thailand
| | - Kuntarat Arunrungvichian
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Rd., Bangkok 10400, Thailand
- Correspondence:
| |
Collapse
|
45
|
Norris SA, Perlmutter JS. The Future of Functional Neuroimaging in Parkinsonism: Prove It! JAMA Neurol 2023; 80:121-122. [PMID: 36374495 PMCID: PMC10645147 DOI: 10.1001/jamaneurol.2022.3978] [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] [Indexed: 11/16/2022]
Abstract
This Viewpoint discusses the current lack of evidence of functional neuroimaging affecting patient outcomes in parkinsonian conditions.
Collapse
Affiliation(s)
- Scott A Norris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Joel S Perlmutter
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
46
|
Lenka A, Jankovic J. How should future clinical trials be designed in the search for disease-modifying therapies for Parkinson's disease? Expert Rev Neurother 2023; 23:107-122. [PMID: 36803618 DOI: 10.1080/14737175.2023.2177535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
INTRODUCTION Although there has been substantial progress in research and innovations in symptomatic treatments, similar success has not been achieved in disease-modifying therapy (DMT) for Parkinson's disease (PD). Considering the enormous motor, psychosocial and financial burden associated with PD, safe and effective DMT is of paramount importance. AREAS COVERED One of the reasons for the lack of progress in DMT for PD is poor or inappropriate design of clinical trials. In the first part of the article, the authors focus on the plausible reasons why the previous trials have failed and in the latter part, they provide their perspectives on future DMT trials. EXPERT OPINION There are several potential reasons why previous trials have failed, including broad clinical and etiopathogenic heterogeneity of PD, poor definition and documentation of target engagement, lack of appropriate biomarkers and outcome measures, and short duration of follow-up. To address these deficiencies, future trials may consider- (i) a more customized approach to select the most suitable participants and therapeutic approaches, (ii) explore combination therapies that would target multiple pathogenetic mechanisms, and (iii) moving beyond targeting only motor symptoms to also assessing non-motor features of PD in well-designed longitudinal studies.
Collapse
Affiliation(s)
- Abhishek Lenka
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Application of Artificial Intelligence in Image Processing of Neurodegenerative Disorders: A Review Study. Neuromodulation 2023. [DOI: 10.5812/ipmn-134223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
: Neurodegenerative diseases can make life difficult and lead to death in many cases. They also can be difficult, time-consuming, and costly to diagnose with enough accuracy/certainty. Artificial intelligence (AI) has shown promise in tackling some of the challenges present in medical imaging and is anticipated to become a crucial tool in health care applications in the near future. In particular, deep learning methods have displayed great performance in various subfields of image processing, including but not limited to image segmentation, image synthesis, and image reconstruction. In this paper, many state-of-the-art applications of deep learning models in image processing were reviewed.
Collapse
|
48
|
Raval NR, Madsen CA, Shalgunov V, Nasser A, Battisti UM, Beaman EE, Juhl M, Jørgensen LM, Herth MM, Hansen HD, Plavén-Sigray P, Knudsen GM. Evaluation of the α-synuclein PET radiotracer (d 3)-[ 11C]MODAG-001 in pigs. Nucl Med Biol 2022; 114-115:42-48. [PMID: 36095921 DOI: 10.1016/j.nucmedbio.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND A positron emission tomography (PET) radiotracer to neuroimage α-synuclein aggregates would be a crucial addition for early diagnosis and treatment development in disorders such as Parkinson's disease, where elevated aggregate levels are a histopathological hallmark. The radiotracer (d3)-[11C]MODAG-001 has recently shown promise for visualization of α-synuclein pre-formed fibrils (α-PFF) in rodents. We here test the radiotracer in a pig model where proteins are intracerebrally injected immediately before scanning. Four pigs were injected in one hemisphere with 150 μg α-PFF, and in the other hemisphere, either 75 μg α-PFF or human brain homogenate from either dementia with Lewy bodies (DLB) or Alzheimer's disease (AD) was injected. All pigs underwent one or two (d3)-[11C]MODAG-001 PET scans, quantified with the non-invasive Logan graphical analysis using the occipital cortex as a reference region. RESULTS The α-PFF and AD homogenate injected brain regions had high uptake of (d3)-[11C]MODAG-001 compared to the occipital cortex or cerebellum. BPND values in 150 μg α-PFF injected regions was 0.78, and in the AD homogenate injected regions was 0.73. By contrast, the DLB homogenate injected region did not differ in uptake and clearance compared to the reference regions. The time-activity curves and BPND values in the 150 μg and 75 μg injected regions of α-PFFs show a dose-dependent effect, and the PET signal could be blocked by pretreatment with unlabeled MODAG-001. CONCLUSION We find that both α-PFF and AD brain homogenates give rise to increased binding of (d3)-[11C]MODAG-001 when injected into the pig brain. Despite its limited specificity for cerebral α-synuclein pathology, (d3)-[11C]MODAG-001 shows promise as a lead tracer for future radiotracer development.
Collapse
Affiliation(s)
- Nakul Ravi Raval
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Clara Aabye Madsen
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Arafat Nasser
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Umberto Maria Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emily Eufaula Beaman
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Morten Juhl
- Cardiology Stem Cell Centre, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Louise Møller Jørgensen
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Copenhagen Spine Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Hanne Demant Hansen
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Pontus Plavén-Sigray
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Gitte Moos Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
49
|
Kaide S, Watanabe H, Iikuni S, Hasegawa M, Ono M. Synthesis and Evaluation of 18F-Labeled Chalcone Analogue for Detection of α-Synuclein Aggregates in the Brain Using the Mouse Model. ACS Chem Neurosci 2022; 13:2982-2990. [PMID: 36197745 DOI: 10.1021/acschemneuro.2c00473] [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] [Indexed: 01/20/2023] Open
Abstract
In the brains of patients with synucleinopathies such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, α-synuclein (α-syn) aggregates deposit abnormally to induce neurodegeneration, although the mechanism is unclear. Thus, in vivo imaging studies targeting α-syn aggregates have attracted much attention to guide medical intervention against synucleinopathy. In our previous study, a chalcone analogue, [125I]PHNP-3, functioned as a feasible probe in terms of α-syn binding in vitro; however, it did not migrate to the mouse brain, and further improvement of brain uptake was required. In the present study, we designed and synthesized two novel 18F-labeled chalcone analogues, [18F]FHCL-1 and [18F]FHCL-2, using a central nervous system multiparameter optimization (CNS MPO) algorithm with the aim of improving blood-brain barrier permeation in the mouse brain. Then, we evaluated their utility for in vivo imaging of α-syn aggregates using a mouse model. In the competitive inhibition assay, both chalcone analogues exhibited high binding affinity for α-syn aggregates (Ki = 2.6 and 3.4 nM, respectively), while no marked amyloid β (Aβ)-binding was observed. The 18F-labeling reaction was successfully performed. In a biodistribution experiment, brain uptake of both chalcone analogues in normal mice (2.09 and 2.40% injected dose/gram (% ID/g) at 2 min postinjection, respectively) was higher than that of [125I]PHNP-3, suggesting that the introduction of 18F into the chalcone analogue led to an improvement in brain uptake in mice while maintaining favorable binding ability for α-syn aggregates. Furthermore, in an ex vivo autoradiography experiment, [18F]FHCL-2 showed the feasibility of the detection of α-syn aggregates in the mouse brain in vivo. These preclinical studies demonstrated the validity of the design of α-syn-targeting probes based on the CNS MPO score and the possibility of in vivo imaging of α-syn aggregates in a mouse model using 18F-labeled chalcone analogues.
Collapse
Affiliation(s)
- Sho Kaide
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
50
|
Nakagawa K, Watanabe H, Kaide S, Ono M. Structure-Activity Relationships of Styrylquinoline and Styrylquinoxaline Derivatives as α-Synuclein Imaging Probes. ACS Med Chem Lett 2022; 13:1598-1605. [PMID: 36262393 PMCID: PMC9575165 DOI: 10.1021/acsmedchemlett.2c00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Synucleinopathies are characterized by the deposition of α-synuclein (α-syn) aggregates before the onset of clinical symptoms. Therefore, in vivo imaging of α-syn may contribute to early diagnosis of these diseases and has attracted much attention in recent years. However, no clinically useful probes have been reported. In the present study, 16 quinoline/quinoxaline derivatives with different styryl and fluorine groups were evaluated in order to develop α-syn imaging probes. Among them, SQ3, which is a quinoline analogue with a p-(dimethylamino)styryl group and fluoroethoxy group at the 2- and 7- positions of the skeleton, displayed moderate selectivity for α-syn aggregates over β-amyloid (Aβ) aggregates (K i = 230 nM), while maintaining high binding affinity for α-syn aggregates (K i = 39.3 nM). In a biodistribution study, [18F]SQ3 exhibited high uptake (2.08% ID/g at 2 min after intravenous injection) into a normal mouse brain. Taken together, we demonstrate that [18F]SQ3 has basic properties as a lead compound for the development of a useful α-syn imaging probe.
Collapse
Affiliation(s)
- Kohei Nakagawa
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Sho Kaide
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional
Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|