1
|
Qu N, Song K, Ji Y, Liu M, Chen L, Lee RJ, Teng L. Albumin Nanoparticle-Based Drug Delivery Systems. Int J Nanomedicine 2024; 19:6945-6980. [PMID: 39005962 PMCID: PMC11246635 DOI: 10.2147/ijn.s467876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/28/2024] [Indexed: 07/16/2024] Open
Abstract
Nanoparticle-based systems are extensively investigated for drug delivery. Among others, with superior biocompatibility and enhanced targeting capacity, albumin appears to be a promising carrier for drug delivery. Albumin nanoparticles are highly favored in many disease therapies, as they have the proper chemical groups for modification, cell-binding sites for cell adhesion, and affinity to protein drugs for nanocomplex generation. Herein, this review summarizes the recent fabrication techniques, modification strategies, and application of albumin nanoparticles. We first discuss various albumin nanoparticle fabrication methods, from both pros and cons. Then, we provide a comprehensive introduction to the modification section, including organic albumin nanoparticles, metal albumin nanoparticles, inorganic albumin nanoparticles, and albumin nanoparticle-based hybrids. We finally bring further perspectives on albumin nanoparticles used for various critical diseases.
Collapse
Affiliation(s)
- Na Qu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Ke Song
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, the Netherlands
| | - Yating Ji
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Mingxia Liu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Lijiang Chen
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Yantai, 264000, People's Republic of China
| |
Collapse
|
2
|
Warmenhoven N, Salvadó G, Janelidze S, Mattsson-Carlgren N, Bali D, Dolado AO, Kolb H, Triana-Baltzer G, Barthélemy NR, Schindler SE, Aschenbrenner AJ, Raji CA, Benzinger TL, Morris JC, Ibanez L, Timsina J, Cruchaga C, Bateman RJ, Ashton N, Arslan B, Zetterberg H, Blennow K, Pichet Binette A, Hansson O. A Comprehensive Head-to-Head Comparison of Key Plasma Phosphorylated Tau 217 Biomarker Tests. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.02.24309629. [PMID: 39006421 PMCID: PMC11245081 DOI: 10.1101/2024.07.02.24309629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Plasma phosphorylated-tau 217 (p-tau217) is currently the most promising biomarkers for reliable detection of Alzheimer's disease (AD) pathology. Various p-tau217 assays have been developed, but their relative performance is unclear. We compared key plasma p-tau217 tests using cross-sectional and longitudinal measures of amyloid-β (Aβ)-PET, tau-PET, and cognition as outcomes, and benchmarked them against cerebrospinal fluid (CSF) biomarker tests. Samples from 998 individuals (mean[range] age 68.5[20.0-92.5], 53% female) from the Swedish BioFINDER-2 cohort were analyzed. Plasma p-tau217 was measured with mass spectrometry (MS) assays (the ratio between phosphorylated and non-phosphorylated [%p-tau217WashU]and ptau217WashU) as well as with immunoassays (p-tau217Lilly, p-tau217Janssen, p-tau217ALZpath). CSF biomarkers included p-tau217Lilly, and the FDA-approved p-tau181/Aβ42Elecsys and p-tau181Elecsys. All plasma p-tau217 tests exhibited high ability to detect abnormal Aβ-PET (AUC range: 0.91-0.96) and tau-PET (AUC range: 0.94-0.97). Plasma %p-tau217WashU had the highest performance, with significantly higher AUCs than all the immunoassays (P diff<0.007). For detecting Aβ-PET status, %p-tau217WashU had an accuracy of 0.93 (immunoassays: 0.83-0.88), sensitivity of 91% (immunoassays: 84-87%), and a specificity of 94% (immunoassays: 85-89%). Among immunoassays, p-tau217Lilly and plasma p-tau217ALZpath had higher AUCs than plasma p-tau217Janssen for Aβ-PET status (P diff<0.006), and p-tau217Lilly outperformed plasma p-tau217ALZpath for tau-PET status (P diff=0.025). Plasma %p-tau217WashU exhibited higher associations with all PET load outcomes compared to immunoassays; baseline Aβ-PET load (R2: 0.72; immunoassays: 0.47-0.58; Pdiff<0.001), baseline tau-PET load (R2: 0.51; immunoassays: 0.38-0.45; Pdiff<0.001), longitudinal Aβ-PET load (R2: 0.53; immunoassays: 0.31-0.38; Pdiff<0.001) and longitudinal tau-PET load (R2: 0.50; immunoassays: 0.35-0.43; Pdiff<0.014). Among immunoassays, plasma p-tau217Lilly was more strongly associated with Aβ-PET load than plasma p-tau217Janssen (P diff<0.020) and with tau-PET load than both plasma p-tau217Janssen and plasma p-tau217ALZpath (all P diff<0.010). Plasma %p-tau217 also correlated more strongly with baseline cognition (Mini-Mental State Examination[MMSE]) than all immunoassays (R2 %p-tau217WashU: 0.33; immunoassays: 0.27-0.30; P diff<0.024). The main results were replicated in an external cohort from Washington University in St Louis (n =219). Finally, p-tau217Nulisa showed similar performance to other immunoassays in subsets of both cohorts. In summary, both MS- and immunoassay-based p-tau217 tests generally perform well in identifying Aβ-PET, tau-PET, and cognitive abnormalities, but %p-tau217WashU performed significantly better than all the examined immunoassays. Plasma %p-tau217 may be considered as a stand-alone confirmatory test for AD pathology, while some immunoassays might be better suited as triage tests where positive results are confirmed with a second test.
Collapse
Affiliation(s)
- Noëlle Warmenhoven
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Anna Orduña Dolado
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Johnson and Johnson Innovative Medicine, San Diego, CA, USA
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Johnson and Johnson Innovative Medicine, San Diego, CA, USA
| | - Nicolas R. Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Cyrus A. Raji
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO, USA
- Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University St. Louis, MO, USA
| | - Randall J. Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Smith AM, Obuchowski NA, Foster NL, Klein G, Mozley PD, Lammertsma AA, Wahl RL, Sunderland JJ, Vanderheyden JL, Benzinger TLS, Kinahan PE, Wong DF, Perlman ES, Minoshima S, Matthews D. The RSNA QIBA Profile for Amyloid PET as an Imaging Biomarker for Cerebral Amyloid Quantification. J Nucl Med 2023; 64:294-303. [PMID: 36137760 PMCID: PMC9902844 DOI: 10.2967/jnumed.122.264031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 02/04/2023] Open
Abstract
A standardized approach to acquiring amyloid PET images increases their value as disease and drug response biomarkers. Most 18F PET amyloid brain scans often are assessed only visually (per regulatory labels), with a binary decision indicating the presence or absence of Alzheimer disease amyloid pathology. Minimizing technical variance allows precise, quantitative SUV ratios (SUVRs) for early detection of β-amyloid plaques and allows the effectiveness of antiamyloid treatments to be assessed with serial studies. Methods: The Quantitative Imaging Biomarkers Alliance amyloid PET biomarker committee developed and validated a profile to characterize and reduce the variability of SUVRs, increasing statistical power for these assessments. Results: On achieving conformance, sites can justify a claim that brain amyloid burden reflected by the SUVR is measurable to a within-subject coefficient of variation of no more than 1.94% when the same radiopharmaceutical, scanner, acquisition, and analysis protocols are used. Conclusion: This overview explains the claim, requirements, barriers, and potential future developments of the profile to achieve precision in clinical and research amyloid PET imaging.
Collapse
Affiliation(s)
- Anne M Smith
- Siemens Medical Solutions USA, Inc., Knoxville, Tennessee;
| | | | - Norman L Foster
- Department of Neurology, University of Utah, Salt Lake City, Utah
| | | | - P David Mozley
- Weill Medical College of Cornell University, New York, New York
| | - Adriaan A Lammertsma
- Amsterdam Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University in Saint Louis, St. Louis, Missouri
| | - John J Sunderland
- Division of Nuclear Medicine, Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University in Saint Louis, St. Louis, Missouri
| | - Paul E Kinahan
- Department of Radiology, School of Medicine, University of Washington, Seattle, Washington
| | - Dean F Wong
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah; and
| | | |
Collapse
|
4
|
Matsuoka K, Takado Y, Tagai K, Kubota M, Sano Y, Takahata K, Ono M, Seki C, Matsumoto H, Endo H, Shinotoh H, Sahara Y, Obata T, Near J, Kawamura K, Zhang MR, Suhara T, Shimada H, Higuchi M. Two pathways differentially linking tau depositions, oxidative stress, and neuronal loss to apathetic phenotypes in progressive supranuclear palsy. J Neurol Sci 2023; 444:120514. [PMID: 36473346 DOI: 10.1016/j.jns.2022.120514] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022]
Abstract
Patients with progressive supranuclear palsy (PSP) frequently exhibit apathy but the neuropathological processes leading to this phenotype remain elusive. We aimed to examine the involvement of tau protein depositions, oxidative stress (OS), and neuronal loss in the apathetic manifestation of PSP. Twenty patients with PSP and twenty-three healthy controls were enrolled. Tau depositions and brain volumes were evaluated via positron-emission tomography (PET) using a specific probe, 18F-PM-PBB3, and magnetic resonance imaging, respectively. Glutathione (GSH) levels in the anterior and posterior cingulate cortices were quantified by magnetic resonance spectroscopy. Tau pathologies were observed in the subcortical and cortical structures of the patient brains. The angular gyrus exhibited a positive correlation between tau accumulations and apathy scale (AS). Although PSP cases did not show GSH level alterations compared with healthy controls, GSH levels in posterior cingulate cortex were correlated with AS and tau depositions in the angular gyrus. Marked atrophy was observed in subcortical areas, and gray matter volumes in the inferior frontal gyrus and anterior cingulate cortex were positively correlated with AS but showed no correlation with tau depositions and GSH levels. Path analysis highlighted synergistic contributions of tau pathologies and GSH reductions in the posterior cortex to AS, in parallel with associations of gray matter atrophy in the anterior cortex with AS. Apathetic phenotypes may arise from PET-visible tau aggregation and OS compromising the neural circuit resilience in the posterior cortex, along with neuronal loss, with neither PET-detectable tau pathologies nor OS in the anterior cortex.
Collapse
Affiliation(s)
- Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan; Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan.
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasunori Sano
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hideki Matsumoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan; Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Tokyo, Japan
| | - Hironobu Endo
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hitoshi Shinotoh
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan; Neurology Clinic, Chiba, Chiba, Japan
| | - Yasuka Sahara
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takayuki Obata
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Jamie Near
- Douglas Mental Health University Institute and Department of Psychiatry, McGill University, Quebec City, Canada
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan; Department of Functional Neurology & Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
5
|
Tau PET imaging in progressive supranuclear palsy: a systematic review and meta-analysis. J Neurol 2023; 270:2451-2467. [PMID: 36633672 DOI: 10.1007/s00415-022-11556-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVES To evaluate the difference of tau burden between patients with progressive supranuclear palsy (PSP) and healthy controls (HCs) or other neurodegenerative diseases using tau-positron emission tomography (PET) imaging. METHODS A systematic search on PubMed, Embase, and Web of Science databases was performed for tau-PET studies in PSP patients, up to April 1, 2022. Standardized mean differences (SMDs) of tau tracer uptake were calculated using random-effects models. Subgroup analysis based on the type of tau tracers, meta-regression, and sensitivity analysis were conducted. RESULTS Twenty-seven studies comprising 553 PSP, 626 HCs, and 406 other neurodegenerative diseases were included. Compared with HCs, PSP patients showed elevated tau binding in basal ganglia, midbrain, dentate nucleus, cerebellar white matter, and frontal lobe with decreasing SMD (SMD: 0.390-1.698). Compared with Parkinson's disease patients, increased tau binding was identified in the midbrain, basal ganglia, dentate nucleus, and frontal and parietal lobe in PSP patients with decreasing SMD (SMD: 0.503-1.853). PSP patients showed higher tau binding in the subthalamic nucleus (SMD = 1.351) and globus pallidus (SMD = 1.000), and lower binding in the cortex and parahippocampal gyrus than Alzheimer's disease patients (SMD: - 2.976 to - 1.018). PSP patients showed higher midbrain tau binding than multiple system atrophy patients (SMD = 1.269). CONCLUSION Tau PET imaging indicates different topography of tau deposition between PSP patients and HCs or other neurodegenerative disorders. The affinity and selectivity of tracers for 4R-tau and the off-target binding of tracers should be considered when interpreting the results.
Collapse
|
6
|
Cui J, Gong K, Guo N, Kim K, Liu H, Li Q. Unsupervised PET logan parametric image estimation using conditional deep image prior. Med Image Anal 2022; 80:102519. [PMID: 35767910 DOI: 10.1016/j.media.2022.102519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
Recently, deep learning-based denoising methods have been gradually used for PET images denoising and have shown great achievements. Among these methods, one interesting framework is conditional deep image prior (CDIP) which is an unsupervised method that does not need prior training or a large number of training pairs. In this work, we combined CDIP with Logan parametric image estimation to generate high-quality parametric images. In our method, the kinetic model is the Logan reference tissue model that can avoid arterial sampling. The neural network was utilized to represent the images of Logan slope and intercept. The patient's computed tomography (CT) image or magnetic resonance (MR) image was used as the network input to provide anatomical information. The optimization function was constructed and solved by the alternating direction method of multipliers (ADMM) algorithm. Both simulation and clinical patient datasets demonstrated that the proposed method could generate parametric images with more detailed structures. Quantification results showed that the proposed method results had higher contrast-to-noise (CNR) improvement ratios (PET/CT datasets: 62.25%±29.93%; striatum of brain PET datasets : 129.51%±32.13%, thalamus of brain PET datasets: 128.24%±31.18%) than Gaussian filtered results (PET/CT datasets: 23.33%±18.63%; striatum of brain PET datasets: 74.71%±8.71%, thalamus of brain PET datasets: 73.02%±9.34%) and nonlocal mean (NLM) denoised results (PET/CT datasets: 37.55%±26.56%; striatum of brain PET datasets: 100.89%±16.13%, thalamus of brain PET datasets: 103.59%±16.37%).
Collapse
Affiliation(s)
- Jianan Cui
- The State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; The Center for Advanced Medical Computing and Analysis, Massachusetts General Hospital/Harvard Medical School, Boston MA 02114, USA
| | - Kuang Gong
- The Center for Advanced Medical Computing and Analysis, Massachusetts General Hospital/Harvard Medical School, Boston MA 02114, USA
| | - Ning Guo
- The Center for Advanced Medical Computing and Analysis, Massachusetts General Hospital/Harvard Medical School, Boston MA 02114, USA
| | - Kyungsang Kim
- The Center for Advanced Medical Computing and Analysis, Massachusetts General Hospital/Harvard Medical School, Boston MA 02114, USA
| | - Huafeng Liu
- The State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China; Jiaxing Key Laboratory of Photonic Sensing and Intelligent Imaging, Jiaxing, Zhejiang 314000, China; Intelligent Optics and Photonics Research Center, Jiaxing Research Institute, Zhejiang University, Zhejiang 314000, China.
| | - Quanzheng Li
- The Center for Advanced Medical Computing and Analysis, Massachusetts General Hospital/Harvard Medical School, Boston MA 02114, USA.
| |
Collapse
|
7
|
Song M, Beyer L, Kaiser L, Barthel H, van Eimeren T, Marek K, Nitschmann A, Scheifele M, Palleis C, Respondek G, Kern M, Biechele G, Hammes J, Bischof G, Barbe M, Onur Ö, Jessen F, Saur D, Schroeter ML, Rumpf JJ, Rullmann M, Schildan A, Patt M, Neumaier B, Barret O, Madonia J, Russell DS, Stephens AW, Mueller A, Roeber S, Herms J, Bötzel K, Danek A, Levin J, Classen J, Höglinger GU, Bartenstein P, Villemagne V, Drzezga A, Seibyl J, Sabri O, Boening G, Ziegler S, Brendel M. Binding characteristics of [ 18F]PI-2620 distinguish the clinically predicted tau isoform in different tauopathies by PET. J Cereb Blood Flow Metab 2021; 41:2957-2972. [PMID: 34044665 PMCID: PMC8545042 DOI: 10.1177/0271678x211018904] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The novel tau-PET tracer [18F]PI-2620 detects the 3/4-repeat-(R)-tauopathy Alzheimer's disease (AD) and the 4R-tauopathies corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP). We determined whether [18F]PI-2620 binding characteristics deriving from non-invasive reference tissue modelling differentiate 3/4R- and 4R-tauopathies. Ten patients with a 3/4R tauopathy (AD continuum) and 29 patients with a 4R tauopathy (CBS, PSP) were evaluated. [18F]PI-2620 PET scans were acquired 0-60 min p.i. and the distribution volume ratio (DVR) was calculated. [18F]PI-2620-positive clusters (DVR ≥ 2.5 SD vs. 11 healthy controls) were evaluated by non-invasive kinetic modelling. R1 (delivery), k2 & k2a (efflux), DVR, 30-60 min standardized-uptake-value-ratios (SUVR30-60) and the linear slope of post-perfusion phase SUVR (9-60 min p.i.) were compared between 3/4R- and 4R-tauopathies. Cortical clusters of 4R-tau cases indicated higher delivery (R1SRTM: 0.92 ± 0.21 vs. 0.83 ± 0.10, p = 0.0007), higher efflux (k2SRTM: 0.17/min ±0.21/min vs. 0.06/min ± 0.07/min, p < 0.0001), lower DVR (1.1 ± 0.1 vs. 1.4 ± 0.2, p < 0.0001), lower SUVR30-60 (1.3 ± 0.2 vs. 1.8 ± 0.3, p < 0.0001) and flatter slopes of the post-perfusion phase (slope9-60: 0.006/min ± 0.007/min vs. 0.016/min ± 0.008/min, p < 0.0001) when compared to 3/4R-tau cases. [18F]PI-2620 binding characteristics in cortical regions differentiate 3/4R- and 4R-tauopathies. Higher tracer clearance indicates less stable binding in 4R tauopathies when compared to 3/4R-tauopathies.
Collapse
Affiliation(s)
- Mengmeng Song
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Thilo van Eimeren
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ken Marek
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Alexander Nitschmann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Gesine Respondek
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Maike Kern
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Hammes
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Gèrard Bischof
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Michael Barbe
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Özgür Onur
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry, University Hospital Cologne, Cologne, Germany.,Center for Memory Disorders, University Hospital Cologne, Cologne, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany.,LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.,Max- Planck-Institute of Human Cognitive and Brain Sciences, Leipzig, Germany.,FTLD Consortium Germany, Ulm, Germany
| | | | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Bernd Neumaier
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Olivier Barret
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA.,Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, CEA, CNRS, MIRCen, Fontenay-aux-Roses, France
| | - Jennifer Madonia
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - David S Russell
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | | | | | - Sigrun Roeber
- Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Center for Neuropathology and Prion Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Kai Bötzel
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Günter U Höglinger
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Technical University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Victor Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - John Seibyl
- InviCRO, LLC, Boston, MA, USA.,Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
8
|
Young CB, Landau SM, Harrison TM, Poston KL, Mormino EC. Influence of common reference regions on regional tau patterns in cross-sectional and longitudinal [ 18F]-AV-1451 PET data. Neuroimage 2021; 243:118553. [PMID: 34487825 PMCID: PMC8785682 DOI: 10.1016/j.neuroimage.2021.118553] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/06/2021] [Accepted: 09/02/2021] [Indexed: 10/30/2022] Open
Abstract
Tau PET has allowed for critical insights into in vivo patterns of tau accumulation and change in individuals early in the Alzheimer's disease (AD) continuum. A key methodological step in tau PET analyses is the selection of a reference region, but there is not yet consensus on the optimal region especially for longitudinal tau PET analyses. This study examines how reference region selection influences results related to disease stage at baseline and over time. Longitudinal flortaucipir ([18F]-AV1451) PET scans were examined using several common reference regions (e.g., eroded subcortical white matter, inferior cerebellar gray matter) in 62 clinically unimpaired amyloid negative (CU A-) individuals, 73 CU amyloid positive (CU A+) individuals, and 64 amyloid positive individuals with mild cognitive impairment (MCI A+) from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Cross-sectionally, both reference regions resulted in robust group differences between CU A-, CU A+, and MCI A+ groups, along with significant associations with CSF phosphorylated tau (pTau-181). However, these results were more focally specific and akin to Braak Staging when using eroded white matter, whereas effects with inferior cerebellum were globally distributed across most cortical regions. Longitudinally, utilization of eroded white matter revealed significant accumulation greater than zero across more regions whereas change over time was diminished using inferior cerebellum. Interestingly, the inferior temporal target region seemed most robust to reference region selection with expected cross-sectional and longitudinal signal across both reference regions. With few exceptions, baseline tau did not significantly predict longitudinal change in tau in the same region regardless of reference region. In summary, reference region selection deserves further evaluation as this methodological step may lead to disparate findings. Inferior cerebellar gray matter may be more sensitive to cross-sectional flortaucipir differences, whereas eroded subcortical white matter may be more sensitive for longitudinal analyses examining regional patterns of change.
Collapse
Affiliation(s)
- Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA United States.
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA United States
| | - Theresa M Harrison
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA United States
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA United States
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA United States
| | | |
Collapse
|
9
|
Zhou Y, Flores S, Mansor S, Hornbeck RC, Tu Z, Perlmutter JS, Ances B, Morris JC, Gropler RJ, Benzinger TLS. Spatially constrained kinetic modeling with dual reference tissues improves 18F-flortaucipir PET in studies of Alzheimer disease. Eur J Nucl Med Mol Imaging 2021; 48:3172-3186. [PMID: 33599811 PMCID: PMC8371062 DOI: 10.1007/s00259-020-05134-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/20/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Recent studies have shown that standard compartmental models using plasma input or the cerebellum reference tissue input are generally not reliable for quantifying tau burden in dynamic 18F-flortaucipir PET studies of Alzheimer disease. So far, the optimal reference region for estimating 18F-flortaucipir delivery and specific tau binding has yet to be determined. The objective of the study is to improve 18F-flortaucipir brain tau PET quantification using a spatially constrained kinetic model with dual reference tissues. METHODS Participants were classified as either cognitively normal (CN) or cognitively impaired (CI) based on clinical assessment. T1-weighted structural MRI and 105-min dynamic 18F-flortaucipir PET scans were acquired for each participant. Using both a simplified reference tissue model (SRTM2) and Logan plot with either cerebellum gray matter or centrum semiovale (CS) white matter as the reference tissue, we estimated distribution volume ratios (DVRs) and the relative transport rate constant R1 for region of interest-based (ROI) and voxelwise-based analyses. Conventional linear regression (LR) and LR with spatially constrained (LRSC) parametric imaging algorithms were then evaluated. Noise-induced bias in the parametric images was compared to estimates from ROI time activity curve-based kinetic modeling. We finally evaluated standardized uptake value ratios at early phase (SUVREP, 0.7-2.9 min) and late phase (SUVRLP, 80-105 min) to approximate R1 and DVR, respectively. RESULTS The percent coefficients of variation of R1 and DVR estimates from SRTM2 with spatially constrained modeling were comparable to those from the Logan plot and SUVRs. The SRTM2 using CS reference tissue with LRSC reduced noise-induced underestimation in the LR generated DVR images to negligible levels (< 1%). Inconsistent overestimation of DVR in the SUVRLP only occurred using the cerebellum reference tissue-based measurements. The CS reference tissue-based DVR and SUVRLP, and cerebellum-based SUVREP and R1 provided higher Cohen's effect size d to detect increased tau deposition and reduced relative tracer transport rate in CI individuals. CONCLUSION Using a spatially constrained kinetic model with dual reference tissues significantly improved quantification of relative perfusion and tau binding. Cerebellum and CS are the suggested reference tissues to estimate R1 and DVR, respectively, for dynamic 18F-flortaucipir PET studies. Cerebellum-based SUVREP and CS-based SUVRLP may be used to simplify 18F-flortaucipir PET study.
Collapse
Affiliation(s)
- Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA.
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Syahir Mansor
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Zhude Tu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Joel S Perlmutter
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
| | - Beau Ances
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
10
|
Wolters EE, Dodich A, Boccardi M, Corre J, Drzezga A, Hansson O, Nordberg A, Frisoni GB, Garibotto V, Ossenkoppele R. Clinical validity of increased cortical uptake of [ 18F]flortaucipir on PET as a biomarker for Alzheimer's disease in the context of a structured 5-phase biomarker development framework. Eur J Nucl Med Mol Imaging 2021; 48:2097-2109. [PMID: 33547556 PMCID: PMC8175307 DOI: 10.1007/s00259-020-05118-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE In 2017, the Geneva Alzheimer's disease (AD) Biomarker Roadmap initiative adapted the framework of the systematic validation of oncological diagnostic biomarkers to AD biomarkers, with the aim to accelerate their development and implementation in clinical practice. With this work, we assess the maturity of [18F]flortaucipir PET and define its research priorities. METHODS The level of maturity of [18F]flortaucipir was assessed based on the AD Biomarker Roadmap. The framework assesses analytical validity (phases 1-2), clinical validity (phases 3-4), and clinical utility (phase 5). RESULTS The main aims of phases 1 (rationale for use) and 2 (discriminative ability) have been achieved. [18F]Flortaucipir binds with high affinity to paired helical filaments of tau and has favorable kinetic properties and excellent discriminative accuracy for AD. The majority of secondary aims of phase 2 were fully achieved. Multiple studies showed high correlations between ante-mortem [18F]flortaucipir PET and post-mortem tau (as assessed by histopathology), and also the effects of covariates on tracer binding are well studied. The aims of phase 3 (early detection ability) were only partially or preliminarily achieved, and the aims of phases 4 and 5 were not achieved. CONCLUSION Current literature provides partial evidence for clinical utility of [18F]flortaucipir PET. The aims for phases 1 and 2 were mostly achieved. Phase 3 studies are currently ongoing. Future studies including representative MCI populations and a focus on healthcare outcomes are required to establish full maturity of phases 4 and 5.
Collapse
Affiliation(s)
- E E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Centre for Mind/Brain Sciences-CIMeC, University of Trento, Rovereto, Italy
| | - M Boccardi
- Late Translational Dementia Studies Group, German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Rostock, Germany
| | - J Corre
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- CURIC, Centre Universitaire Romand d'Implants Cochléaires, Department of Clinical Neurosciences, University of Geneva, Geneva, Switzerland
| | - A Drzezga
- Faculty of Medicine, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - O Hansson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
| | - R Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Pereira JB, Janelidze S, Ossenkoppele R, Kvartsberg H, Brinkmalm A, Mattsson-Carlgren N, Stomrud E, Smith R, Zetterberg H, Blennow K, Hansson O. Untangling the association of amyloid-β and tau with synaptic and axonal loss in Alzheimer's disease. Brain 2021; 144:310-324. [PMID: 33279949 PMCID: PMC8210638 DOI: 10.1093/brain/awaa395] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/04/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023] Open
Abstract
It is currently unclear how amyloid-β and tau deposition are linked to changes in
synaptic function and axonal structure over the course of Alzheimer’s disease. Here, we
assessed these relationships by measuring presynaptic (synaptosomal-associated protein 25,
SNAP25; growth-associated protein 43, GAP43), postsynaptic (neurogranin, NRGN) and axonal
(neurofilament light chain) markers in the CSF of individuals with varying levels of
amyloid-β and tau pathology based on 18F-flutemetamol PET and
18F-flortaucipir PET. In addition, we explored the relationships between
synaptic and axonal markers with cognition as well as functional and anatomical brain
connectivity markers derived from resting-state functional MRI and diffusion tensor
imaging. We found that the presynaptic and postsynaptic markers SNAP25, GAP43 and NRGN are
elevated in early Alzheimer’s disease i.e. in amyloid-β-positive individuals without
evidence of tau pathology. These markers were associated with greater amyloid-β pathology,
worse memory and functional changes in the default mode network. In contrast,
neurofilament light chain was abnormal in later disease stages, i.e. in individuals with
both amyloid-β and tau pathology, and correlated with more tau and worse global cognition.
Altogether, these findings support the hypothesis that amyloid-β and tau might have
differential downstream effects on synaptic and axonal function in a stage-dependent
manner, with amyloid-related synaptic changes occurring first, followed by tau-related
axonal degeneration.
Collapse
Affiliation(s)
- Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Hlin Kvartsberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
12
|
Leuzy A, Smith R, Ossenkoppele R, Santillo A, Borroni E, Klein G, Ohlsson T, Jögi J, Palmqvist S, Mattsson-Carlgren N, Strandberg O, Stomrud E, Hansson O. Diagnostic Performance of RO948 F 18 Tau Positron Emission Tomography in the Differentiation of Alzheimer Disease From Other Neurodegenerative Disorders. JAMA Neurol 2021; 77:955-965. [PMID: 32391858 PMCID: PMC7215644 DOI: 10.1001/jamaneurol.2020.0989] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Question How does RO948 F 18 positron emission tomographic scanning discriminate between Alzheimer disease and other neurodegenerative disorders in comparison with magnetic resonance imaging and cerebrospinal fluid measures? Findings In this diagnostic study including 613 patients from the Swedish BioFINDER-2 clinical trial, standard uptake value ratios of RO948 F 18 were higher in patients with Alzheimer disease dementia compared with cognitively unimpaired controls and patients with other neurodegenerative disorders; furthermore, RO948 F 18 outperformed magnetic resonance imaging and cerebrospinal fluid measures. Generally, tau positron emission tomographic positivity was confined to amyloid β–positive cases or MAPT R406W mutation carriers in this cohort; in patients with semantic variant primary progressive aphasia, RO948 F 18 retention was lower than that for flortaucipir F 18. Meaning These findings suggest that RO948 F 18 has a high specificity for Alzheimer disease–type tau and highlight its potential as a diagnostic marker in the workup of patients treated in memory clinics. Importance The diagnostic performance of second-generation tau positron emission tomographic (PET) tracers is not yet known. Objective To examine the novel tau PET tracer RO948 F 18 ([18F]RO948) performance in discriminating Alzheimer disease (AD) from non-AD neurodegenerative disorders. Design, Setting, and Participants In this diagnostic study, 613 participants in the Swedish BioFINDER-2 study were consecutively enrolled in a prospective cross-sectional study from September 4, 2017, to August 28, 2019. Participants included 257 cognitively unimpaired controls, 154 patients with mild cognitive impairment, 100 patients with AD dementia, and 102 with non-AD neurodegenerative disorders. Evaluation included a comparison of tau PET tracer [18F]RO948 with magnetic resonance imaging (MRI) and cerebrospinal fluid and a head-to-head comparison between [18F]RO948 and flortaucipir F 18 ([18F]flortaucipir) in patients with semantic variant primary progressive aphasia (svPPA). Exposures [18F]RO948 (all patients) and [18F]flortaucipir (3 patients with svPPA) tau PET; MRI (hippocampal volume, composite temporal lobe cortical thickness, whole-brain cortical thickness) and cerebrospinal fluid measures (p-tau181 and amyloid Aβ42 and Aβ40 ratio[Aβ42/Aβ40], and Aβ42/p-tau181 ratio). Main Outcomes and Measures Standard uptake value ratios (SUVRs) in 4 predefined regions of interest (ROIs) reflecting Braak staging scheme for tau pathology and encompass I-II (entorhinal cortex), III-IV (inferior/middle temporal, fusiform gyrus, parahippocampal cortex, and amygdala), I-IV, and V-VI (widespread neocortical areas), area under the receiver operating characteristic curve (AUC) values, and subtraction images between [18F]RO948 and [18F]flortaucipir. Results Diagnostic groups among the 613 participants included cognitively unimpaired (mean [SD] age, 65.8 [12.1] years; 117 men [46%]), mild cognitive impairment (age, 70.8 [8.3] years; 82 men [53%]), AD dementia (age, 73.5 [6.7] years; 57 men [57%]), and non-AD disorders (age, 70.5 [8.6] years; 41 men [40%]). Retention of [18F]RO948 was higher in AD dementia compared with all other diagnostic groups. [18F]RO948 could distinguish patients with AD dementia from individuals without cognitive impairment and those with non-AD disorders, and the highest AUC was obtained using the I-IV ROI (AUC = 0.98; 95% CI, 0.96-0.99 for AD vs no cognitive impairment and AUC = 0.97; 95% CI, 0.95-0.99 for AD vs non-AD disorders), which outperformed MRI (highest AUC = 0.91 for AD vs no cognitive impairment using whole-brain thickness, and AUC = 0.80 for AD vs non-AD disorders using temporal lobe thickness) and cerebrospinal fluid measures (highest AUC = 0.94 for AD vs no cognitive impairment using Aβ42/p-tau181, and AUC = 0.93 for AD vs non-AD disorders using Aβ42/Aβ40). Generally, tau PET positivity using [18F]RO948 was observed only in Aβ-positive cases or in MAPT R406W mutation carriers. Retention of [18F]RO948 was not pronounced in patients with svPPA, and head-to-head comparison revealed lower temporal lobe uptake than with [18F]flortaucipir. Conclusions and Relevance In this study, elevated [18F]RO948 SUVRs were most often seen among Aβ-positive cases, which suggests that [18F]RO948 has high specificity for AD-type tau and highlights its potential as a diagnostic marker in the differential diagnosis of AD.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | | | | | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Jonas Jögi
- Skåne University Hospital, Department of Clinical Physiology and Nuclear Medicine, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Smith R, Strandberg O, Mattsson-Carlgren N, Leuzy A, Palmqvist S, Pontecorvo MJ, Devous MD, Ossenkoppele R, Hansson O. The accumulation rate of tau aggregates is higher in females and younger amyloid-positive subjects. Brain 2021; 143:3805-3815. [PMID: 33439987 PMCID: PMC7805812 DOI: 10.1093/brain/awaa327] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
The development of tau-PET allows paired helical filament tau pathology to be visualized in vivo. Increased knowledge about conditions affecting the rate of tau accumulation could guide the development of therapies halting the progression of Alzheimer’s disease. However, the factors modifying the rate of tau accumulation over time in Alzheimer’s disease are still largely unknown. Large-scale longitudinal cohort studies, adjusting for baseline tau load, are needed to establish such risk factors. In the present longitudinal study, 419 participants from four cohorts in the USA (Avid 05e, n = 157; Expedition-3, n = 82; ADNI, n = 123) and Sweden (BioFINDER, n = 57) were scanned repeatedly with tau-PET. The study participants were cognitively unimpaired (n = 153), or patients with mild cognitive impairment (n = 139) or Alzheimer’s disease dementia (n = 127). Participants underwent two to four tau-PET (18F-flortaucipir) scans with a mean (± standard deviation) of 537 (±163) days between the first and last scan. The change in tau-PET signal was estimated in temporal meta- and neocortical regions of interest. Subject specific tau-PET slopes were predicted simultaneously by age, sex, amyloid status (determined by amyloid-β PET), APOE ε4 genotype, study cohort, diagnosis and baseline tau load. We found that accelerated increase in tau-PET signal was observed in amyloid-β-positive mild cognitive impairment (3.0 ± 5.3%) and Alzheimer’s disease dementia (2.9 ± 5.7%), respectively, when compared to either amyloid-β-negative cognitively unimpaired (0.4 ± 2.7%), amyloid-β-negative mild cognitive impairment (−0.4 ± 2.3%) or amyloid-β-positive cognitively unimpaired (1.2 ± 2.8%). Tau-PET uptake was accelerated in females (temporal region of interest: t = 2.86, P = 0.005; neocortical region of interest: t = 2.90, P = 0.004), younger individuals (temporal region of interest: t = −2.49, P = 0.013), and individuals with higher baseline tau-PET signal (temporal region of interest: t = 3.83, P < 0.001; neocortical region of interest: t = 5.01, P < 0.001). Tau-PET slopes decreased with age in amyloid-β-positive subjects, but were stable by age in amyloid-β-negative subjects (age × amyloid-β status interaction: t = −2.39, P = 0.018). There were no effects of study cohort or APOE ε4 positivity. In a similar analysis on longitudinal amyloid-β-PET (in ADNI subjects only, n = 639), we found significant associations between the rate of amyloid-β accumulation and APOE ε4 positivity, older age and baseline amyloid-β positivity, but no effect of sex. In conclusion, in this longitudinal PET study comprising four cohorts, we found that the tau accumulation rate is greater in females and younger amyloid-β-positive individuals, while amyloid-β accumulation is greater in APOE ε4 carriers and older individuals. These findings are important considerations for the design of clinical trials, and might improve our understanding of factors associated with faster tau aggregation and spread.
Collapse
Affiliation(s)
- Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | | | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Amsterdam University Medical Center, Alzheimercenter, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
14
|
Herholz K. Hide-and-seek for radiotracers and neurodegenerative pathology. Eur J Nucl Med Mol Imaging 2020; 47:1791-1792. [PMID: 31897583 DOI: 10.1007/s00259-019-04661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Karl Herholz
- Division of Neuroscience and Experimental Psychology, University of Manchester, 27 Palatine Road, Manchester, M20 3LJ, UK.
| |
Collapse
|
15
|
Franzmeier N, Dewenter A, Frontzkowski L, Dichgans M, Rubinski A, Neitzel J, Smith R, Strandberg O, Ossenkoppele R, Buerger K, Duering M, Hansson O, Ewers M. Patient-centered connectivity-based prediction of tau pathology spread in Alzheimer's disease. SCIENCE ADVANCES 2020; 6:eabd1327. [PMID: 33246962 PMCID: PMC7695466 DOI: 10.1126/sciadv.abd1327] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/02/2020] [Indexed: 05/25/2023]
Abstract
In Alzheimer's disease (AD), the Braak staging scheme suggests a stereotypical tau spreading pattern that does, however, not capture interindividual variability in tau deposition. This complicates the prediction of tau spreading, which may become critical for defining individualized tau-PET readouts in clinical trials. Since tau is assumed to spread throughout connected regions, we used functional connectivity to improve tau spreading predictions over Braak staging methods. We included two samples with longitudinal tau-PET from controls and AD patients. Cross-sectionally, we found connectivity of tau epicenters (i.e., regions with earliest tau) to predict estimated tau spreading sequences. Longitudinally, we found tau accumulation rates to correlate with connectivity strength to patient-specific tau epicenters. A connectivity-based, patient-centered tau spreading model improved the assessment of tau accumulation rates compared to Braak stage-specific readouts and reduced sample sizes by ~40% in simulated tau-targeting interventions. Thus, connectivity-based tau spreading models may show utility in clinical trials.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lukas Frontzkowski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Anna Rubinski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Julia Neitzel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Ruben Smith
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Katharina Buerger
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
16
|
Pawlik D, Leuzy A, Strandberg O, Smith R. Compensating for choroid plexus based off-target signal in the hippocampus using 18F-flortaucipir PET. Neuroimage 2020; 221:117193. [PMID: 32711062 DOI: 10.1016/j.neuroimage.2020.117193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/18/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE The hippocampus is affected by tau pathology early in Alzheimer's disease (AD) development. Accurate quantification of hippocampal tau signal using the tau-PET tracer 18F-flortaucipir is complicated, however, by off-target binding in the adjacent choroid plexus. We here present a new method for compensating for this off-target choroid plexus signal. METHODS As off-target binding in the choroid plexus is known to be higher using 18F-flortaucipir compared to 18F-RO948, we created a binary hippocampal mask in template space where 18F-flortaucipir signal was higher than 18F-RO948, using data from 30 patients that underwent both 18F-flortaucipir and 18F-RO948 PET. This mask, presumably representing hippocampal voxels affected by off-target binding from the choroid plexus, was then converted to native space and applied as an exclusion mask to 145 patients across the AD-spectrum scanned with 18F-flortaucipir. As an alternative approach exclusion masks were generated by expanding the choroid plexus ROI in native space. Results were analysed both without and with partial volume error correction (non-PVEc/PVEc). RESULTS Unmasked hippocampal standardized uptake value ratios (SUVR) were significantly correlated to choroid plexus SUVRs using both non-PVEc (p < 0.001, r = 0.28) and PVEc data (p < 0.05, r = 0.18). After applying the mask, however, these correlations disappeared. The diagnostic accuracy in separating cognitively impaired (CI) from cognitively unimpaired (CU) subjects improved after masking, from an AUC of 0.792 (95% C.I.,0.715-0.869) to 0.837 (95% C.I.,0.768-0.906) for non-PVEc data (p < 0.001), and from 0.798 (95% C.I.,0.722-0.873) to 0.834 (95% C.I.,0.766-0.903) for PVEc data (p < 0.001). The correlations to memory improved significantly for MMSE for unmasked vs. masked data both without (r = -0.440 vs. r = -0.499, p < 0.001) and with (r = -0.454 vs. r = -0.503, p < 0.001) PVEc. Similar results were found using the ADAS-Cog Delayed Word Recall test. CONCLUSION Choroid plexus off-target binding interferes with the estimation of true hippocampal retention using 18F-flortaucipir PET. Using a mask to correct for this off-target signal, we improved the diagnostic accuracy of 18F-flortaucipir in the hippocampus and the correlation between 18F-flortaucipir hippocampal SUVR and cognitive measures.
Collapse
Affiliation(s)
- Daria Pawlik
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Neurology, Skåne University Hospital, SE-20502 Malmö, Lund, Sweden.
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Neurology, Skåne University Hospital, SE-20502 Malmö, Lund, Sweden.
| |
Collapse
|
17
|
Gao C, Gong W, Yang M, Chu X, Wang Y, Li Z, Yang Y, Gao C. T807-modified human serum albumin biomimetic nanoparticles for targeted drug delivery across the blood-brain barrier. J Drug Target 2020; 28:1085-1095. [PMID: 32614677 DOI: 10.1080/1061186x.2020.1777420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Novel biocompatible Human Serum Albumin (HSA) nanoparticles composed of membrane of erythrocytes (ETm)-coated and DSPE-PEG3400-T807 segments have been designed for sustained drug delivery across the blood-brain barrier (BBB). The nanoparticles have developed by induced albumin self-assembly with glutathione as reducing agent. The chemical, physical and biocompatible properties of the T807/ETm-HSA nanoparticles have been characterised by hydrogen nuclear magnetic resonance, matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry, transmission electron microscopy, dynamic light scattering and confocal laser scanning microscopy techniques. The unique targeting properties of the nanoparticles after fabrication with the brain-targeted ligand T807 was demonstrated by their attaching to brain cells as well as their enhanced transport ability to cross the BBB. In a further demonstration of their ability to target brain cells, in vivo living imaging revealed that T807/ETm-HSA nanoparticles accumulated in the mice brain after intravenous injection. The surface modification of ETm/HSA nanoparticles with the brain-targeted T807 demonstrated in this work represents a highly novel and effective strategy to provide efficient brain targeting and shows promise for the future in using modified ETm-coated HSA nanoparticles to penetrate the brain.
Collapse
Affiliation(s)
- Chunhong Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaoyang Chu
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
18
|
Snitz BE, Tudorascu DL, Yu Z, Campbell E, Lopresti BJ, Laymon CM, Minhas DS, Nadkarni NK, Aizenstein HJ, Klunk WE, Weintraub S, Gershon RC, Cohen AD. Associations between NIH Toolbox Cognition Battery and in vivo brain amyloid and tau pathology in non-demented older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12018. [PMID: 32426450 PMCID: PMC7228102 DOI: 10.1002/dad2.12018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/04/2022]
Abstract
INTRODUCTION The National Institutes of Health (NIH) Toolbox Cognition Battery (NIHTB-CB) was developed to be a common assessment metric across a broad array of research studies. We investigated associations between NIHTB-CB and brain amyloid and tau deposition in cognitively unimpaired older adults. METHODS One hundred eighteen community-based volunteers completed magnetic resonance imaging (MRI), Pittsburgh compound B (PiB)-PET (positron emission tomography) and AV-1451-PET neuroimaging, a neuropsychological evaluation, NIHTB-CB, and the Clinical Dementia Rating (CDR) scale. Demographically adjusted regression models evaluated cognition-biomarker associations; standardized effect sizes allowed comparison of association strength across measures. RESULTS No NIHTB-CB measures were associated with amyloid deposition. NIHTB-CB measures of fluid cognition, including Pattern Comparison Processing Speed, Dimensional Change Card Sort, and Fluid Cognition Composite, were associated with tau deposition in higher Braak regions. Pattern Comparison Processing Speed was the most robust association with sensitivity analyses. DISCUSSION NIHTB-CB tasks of processing speed and executive functions may be sensitive to pathologic tau deposition on imaging in normal aging.
Collapse
Affiliation(s)
- Beth E. Snitz
- Department of NeurologyUniversity of PittsburghSchool of MedicinePittsburghPennsylvania
| | - Dana L. Tudorascu
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Zheming Yu
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Elizabeth Campbell
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Brian J. Lopresti
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Charles M. Laymon
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
- Department of BioengineeringUniversity of Pittsburgh School of EngineeringPittsburghPennsylvania
| | - Davneet S. Minhas
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Neelesh K. Nadkarni
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Howard J. Aizenstein
- Department of BioengineeringUniversity of Pittsburgh School of EngineeringPittsburghPennsylvania
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - William E. Klunk
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Sandra Weintraub
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinois
| | - Richard C. Gershon
- Department of Medical Social SciencesNorthwestern University Feinberg School of MedicineChicagoIllinois
| | - Ann D. Cohen
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| |
Collapse
|
19
|
Mattsson-Carlgren N, Leuzy A, Janelidze S, Palmqvist S, Stomrud E, Strandberg O, Smith R, Hansson O. The implications of different approaches to define AT(N) in Alzheimer disease. Neurology 2020; 94:e2233-e2244. [PMID: 32398359 PMCID: PMC7357296 DOI: 10.1212/wnl.0000000000009485] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/19/2019] [Indexed: 12/05/2022] Open
Abstract
Objective To compare different β-amyloid (Aβ), tau, and neurodegeneration (AT[N]) variants within the Swedish BioFINDER studies. Methods A total of 490 participants were classified into AT(N) groups. These include 53 cognitively unimpaired (CU) and 48 cognitively impaired (CI) participants (14 mild cognitive impairment [MCI] and 34 Alzheimer disease [AD] dementia) from BioFINDER-1 and 389 participants from BioFINDER-2 (245 CU and 144 CI [138 MCI and 6 AD dementia]). Biomarkers for A were CSF Aβ42 and amyloid-PET ([18F]flutemetamol); for T, CSF phosphorylated tau (p-tau) and tau PET ([18F]flortaucipir); and for (N), hippocampal volume, temporal cortical thickness, and CSF neurofilament light (NfL). Binarization of biomarkers was achieved using cutoffs defined in other cohorts. The relationship between different AT(N) combinations and cognitive trajectories (longitudinal Mini-Mental State Examination scores) was examined using linear mixed modeling and coefficient of variation. Results Among CU participants, A−T−(N)− or A+T−(N)− variants were most common. However, more T+ cases were seen using p-tau than tau PET. Among CI participants, A+T+(N)+ was more common; however, more (N)+ cases were seen for MRI measures relative to CSF NfL. Tau PET best predicted longitudinal cognitive decline in CI and p-tau in CU participants. Among CI participants, continuous T (especially tau PET) and (N) measures improved the prediction of cognitive decline compared to binary measures. Conclusions Our findings show that different AT(N) variants are not interchangeable, and that optimal variants differ by clinical stage. In some cases, dichotomizing biomarkers may result in loss of important prognostic information.
Collapse
Affiliation(s)
- Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden.
| | - Antoine Leuzy
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Shorena Janelidze
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Ruben Smith
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit, Department of Clinical Sciences (N.M.-C., A.L., S.J., S.P., E.S., O.S., R.S., O.H.), and Wallenberg Centre for Molecular Medicine (N.M.-C.), Lund University, Malmö; and Department of Neurology (N.M.-C., S.P., R.S.) and Memory Clinic (E.S., O.H.), Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
20
|
Gao C, Wang Y, Sun J, Han Y, Gong W, Li Y, Feng Y, Wang H, Yang M, Li Z, Yang Y, Gao C. Neuronal mitochondria-targeted delivery of curcumin by biomimetic engineered nanosystems in Alzheimer's disease mice. Acta Biomater 2020; 108:285-299. [PMID: 32251785 DOI: 10.1016/j.actbio.2020.03.029] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Biomimetic nanotechnology represents a promising approach for the delivery of therapeutic agents for the treatment of complex diseases. Recently, neuronal mitochondria have been proposed to serve as a promising therapeutic target for sporadic Alzheimer's disease (AD). However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological and pathological environment. Herein, we devised and tested a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-camouflaged human serum albumin nanoparticles bearing T807 and triphenylphosphine (TPP) molecules attached to the RBC membrane surface (T807/TPP-RBC-NPs). With the advantage of the suitable physicochemical properties of the nanoparticles and the unique biological functions of the RBC membrane, the T807/TPP-RBC-NPs are stabilized and promote sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of T807 and TPP, T807/TPP-RBC-NPs can not only penetrate the blood-brain barrier (BBB) but also target nerve cells and further localize in the mitochondria. After encapsulating curcumin (CUR) as the model antioxidant, the research data demonstrated that CUR-loaded T807/TPP-RBC-NPs can relieve AD symptoms by mitigating mitochondrial oxidative stress and suppressing neuronal death both in vitro and in vivo. In conclusion, the intravenous neuronal mitochondria-targeted biomimetic engineered delivery nanosystems provides an effective drug delivery platform for brain diseases. STATEMENT OF SIGNIFICANCE: The efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge for drug delivery due to the complicated physiological and pathological environment. To address this need, various types of nanovessels have been fabricated using a variety of materials in the last few decades. However, problems with the synthetic materials still exist and even cause toxicology issues. New findings in nanomedicine are promoting the development of biomaterials. Herein, we designed a red blood cell (RBC) membrane-coated human serum albumin nanoparticle dual-modified with T807 and TPP (T807/TPP-RBC-NPs) to accomplish these objectives. After encapsulating curcumin as the model drug, the research data demonstrated that the intravenous neuronal mitochondria-targeted biomimetic engineered delivery nanosystems are a promising therapeutic candidate for mitochondrial dysfunction in Alzheimer's disease (AD).
Collapse
|
21
|
Janelidze S, Stomrud E, Smith R, Palmqvist S, Mattsson N, Airey DC, Proctor NK, Chai X, Shcherbinin S, Sims JR, Triana-Baltzer G, Theunis C, Slemmon R, Mercken M, Kolb H, Dage JL, Hansson O. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer's disease. Nat Commun 2020; 11:1683. [PMID: 32246036 PMCID: PMC7125218 DOI: 10.1038/s41467-020-15436-0] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/06/2020] [Indexed: 01/01/2023] Open
Abstract
Cerebrospinal fluid (CSF) p-tau181 (tau phosphorylated at threonine 181) is an established biomarker of Alzheimer's disease (AD), reflecting abnormal tau metabolism in the brain. Here we investigate the performance of CSF p-tau217 as a biomarker of AD in comparison to p-tau181. In the Swedish BioFINDER cohort (n = 194), p-tau217 shows stronger correlations with the tau positron emission tomography (PET) tracer [18F]flortaucipir, and more accurately identifies individuals with abnormally increased [18F]flortaucipir retention. Furthermore, longitudinal increases in p-tau217 are higher compared to p-tau181 and better correlate with [18F]flortaucipir uptake. P-tau217 correlates better than p-tau181 with CSF and PET measures of neocortical amyloid-β burden and more accurately distinguishes AD dementia from non-AD neurodegenerative disorders. Higher correlations between p-tau217 and [18F]flortaucipir are corroborated in an independent EXPEDITION3 trial cohort (n = 32). The main results are validated using a different p-tau217 immunoassay. These findings suggest that p-tau217 might be more useful than p-tau181 in the diagnostic work up of AD.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden.
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
| | - Niklas Mattsson
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Entrégatan 7, 222 42, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Klinikgatan 32, 221 84, Lund, Sweden
| | | | | | - Xiyun Chai
- Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | - John R Sims
- Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Gallen Triana-Baltzer
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA
| | - Clara Theunis
- Janssen Pharmaceutical Companies of Johnson & Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Randy Slemmon
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA
| | - Marc Mercken
- Janssen Pharmaceutical Companies of Johnson & Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Hartmuth Kolb
- Neuroscience Biomarkers, Janssen Research & Development, 3210 Merryfield Row, San Diego, CA, CA 92121, USA.
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Sölvegatan 18, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Simrisbanvägen 14, 205 02, Malmö, Sweden.
| |
Collapse
|
22
|
Janelidze S, Mattsson N, Palmqvist S, Smith R, Beach TG, Serrano GE, Chai X, Proctor NK, Eichenlaub U, Zetterberg H, Blennow K, Reiman EM, Stomrud E, Dage JL, Hansson O. Plasma P-tau181 in Alzheimer's disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer's dementia. Nat Med 2020; 26:379-386. [PMID: 32123385 DOI: 10.1038/s41591-020-0755-1] [Citation(s) in RCA: 609] [Impact Index Per Article: 152.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
Plasma phosphorylated tau181 (P-tau181) might be increased in Alzheimer's disease (AD), but its usefulness for differential diagnosis and prognosis is unclear. We studied plasma P-tau181 in three cohorts, with a total of 589 individuals, including cognitively unimpaired participants and patients with mild cognitive impairment (MCI), AD dementia and non-AD neurodegenerative diseases. Plasma P-tau181 was increased in preclinical AD and further increased at the MCI and dementia stages. It correlated with CSF P-tau181 and predicted positive Tau positron emission tomography (PET) scans (area under the curve (AUC) = 0.87-0.91 for different brain regions). Plasma P-tau181 differentiated AD dementia from non-AD neurodegenerative diseases with an accuracy similar to that of Tau PET and CSF P-tau181 (AUC = 0.94-0.98), and detected AD neuropathology in an autopsy-confirmed cohort. High plasma P-tau181 was associated with subsequent development of AD dementia in cognitively unimpaired and MCI subjects. In conclusion, plasma P-tau181 is a noninvasive diagnostic and prognostic biomarker of AD, which may be useful in clinical practice and trials.
Collapse
Affiliation(s)
| | - Niklas Mattsson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | | | | - Xiyun Chai
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
23
|
Golla SSV, Wolters EE, Timmers T, Ossenkoppele R, van der Weijden CWJ, Scheltens P, Schwarte L, Mintun MA, Devous Sr MD, Schuit RC, Windhorst AD, Lammertsma AA, Yaqub M, van Berckel BNM, Boellaard R. Parametric methods for [ 18F]flortaucipir PET. J Cereb Blood Flow Metab 2020; 40:365-373. [PMID: 30569813 PMCID: PMC7044757 DOI: 10.1177/0271678x18820765] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 12/22/2022]
Abstract
[18F]Flortaucipir is a PET tau tracer used to visualize tau binding in Alzheimer's disease (AD) in vivo. The present study evaluated the performance of several methods to obtain parametric images of [18F]flortaucipir. One hundred and thirty minutes dynamic PET scans were performed in 10 AD patients and 10 controls. Parametric images were generated using different linearization and basis function approaches. Regional binding potential (BPND) and volume of distribution (VT) values obtained from the parametric images were compared with corresponding values derived using the reversible two-tissue compartment model (2T4k_VB). Performance of SUVr parametric images was assessed by comparing values with distribution volume ratio (DVR) and SRTM-derived BPND estimates obtained using non-linear regression (NLR). Spectral analysis (SA) (r2 = 0.92; slope = 0.99) derived VT correlated well with NLR-derived VT. RPM (r2 = 0.95; slope = 0.98) derived BPND correlated well with NLR-derived DVR. Although SUVr80-100 min correlated well with NLR-derived DVR (r2 = 0.91; slope = 1.09), bias in SUVr appeared to depend on uptake time and underlying level of specific binding. In conclusion, RPM and SA provide parametric images comparable to the NLR estimates. Individual SUVr values are biased compared with DVR and this bias requires further study in a larger dataset in order to understand its consequences.
Collapse
Affiliation(s)
- Sandeep SV Golla
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Emma E Wolters
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
- Alzheimer Center and Department of
Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Tessa Timmers
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
- Alzheimer Center and Department of
Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Rik Ossenkoppele
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
- Alzheimer Center and Department of
Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Chris WJ van der Weijden
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
- Alzheimer Center and Department of
Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Philip Scheltens
- Alzheimer Center and Department of
Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Lothar Schwarte
- Department of Anaesthesiology, Amsterdam
Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | - Robert C Schuit
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Bart NM van Berckel
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear
Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The
Netherlands
| |
Collapse
|
24
|
Franzmeier N, Neitzel J, Rubinski A, Smith R, Strandberg O, Ossenkoppele R, Hansson O, Ewers M. Functional brain architecture is associated with the rate of tau accumulation in Alzheimer's disease. Nat Commun 2020; 11:347. [PMID: 31953405 PMCID: PMC6969065 DOI: 10.1038/s41467-019-14159-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
In Alzheimer's diseases (AD), tau pathology is strongly associated with cognitive decline. Preclinical evidence suggests that tau spreads across connected neurons in an activity-dependent manner. Supporting this, cross-sectional AD studies show that tau deposition patterns resemble functional brain networks. However, whether higher functional connectivity is associated with higher rates of tau accumulation is unclear. Here, we combine resting-state fMRI with longitudinal tau-PET in two independent samples including 53 (ADNI) and 41 (BioFINDER) amyloid-biomarker defined AD subjects and 28 (ADNI) vs. 16 (BioFINDER) amyloid-negative healthy controls. In both samples, AD subjects show faster tau accumulation than controls. Second, in AD, higher fMRI-assessed connectivity between 400 regions of interest (ROIs) is associated with correlated tau-PET accumulation in corresponding ROIs. Third, we show that a model including baseline connectivity and tau-PET is associated with future tau-PET accumulation. Together, connectivity is associated with tau spread in AD, supporting the view of transneuronal tau propagation.
Collapse
Affiliation(s)
- Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universitat München, Ludwig-Maximilians-Universitat LMU, Munich, Germany.
| | - Julia Neitzel
- Institute for Stroke and Dementia Research, Klinikum der Universitat München, Ludwig-Maximilians-Universitat LMU, Munich, Germany
| | - Anna Rubinski
- Institute for Stroke and Dementia Research, Klinikum der Universitat München, Ludwig-Maximilians-Universitat LMU, Munich, Germany
| | - Ruben Smith
- Department of Neurology, Skane University Hospital, Lund, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences Malmo, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmo, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmo, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmo, Lund University, Lund, Sweden.,Memory Clinic, Skane University Hospital, Malmo, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universitat München, Ludwig-Maximilians-Universitat LMU, Munich, Germany
| | | |
Collapse
|
25
|
Pereira JB, Ossenkoppele R, Palmqvist S, Strandberg TO, Smith R, Westman E, Hansson O. Amyloid and tau accumulate across distinct spatial networks and are differentially associated with brain connectivity. eLife 2019; 8:50830. [PMID: 31815669 PMCID: PMC6938400 DOI: 10.7554/elife.50830] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
Abstract
The abnormal accumulation of amyloid-β and tau targets specific spatial networks in Alzheimer's disease. However, the relationship between these networks across different disease stages and their association with brain connectivity has not been explored. In this study, we applied a joint independent component analysis to 18F- Flutemetamol (amyloid-β) and 18F-Flortaucipir (tau) PET images to identify amyloid-β and tau networks across different stages of Alzheimer's disease. We then assessed whether these patterns were associated with resting-state functional networks and white matter tracts. Our analyses revealed nine patterns that were linked across tau and amyloid-β data. The amyloid-β and tau patterns showed a fair to moderate overlap with distinct functional networks but only tau was associated with white matter integrity loss and multiple cognitive functions. These findings show that amyloid-β and tau have different spatial affinities, which can be used to understand how they accumulate in the brain and potentially damage the brain's connections.
Collapse
Affiliation(s)
- Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Tor Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
26
|
Heurling K, Smith R, Strandberg OT, Schain M, Ohlsson T, Hansson O, Schöll M. Regional times to equilibria and their impact on semi-quantification of [ 18F]AV-1451 uptake. J Cereb Blood Flow Metab 2019; 39:2223-2232. [PMID: 30073880 PMCID: PMC6827127 DOI: 10.1177/0271678x18791430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/08/2018] [Accepted: 06/20/2018] [Indexed: 11/17/2022]
Abstract
The semi-quantitative estimate standardised uptake value ratios (SUVR) correlate well with specific binding of the tracer expressed as distribution volume ratios (DVR) for the tau positron emission tomography tracer [18F]AV-1451 uptake and are therefore widely used as proxy for tracer binding. With regard to tracer kinetic modelling, there exists a time point when SUVR deviates minimally from DVR, occurring when the specific binding reaches a transient equilibrium. Here, we have investigated whether the time to equilibrium affects the agreement between SUVR and DVR across different brain regions. We show that the time required to reach equilibrium differs across brain regions, resulting in region-specific biases. However, even though the 80-100 min post-injection time window did not show the smallest bias numerically, the disagreement between SUVR and DVR varied least between regions during this time. In conclusion, our findings suggest a regional component to the bias of SUVR related to the time to transient equilibrium of the specific binding. [18F]AV-1451 uptake should consequently be interpreted with some caution when compared across brain regions using this method of quantification. The commonly used time window 80-100 min post-injection shows the most consistent bias across regions and is recommended for semi-quantification of [18F]AV-1451.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Ruben Smith
- Department of Neurology, Lund University, Skåne University Hospital, Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| | - Olof T Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| | - Martin Schain
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund/Malmö, Sweden
| |
Collapse
|
27
|
Head-to-head comparison of tau positron emission tomography tracers [ 18F]flortaucipir and [ 18F]RO948. Eur J Nucl Med Mol Imaging 2019; 47:342-354. [PMID: 31612245 PMCID: PMC6974501 DOI: 10.1007/s00259-019-04496-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE [18F]flortaucipir binds to paired helical filament tau and accurately identifies tau in Alzheimer's disease (AD). However, "off-target" binding interferes with the quantification of [18F]flortaucipir in several brain regions. Recently, other tau PET tracers have been developed. Here, we compare [18F]flortaucipir with the novel tau tracer [18F]RO948 head-to-head in vivo. METHODS We included 18 participants with AD, three with amyloid-β-positive amnestic mild cognitive impairment, and four healthy controls. All underwent [18F]flortaucipir (80-100 min) and [18F]RO948 (70-90) PET scans within approximately 1 month. Four study participants underwent 0-100-min dynamic scanning. Standardized uptake value ratios (SUVRs) were created using an inferior cerebellar reference region. RESULTS Neocortical tracer retention was highly comparable using both SUVR and distribution volume ratio-1 values obtained from dynamic scans. However, [18F]RO948 retention was significantly higher in the entorhinal cortex and lower in the basal ganglia, thalamus, and choroid plexus compared with [18F]flortaucipir. Increased off-target binding was observed with age for both tracers. Several cases exhibited strong [18F]RO948 retention in the skull/meninges. This extra-cerebral signal, however, did not affect diagnostic accuracy and remained relatively unchanged when re-examining a subsample after 1 year. Kinetic modeling showed an increase in [18F]flortaucipir SUVR over the scanning interval, compared with a plateau for [18F]RO948. CONCLUSION [18F]RO948 and [18F]flortaucipir bound comparably in neocortical regions, but [18F]RO948 showed higher retention in the medial temporal lobe and lower intracerebral "off-target" binding. Time-dependent bias of SUVR estimates may prove less of a factor with [18F]RO948, compared with previous tau ligands.
Collapse
|
28
|
Perani D, Iaccarino L, Lammertsma AA, Windhorst AD, Edison P, Boellaard R, Hansson O, Nordberg A, Jacobs AH. A new perspective for advanced positron emission tomography-based molecular imaging in neurodegenerative proteinopathies. Alzheimers Dement 2019; 15:1081-1103. [PMID: 31230910 DOI: 10.1016/j.jalz.2019.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Recent studies in neurodegenerative conditions have increasingly highlighted that the same neuropathology can trigger different clinical phenotypes or, vice-versa, that similar phenotypes can be triggered by different neuropathologies. This evidence has called for the adoption of a pathology spectrum-based approach to study neurodegenerative proteinopathies. These conditions share brain deposition of abnormal protein aggregates, leading to aberrant biochemical, metabolic, functional, and structural changes. Positron emission tomography (PET) is a well-recognized and unique tool for the in vivo assessment of brain neuropathology, and novel PET techniques are emerging for the study of specific protein species. Today, key applications of PET range from early research and clinical diagnostic tools to their use in clinical trials for both participants screening and outcome evaluation. This position article critically reviews the role of distinct PET molecular tracers for different neurodegenerative proteinopathies, highlighting their strengths, weaknesses, and opportunities, with special emphasis on methodological challenges and future applications.
Collapse
Affiliation(s)
- Daniela Perani
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Iaccarino
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK; Neurology Imaging Unit, Imperial College London, London, UK
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Center for Alzheimer Research, Stockholm, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany; Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn, Germany.
| | | |
Collapse
|
29
|
Abstract
Purpose of Review Abnormal accumulation of tau protein is the main hallmark of tauopathies and is closely associated with neurodegeneration and cognitive impairment, whereas the advance in PET imaging provides a non-invasive detection of tau inclusions in the brain. In this review, we discuss the potential of PET imaging as a biomarker in tauopathies, the latest development of novel tau tracers with new clinical information that has been disclosed, and the opportunities for improving diagnosis and designing clinical trials in the future. Recent Findings In recent years, several first-generation tau PET tracers including [11C]PBB3, [18F]THK-5117, [18F]THK-5351 and [18F]AV-1451 have been developed and succeeded in imaging neurofibrillary pathology in vivo. Due to the common off-target binding and subcortical white matter uptake seen in the first-generation tracers, several research institutes and pharmaceutical companies have been working on developing second-generation tau PET tracers which exhibit higher binding affinity and selectivity. Summary Tau PET imaging is promising to serve as a biomarker to support differential diagnosis and monitor disease progression in many neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Ting Wang
- Neurology Imaging Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paul Edison
- Neurology Imaging Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK. .,Cardiff University, Cardiff, CF10 3AT, UK.
| |
Collapse
|
30
|
Smith R, Santillo AF, Waldö ML, Strandberg O, Berron D, Vestberg S, van Westen D, van Swieten J, Honer M, Hansson O. 18F-Flortaucipir in TDP-43 associated frontotemporal dementia. Sci Rep 2019; 9:6082. [PMID: 30988363 PMCID: PMC6465310 DOI: 10.1038/s41598-019-42625-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
Retention of 18F-Flortaucipir is reportedly increased in the semantic variant of primary progressive aphasia (svPPA), which is dominated by TDP-43 pathology. However, it is unclear if 18F-Flortaucipir is also increased in other TDP-43 diseases, such as bvFTD caused by a C9orf72 gene mutation. We therefore recruited six C9orf72 expansion carriers, six svPPA patients, and 54 healthy controls. All underwent 18F-Flortaucipir PET and MRI scanning. Data from 39 Alzheimer’s Disease patients were used for comparison. PET tracer retention was assessed both at the region-of-interest (ROI) and at the voxel-level. Further, autoradiography using 3H-Flortaucipir was performed. SvPPA patients exhibited higher 18F-Flortaucipir retention in the lateral temporal cortex bilaterally according to ROI- and voxel-based analyses. In C9orf72 patients, 18F-Flortaucipir binding was slightly increased in the inferior frontal lobes in the ROI based analysis, but these results were not replicated in the voxel-based analysis. Autoradiography did not show specific binding in svPPA cases or in C9orf72-mutation carriers. In conclusion, temporal lobe 18F-Flortaucipir retention was observed in some cases of svPPA, but the uptake was of a lower magnitude compared to AD dementia. C9orf72-mutation carriers exhibited none or limited 18F-Flortaucipir retention, indicating that 18F-Flortaucipir binding in TDP-43 proteinopathies is not a general TDP-43 related phenomenon.
Collapse
Affiliation(s)
- R Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden. .,Department of Neurology, Skåne University Hospital, Lund, Sweden.
| | - A F Santillo
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - M Landqvist Waldö
- Clinical Sciences Helsingborg, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - O Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - D Berron
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - S Vestberg
- Department of Psychology, Lund University, Lund, Sweden
| | - D van Westen
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - J van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - M Honer
- Roche Pharmaceutical Research and Early Development, Neuroscience Translational Technologies, Roche Innovation Center, Basel, Switzerland
| | - O Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
31
|
Lois C, Gonzalez I, Johnson KA, Price JC. PET imaging of tau protein targets: a methodology perspective. Brain Imaging Behav 2019; 13:333-344. [PMID: 29497982 PMCID: PMC6119534 DOI: 10.1007/s11682-018-9847-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The two neuropathological hallmarks of Alzheimer's disease (AD) are amyloid-[Formula: see text] plaques and neurofibrillary tangles of tau protein. Fifteen years ago, Positron Emission Tomography (PET) with Pittsburgh Compound B (11C-PiB) enabled selective in-vivo visualization of amyloid-[Formula: see text] plaque deposits and has since provided valuable information about the role of amyloid-[Formula: see text] deposition in AD. The progression of tau deposition has been shown to be highly associated with neuronal loss, neurodegeneration, and cognitive decline. Until recently it was not possible to visualize tau deposition in-vivo, but several tau PET tracers are now available in different stages of clinical development. To date, no tau tracer has been approved by the Food and Drug Administration for use in the evaluation of AD or other tauopathies, despite very active research efforts. In this paper we review the recent developments in tau PET imaging with a focus on in-vivo findings in AD and discuss the challenges associated with tau tracer development, the status of development and validation of different tau tracers, and the clinical information these provide.
Collapse
Affiliation(s)
- Cristina Lois
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA.
| | - Ivan Gonzalez
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Radiotracers for imaging of Parkinson's disease. Eur J Med Chem 2019; 166:75-89. [DOI: 10.1016/j.ejmech.2019.01.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/12/2019] [Accepted: 01/13/2019] [Indexed: 12/22/2022]
|
33
|
Mattsson N, Insel PS, Donohue M, Jögi J, Ossenkoppele R, Olsson T, Schöll M, Smith R, Hansson O. Predicting diagnosis and cognition with 18F-AV-1451 tau PET and structural MRI in Alzheimer's disease. Alzheimers Dement 2019; 15:570-580. [PMID: 30639421 DOI: 10.1016/j.jalz.2018.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 11/01/2018] [Accepted: 12/02/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION The relative importance of structural magnetic resonance imaging (MRI) and tau positron emission tomography (PET) to predict diagnosis and cognition in Alzheimer's disease (AD) is unclear. METHODS We tested 56 cognitively unimpaired controls (including 27 preclinical AD), 32 patients with prodromal AD, and 39 patients with AD dementia. Optimal classifiers were constructed using the least absolute shrinkage and selection operator with 18F-AV-1451 (tau) PET and structural MRI data (regional cortical thickness and subcortical volumes). RESULTS 18F-AV-1451 in the amygdala, entorhinal cortex, parahippocampal gyrus, fusiform, and inferior parietal lobule had 93% diagnostic accuracy for AD (prodromal or dementia). The MRI classifier involved partly the same regions plus the hippocampus, with 83% accuracy, but did not improve upon the tau classifier. 18F-AV-1451 retention and MRI were independently associated with cognition. DISCUSSION Optimized tau PET classifiers may diagnose AD with high accuracy, but both tau PET and structural brain MRI capture partly unique information relevant for the clinical deterioration in AD.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden; Lund University, Skåne University Hospital, Department of Clinical Sciences, Neurology, Lund, Sweden.
| | - Philip S Insel
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; Center for Imaging of Neurodegenerative Diseases, Department of Veterans Affairs Medical Center, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Michael Donohue
- Department of Neurology, Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | - Jonas Jögi
- Department of Clinical Physiology and Nuclear Medicine, Skåne University Hospital, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; VU University Medical Center, Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Tomas Olsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Michael Schöll
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; Lund University, Skåne University Hospital, Department of Clinical Sciences, Neurology, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
34
|
Optimal timing of tau pathology imaging and automatic extraction of a reference region using dynamic [ 18F]THK5317 PET. NEUROIMAGE-CLINICAL 2019; 22:101681. [PMID: 30710871 PMCID: PMC6357848 DOI: 10.1016/j.nicl.2019.101681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/18/2018] [Accepted: 01/20/2019] [Indexed: 12/27/2022]
Abstract
[18F]THK5317 is a PET tracer for in-vivo imaging of tau associated with Alzheimer's disease (AD). This work aimed to evaluate optimal timing for standardized uptake value ratio (SUVR) measures with [18F]THK5317 and automated generation of SUVR-1 and relative cerebral blood flow (R1) parametric images. Nine AD patients and nine controls underwent 90 min [18F]THK5317 scans. SUVR-1 was calculated at transient equilibrium (TE) and for seven different 20 min intervals and compared with distribution volume ratio (DVR; reference Logan). Cerebellar grey matter (MRI) was used as reference region. A supervised cluster analysis (SVCA) method was implemented to automatically generate a reference region, directly from the dynamic PET volume without the need of a structural MRI scan, for computation of SUVR-1 and R1 images for a scan duration matching the optimal timing. TE was reached first in putamen, frontal- and parietal cortex at 22 ± 4 min for AD patients and in putamen at 20 ± 0 min in controls. Over all regions and subjects, SUVR20-40-1 correlated best with DVR-1, R2 = 0.97. High correlation was found between values generated using MRI- and SVCA-based reference (R2 = 0.93 for SUVR20-40-1; R2 = 0.94 for R1). SUVR20-40 allows for accurate semi-quantitative assessment of tau pathology and SVCA may be used to obtain a reference region for calculation of both SUVR-1 and R1 with 40 min scan duration.
Collapse
|
35
|
Schöll M, Maass A, Mattsson N, Ashton NJ, Blennow K, Zetterberg H, Jagust W. Biomarkers for tau pathology. Mol Cell Neurosci 2018; 97:18-33. [PMID: 30529601 PMCID: PMC6584358 DOI: 10.1016/j.mcn.2018.12.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/01/2018] [Indexed: 12/14/2022] Open
Abstract
The aggregation of fibrils of hyperphosphorylated and C-terminally truncated microtubule-associated tau protein characterizes 80% of all dementia disorders, the most common neurodegenerative disorders. These so-called tauopathies are hitherto not curable and their diagnosis, especially at early disease stages, has traditionally proven difficult. A keystone in the diagnosis of tauopathies was the development of methods to assess levels of tau protein in vivo in cerebrospinal fluid, which has significantly improved our knowledge about these conditions. Tau proteins have also been measured in blood, but the importance of tau-related changes in blood is still unclear. The recent addition of positron emission tomography ligands to visualize, map and quantify tau pathology has further contributed with information about the temporal and spatial characteristics of tau accumulation in the living brain. Together, the measurement of tau with fluid biomarkers and positron emission tomography constitutes the basis for a highly active field of research. This review describes the current state of biomarkers for tau biomarkers derived from neuroimaging and from the analysis of bodily fluids and their roles in the detection, diagnosis and prognosis of tau-associated neurodegenerative disorders, as well as their associations with neuropathological findings, and aims to provide a perspective on how these biomarkers might be employed prospectively in research and clinical settings. Biomarkers for tau pathology are now essential to the research framework in the diagnosis of Alzheimer's disease (AD) Measurement of t- and p-tau has been possible in cerebrospinal fluid (CSF) for some time, the recent development of positron emission tomography (PET) ligands binding to tau has added the possibility to map and quantify tau in the living brain First-generation tau PET ligands bind predominantly to AD-typical 3R/4R tau isoforms and exhibit off-target binding that can limit accurate ligand uptake quantification Second-generation tau PET ligands appear to bind to comparable binding sites but exhibit fewer issues with brain off-target binding Biomarkers for tau derived from CSF analysis and PET could provide complementary information about disease state and stage At this time, T-tau, but not p-tau, can be reliably measured in plasma using ultra-sensitive immunoassays.
Collapse
Affiliation(s)
- Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden; Clinical Memory Research Unit, Lund University, Malmö, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Anne Maass
- German Center for Neurodegenerative Diseases, Magdeburg, Germany; Helen Wills Neuroscience Institute, University of California, Berkeley, USA
| | - Niklas Mattsson
- Clinical Memory Research Unit, Lund University, Malmö, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden; King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
36
|
Kuwabara H, Comley RA, Borroni E, Honer M, Kitmiller K, Roberts J, Gapasin L, Mathur A, Klein G, Wong DF. Evaluation of 18F-RO-948 PET for Quantitative Assessment of Tau Accumulation in the Human Brain. J Nucl Med 2018; 59:1877-1884. [PMID: 30097505 DOI: 10.2967/jnumed.118.214437] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023] Open
Abstract
The availability of tau PET radioligands enables quantitative assessment of tau density and distribution in the human brain. We evaluated the kinetics of a novel radioligand, 18F-RO-948 (previously referred to as 18F-RO6958948), and its ability to identify tau positivity in individual patients with mild Alzheimer disease (AD). Methods: Eleven subjects with amyloid-positive mild AD, 5 amyloid-negative older control subjects (OC), and 5 younger control subjects (YC) completed 1 or 2 (4 AD and 5 OC) PET scans with 18F-RO-948 for 90, 120, or 200 min. The kinetics of the radioligand was evaluated with standard compartmental and noncompartmental models (with plasma data in 70% of cases), tissue-reference methods, and SUV ratio. These approaches were applied to assess the ability of 18F-RO-948 to discriminate AD subjects from OC subjects. Results: The plasma reference graphical analysis appeared to be the optimal method of quantification for 18F-RO-948, yielding strictly time-consistent values of distribution volume and distribution volume ratio at 90 min against the analyses at 120 and 200 min. The reference tissue graphical analysis and SUV ratio were cross-validated against plasma reference graphical analysis. Test-retest evaluation showed excellent reproducibility. A proposed novel index of tau load, the regional tau-positive fraction, showed high values in the medial and lateral temporal and parietal regions in AD and successfully separated AD subjects from OC and YC subjects with a significant margin. Conclusion: 18F-RO-948 appears to be a promising radioligand for quantitative imaging of tau in the brain of AD patients.
Collapse
Affiliation(s)
- Hiroto Kuwabara
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert A Comley
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Edilio Borroni
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Michael Honer
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Kelly Kitmiller
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joshua Roberts
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lorena Gapasin
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anil Mathur
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gregory Klein
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Dean F Wong
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Mattsson N, Schöll M, Strandberg O, Smith R, Palmqvist S, Insel PS, Hägerström D, Ohlsson T, Zetterberg H, Jögi J, Blennow K, Hansson O. 18F-AV-1451 and CSF T-tau and P-tau as biomarkers in Alzheimer's disease. EMBO Mol Med 2018; 9:1212-1223. [PMID: 28743782 PMCID: PMC5582410 DOI: 10.15252/emmm.201707809] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
To elucidate the relationship between cerebrospinal fluid (CSF) total-tau (T-tau) and phosphorylated tau (P-tau) with the tau PET ligand 18F-AV-1451 in Alzheimer's disease (AD), we examined 30 cognitively healthy elderly (15 with preclinical AD), 14 prodromal AD, and 39 AD dementia patients. CSF T-tau and P-tau were highly correlated (R = 0.92, P < 0.001), but they were only moderately associated with retention of 18F-AV-1451, and mainly in demented AD patients. 18F-AV-1451, but not CSF T-tau or P-tau, was strongly associated with atrophy and cognitive impairment. CSF tau was increased in preclinical AD, despite normal 18F-AV-1451 retention. However, not all dementia AD patients exhibited increased CSF tau, even though 18F-AV-1451 retention was always increased at this disease stage. We conclude that CSF T-tau and P-tau mainly behave as biomarkers of "disease state", since they appear to be increased in many cases of AD at all disease stages, already before the emergence of tau aggregates. In contrast, 18F-AV-1451 is a biomarker of "disease stage", since it is increased in clinical stages of the disease, and is associated with brain atrophy and cognitive decline.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden .,Memory Clinic, Skåne University Hospital, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Michael Schöll
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,MedTech West and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Philip S Insel
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Douglas Hägerström
- Department of Clinical Neurophysiology, Skåne University Hospital, Lund, Sweden
| | - Tomas Ohlsson
- Department of Radiation physics, Skåne University Hospital, Lund, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Jonas Jögi
- Department of Clinical Physiology and Nuclear Medicine, Skåne University Hospital, Lund, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden .,Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
38
|
Smith R, Schöll M, Londos E, Ohlsson T, Hansson O. 18F-AV-1451 in Parkinson's Disease with and without dementia and in Dementia with Lewy Bodies. Sci Rep 2018; 8:4717. [PMID: 29549278 PMCID: PMC5856779 DOI: 10.1038/s41598-018-23041-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/05/2018] [Indexed: 01/08/2023] Open
Abstract
Mixed pathologies of α-synuclein, β-amyloid and tau are relatively common in Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). We therefore wanted to study the retention patterns of 18F-AV-1451 in PD, PD-dementia (PDD), and DLB. To do this 44 healthy controls, 11 non-demented patients with PD, 18 patients with PDD, and six patients with DLB underwent MRI and 18F-AV-1451 PET scanning and cognitive testing. We found that parietal 18F-AV-1451 retention was increased in patients with DLB compared to controls and PD patients, while 18F-AV-1451 uptake was reduced in the substantia nigra in PDD. Increased parietal 18F-AV-1451 PET uptake was associated with impaired performance on verbal fluency tests, and the decreased uptake in the substantia nigra correlated with worse motor function. We found no effect of the monoamine oxidase B inhibitor rasagiline on 18F-AV-1451 binding. In conclusion DLB patients have increased parietal 18F-AV-1451 uptake. Increased parietal tau is associated with executive impairment in patients with synucleinopathies, while decreased uptake in the substantia nigra is associated with parkinsonism. Further, our data indicate that 18F-AV-1451 does not significantly bind to MAO-B in vivo.
Collapse
Affiliation(s)
- Ruben Smith
- Department of Neurology, Skåne University Hospital, Lund, Sweden. .,Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | - Michael Schöll
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Londos
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
39
|
Firouzian A, Whittington A, Searle GE, Koychev I, Zamboni G, Lovestone S, Gunn RN. Imaging Aβ and tau in early stage Alzheimer's disease with [ 18F]AV45 and [ 18F]AV1451. EJNMMI Res 2018; 8:19. [PMID: 29500717 PMCID: PMC5834417 DOI: 10.1186/s13550-018-0371-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/19/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AD is a progressive neurodegenerative disorder that is associated with the accumulation of two different insoluble protein aggregates, Aβ plaques and hyperphosphorylated tau. This study aimed to investigate the optimal acquisition and quantification of [18F]AV45 and [18F]AV1451 to image Aβ and tau, respectively, in subjects with AD. Fifteen subjects with early stage AD underwent a T1-weighted structural MRI and two dynamic PET scans to image Aβ (60 min, [18F]AV45) and tau (120 min, [18F]AV1451). Both dynamic BPND and static SUVR outcome measures were calculated and compared for 12 out of 15 subjects who completed 60 min of the Aβ PET scan and at least 110 min of the tau PET scan. The SRTM and reference Logan graphical analysis were applied to the dynamic data to estimate regional BPND values and SUVR ratios from the static data. Optimal acquisition windows were explored for both the dynamic and static acquisitions. In addition, the spatial correlation between regional Aβ and tau signals was explored. RESULTS Both the SRTM and graphical analysis methods showed a good fit to the dynamic data for both Aβ and tau dynamic PET scans. Mean regional BPND estimates became stable 30 min p.i. for [18F]AV45 and 80 min p.i. for [18F]AV1451. Time stability analysis of static SUVR data showed that the outcome measure starts to become stable for scan windows of 30-50 min p.i. for [18F]AV45 and 80-100 min p.i. for [18F]AV1451. The results from these time windows correlated well with the results from the full dynamic analysis for both tracers (R2 = 0.74 for [18F]AV45 and R2 = 0.88 for [18F]AV1451). There was a high correlation between amyloid uptake estimate using both dynamic analysis methods in thalamus and tau uptake in thalamus, hippocampus and amygdala. CONCLUSIONS Short static PET scans at appropriate time windows provided SUVR values which were in reasonable agreement with BPND values calculated from dynamic scans using SRTM and reference Logan. These simplified methods may be appropriate for classification and intervention studies, although caution should be employed when considering interventional studies where blood flow and extraction could change.
Collapse
Affiliation(s)
- Azadeh Firouzian
- Imanova Ltd., Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
| | - Alex Whittington
- Department of Medicine, Faculty of Medicine, Imperial College London, South Kensington Campus, London, SW7 2AZ UK
| | - Graham E. Searle
- Imanova Ltd., Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
| | - Ivan Koychev
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK
| | - Giovanna Zamboni
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU UK
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK
| | - Roger N. Gunn
- Imanova Ltd., Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
- Department of Medicine, Faculty of Medicine, Imperial College London, South Kensington Campus, London, SW7 2AZ UK
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - on behalf of the Deep and Frequent Phenotyping study team
- Imanova Ltd., Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
- Department of Medicine, Faculty of Medicine, Imperial College London, South Kensington Campus, London, SW7 2AZ UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU UK
- Department of Engineering Science, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Chen Q, Du Y, Zhang K, Liang Z, Li J, Yu H, Ren R, Feng J, Jin Z, Li F, Sun J, Zhou M, He Q, Sun X, Zhang H, Tian M, Ling D. Tau-Targeted Multifunctional Nanocomposite for Combinational Therapy of Alzheimer's Disease. ACS NANO 2018; 12:1321-1338. [PMID: 29364648 DOI: 10.1021/acsnano.7b07625] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Alzheimer's disease (AD) remains an incurable disease and lacks efficient diagnostic methods. Most AD treatments have focused on amyloid-β (Aβ) targeted therapy; however, it is time to consider the alternative theranostics due to accumulated findings of weak correlation between Aβ deposition and cognition, as well as the failures of Phase III clinical trial on Aβ targeted therapy. Recent studies have shown that the tau pathway is closely associated with clinical development of AD symptoms, which might be a potential therapeutic target. We herein construct a methylene blue (MB, a tau aggregation inhibitor) loaded nanocomposite (CeNC/IONC/MSN-T807), which not only possesses high binding affinity to hyperphosphorylated tau but also inhibits multiple key pathways of tau-associated AD pathogenesis. We demonstrate that these nanocomposites can relieve the AD symptoms by mitigating mitochondrial oxidative stress, suppressing tau hyperphosphorylation, and preventing neuronal death both in vitro and in vivo. The memory deficits of AD rats are significantly rescued upon treatment with MB loaded CeNC/IONC/MSN-T807. Our results indicate that hyperphosphorylated tau-targeted multifunctional nanocomposites could be a promising therapeutic candidate for Alzheimer's disease.
Collapse
Affiliation(s)
- Qing Chen
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
| | - Yang Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
| | - Kai Zhang
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
| | - Zeyu Liang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
| | - Jinquan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
| | - Hao Yu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
| | - Rong Ren
- College of Chemical & Biological Engineering, Zhejiang University , Hangzhou, Zhejiang 310027, P.R. China
| | - Jin Feng
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
| | - Zhiming Jin
- Jiangsu Huayi Technology Limited Company , Changshu, Jiangsu 215522, P.R. China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University , Hangzhou, Zhejiang 310058, P.R. China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310016, P.R. China
| | - Min Zhou
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
| | - Qinggang He
- College of Chemical & Biological Engineering, Zhejiang University , Hangzhou, Zhejiang 310027, P.R. China
| | - Xiaolian Sun
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Hong Zhang
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
- Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, P.R. China
| | - Mei Tian
- Department of Nuclear Medicine and PET/CT Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, Zhejiang 310009, P.R. China
- Collaborative Innovation Center for Brain Science, Fudan University , Shanghai 200032, P.R. China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P.R. China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University , Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
41
|
Klunk WE. Molecular imaging: What is right and what is an illusion? ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2018; 10:217-220. [PMID: 29780866 PMCID: PMC5956935 DOI: 10.1016/j.dadm.2018.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Over the past 40 years, brain molecular imaging has evolved from measuring cerebral metabolism with fluorodeoxyglucose, to neuroreceptor imaging, to imaging pathological protein deposits. In the early going, the characteristics of successful molecular imaging radiotracers were defined, and a detailed "Process" was developed for the collection of basic pharmacodynamic and pharmacokinetic data. These data are essential for the interpretation of in vivo imaging data and for defining the strengths, weaknesses, and limitations of new tracers. This perspective discusses the use of this "Process" in the development of the amyloid β positron emission tomography radiotracer, Pittsburgh Compound-B, and discusses some of the current controversies and difficulties in the field of tau positron emission tomography in the context of human data that preceded completion of this radiotracer characterization process-which still remains to be completed. As a field, we must decide which data are valid and which are artifacts and determine that when the artifacts are so overwhelming, the data are merely an illusion.
Collapse
|
42
|
Mattsson N, Smith R, Strandberg O, Palmqvist S, Schöll M, Insel PS, Hägerström D, Ohlsson T, Zetterberg H, Blennow K, Jögi J, Hansson O. Comparing 18F-AV-1451 with CSF t-tau and p-tau for diagnosis of Alzheimer disease. Neurology 2018; 90:e388-e395. [PMID: 29321235 PMCID: PMC5791788 DOI: 10.1212/wnl.0000000000004887] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 10/05/2017] [Indexed: 11/15/2022] Open
Abstract
Objective To compare PET imaging of tau pathology with CSF measurements (total tau [t-tau] and phosphorylated tau [p-tau]) in terms of diagnostic performance for Alzheimer disease (AD). Methods We compared t-tau and p-tau and 18F-AV-1451 in 30 controls, 14 patients with prodromal AD, and 39 patients with Alzheimer dementia, recruited from the Swedish BioFINDER study. All patients with AD (prodromal and dementia) were screened for amyloid positivity using CSF β-amyloid 42. Retention of 18F-AV-1451 was measured in a priori specified regions, selected for known associations with tau pathology in AD. Results Retention of 18F-AV-1451 was markedly elevated in Alzheimer dementia and moderately elevated in prodromal AD. CSF t-tau and p-tau was increased to similar levels in both AD dementia and prodromal AD. 18F-AV-1451 had very good diagnostic performance for Alzheimer dementia (area under the receiver operating characteristic curve [AUROC] ∼1.000), and was significantly better than t-tau (0.876), p-tau (0.890), hippocampal volume (0.824), and temporal cortical thickness (0.860). For prodromal AD, there were no significant AUROC differences between CSF tau and 18F-AV-1451 measures (0.836–0.939), but MRI measures had lower AUROCs (0.652–0.769). Conclusions CSF tau and 18F-AV-1451 have equal performance in early clinical stages of AD, but 18F-AV-1451 is superior in the dementia stage, and exhibits close to perfect diagnostic performance for mild to moderate AD. Classification of evidence This study provides Class III evidence that CSF tau and 18F-AV-1451 PET have similar performance in identifying early AD, and that 18F-AV-1451 PET is superior to CSF tau in identifying mild to moderate AD.
Collapse
Affiliation(s)
- Niklas Mattsson
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| | - Ruben Smith
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Olof Strandberg
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Sebastian Palmqvist
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Michael Schöll
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Philip S Insel
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Douglas Hägerström
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Tomas Ohlsson
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Henrik Zetterberg
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Jonas Jögi
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Oskar Hansson
- From the Clinical Memory Research Unit (N.M., R.S., O.S., S.P., M.S., P.S.I., O.H.), Faculty of Medicine, Lund University; Memory Clinic (N.M., O.H.) and Departments of Neurology (N.M., R.S., S.P.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital, Lund; MedTech West and the Department of Clinical Neuroscience (M.S.), University of Gothenburg, Sweden; Center for Imaging of Neurodegenerative Diseases (P.S.I.), Department of Veterans Affairs Medical Center, San Francisco; Department of Radiology and Biomedical Imaging (P.S.I.), University of California, San Francisco; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology, Department of Molecular Neuroscience (H.Z., K.B.), the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|
43
|
Schonhaut DR, McMillan CT, Spina S, Dickerson BC, Siderowf A, Devous MD, Tsai R, Winer J, Russell DS, Litvan I, Roberson ED, Seeley WW, Grinberg LT, Kramer JH, Miller BL, Pressman P, Nasrallah I, Baker SL, Gomperts SN, Johnson KA, Grossman M, Jagust WJ, Boxer AL, Rabinovici GD. 18 F-flortaucipir tau positron emission tomography distinguishes established progressive supranuclear palsy from controls and Parkinson disease: A multicenter study. Ann Neurol 2017; 82:622-634. [PMID: 28980714 DOI: 10.1002/ana.25060] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/10/2017] [Accepted: 09/24/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE 18 F-flortaucipir (formerly 18 F-AV1451 or 18 F-T807) binds to neurofibrillary tangles in Alzheimer disease, but tissue studies assessing binding to tau aggregates in progressive supranuclear palsy (PSP) have yielded mixed results. We compared in vivo 18 F-flortaucipir uptake in patients meeting clinical research criteria for PSP (n = 33) to normal controls (n = 46) and patients meeting criteria for Parkinson disease (PD; n = 26). METHODS Participants underwent magnetic resonance imaging and positron emission tomography for amyloid-β (11 C-PiB or 18 F-florbetapir) and tau (18 F-flortaucipir). 18 F-flortaucipir standardized uptake value ratios were calculated (t = 80-100 minutes, cerebellum gray matter reference). Voxelwise and region-of-interest group comparisons were performed in template space, with receiver operating characteristic curve analyses to assess single-subject discrimination. Qualitative comparisons with postmortem tau are reported in 1 patient who died 9 months after 18 F-flortaucipir. RESULTS Clinical PSP patients showed bilaterally elevated 18 F-flortaucipir uptake in globus pallidus, putamen, subthalamic nucleus, midbrain, and dentate nucleus relative to controls and PD patients (voxelwise p < 0.05 family wise error corrected). Globus pallidus binding best distinguished PSP patients from controls and PD (area under the curve [AUC] = 0.872 vs controls, AUC = 0.893 vs PD). PSP clinical severity did not correlate with 18 F-flortaucipir in any region. A patient with clinical PSP and pathological diagnosis of corticobasal degeneration had severe tau pathology in PSP-related brain structures with good correspondence between in vivo 18 F-flortaucipir and postmortem tau neuropathology. INTERPRETATION 18 F-flortaucipir uptake was elevated in PSP versus controls and PD patients in a pattern consistent with the expected distribution of tau pathology. Ann Neurol 2017;82:622-634.
Collapse
Affiliation(s)
- Daniel R Schonhaut
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corey T McMillan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Salvatore Spina
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | | | | | - Richard Tsai
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Joseph Winer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | | | - Irene Litvan
- Department of Neurology, University of California, San Diego, San Diego, CA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - William W Seeley
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA.,Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Lea T Grinberg
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA.,Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Joel H Kramer
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Bruce L Miller
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Peter Pressman
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Ilya Nasrallah
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Suzanne L Baker
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | - Stephen N Gomperts
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA
| | - Murray Grossman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | - Adam L Boxer
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
44
|
Schöll M, Ossenkoppele R, Strandberg O, Palmqvist S, Jögi J, Ohlsson T, Smith R, Hansson O. Distinct 18F-AV-1451 tau PET retention patterns in early- and late-onset Alzheimer's disease. Brain 2017; 140:2286-2294. [PMID: 29050382 DOI: 10.1093/brain/awx171] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/29/2017] [Indexed: 11/13/2022] Open
Abstract
Patients with Alzheimer's disease can present with different clinical phenotypes. Individuals with late-onset Alzheimer's disease (>65 years) typically present with medial temporal lobe neurodegeneration and predominantly amnestic symptomatology, while patients with early-onset Alzheimer's disease (<65 years) exhibit greater neocortical involvement associated with a clinical presentation including dyspraxia, executive dysfunction, or visuospatial impairment. We recruited 20 patients with early-onset Alzheimer's disease, 21 with late-onset Alzheimer's disease, three with prodromal early-onset Alzheimer's disease and 13 with prodromal late-onset Alzheimer's disease, as well as 30 cognitively healthy elderly controls, that had undergone 18F-AV-1451 tau positron emission tomography and structural magnetic resonance imaging to explore whether early- and late-onset Alzheimer's disease exhibit differential regional tau pathology and atrophy patterns. Strong associations of lower age at symptom onset with higher 18F-AV-1451 uptake were observed in several neocortical regions, while higher age did not yield positive associations in neither patient group. Comparing patients with early-onset Alzheimer's disease with controls resulted in significantly higher 18F-AV-1451 retention throughout the neocortex, while comparing healthy controls with late-onset Alzheimer's disease patients yielded a distinct pattern of higher 18F-AV-1451 retention, predominantly confined to temporal lobe regions. When compared against each other, the early-onset Alzheimer's disease group exhibited greater uptake than the late-onset group in prefrontal and premotor, as well as in inferior parietal cortex. These preliminary findings indicate that age may constitute an important contributor to Alzheimer's disease heterogeneity highlighting the potential of tau positron emission tomography to capture phenotypic variation across patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Michael Schöll
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Rik Ossenkoppele
- Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | | - Jonas Jögi
- Department of Clinical Physiology and Nuclear Medicine, Skåne University Hospital, Lund, Sweden
| | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
45
|
Bischof GN, Endepols H, van Eimeren T, Drzezga A. Tau-imaging in neurodegeneration. Methods 2017; 130:114-123. [PMID: 28790016 DOI: 10.1016/j.ymeth.2017.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 10/19/2022] Open
Abstract
Pathological cerebral aggregations of proteins are suggested to play a crucial role in the development of neurodegenerative disorders. For example, aggregation of the protein ß-amyloid in form of extracellular amyloid-plaques as well as intraneuronal depositions of the protein tau in form of neurofibrillary tangles represent hallmarks of Alzheimer's disease (AD). Recently, novel tracers for in vivo molecular imaging of tau-aggregates in the brain have been introduced, complementing existing tracers for imaging amyloid-plaques. Available data on these novel tracers indicate that the subject of Tau-PET may be of considerable complexity. On the one hand this refers to the various forms of appearance of tau-pathology in different types of neurodegenerative disorders. On the other hand, a number of hurdles regarding validation of these tracers still need to be overcome with regard to comparability and standardization of the different tracers, observed off-target/non-specific binding and quantitative interpretation of the signal. These issues will have to be clarified before systematic clinical application of this exciting new methodological approach may become possible. Potential applications refer to early detection of neurodegeneration, differential diagnosis between tauopathies and non-tauopathies and specific patient selection and follow-up in therapy trials.
Collapse
Affiliation(s)
| | - Heike Endepols
- Department of Nuclear Medicine, University of Cologne, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany.
| |
Collapse
|
46
|
Smith R, Schöll M, Widner H, van Westen D, Svenningsson P, Hägerström D, Ohlsson T, Jögi J, Nilsson C, Hansson O. In vivo retention of 18F-AV-1451 in corticobasal syndrome. Neurology 2017; 89:845-853. [PMID: 28754841 PMCID: PMC5580862 DOI: 10.1212/wnl.0000000000004264] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/30/2017] [Indexed: 12/24/2022] Open
Abstract
Objective: To study the usefulness of 18F-AV-1451 PET in patients with corticobasal syndrome (CBS). Methods: We recruited 8 patients with CBS, 17 controls, 31 patients with Alzheimer disease (AD), and 11 patients with progressive supranuclear palsy (PSP) from the Swedish BioFINDER study. All patients underwent clinical assessment, 18F-AV-1451 PET, MRI, and quantification of β-amyloid pathology. A subset of participants also underwent 18F-FDG-PET. Results: In the 8 patients with CBS, 6 had imaging findings compatible with the corticobasal degeneration pathology and 2 with typical AD pathology. In the 6 patients with CBS without typical AD pathology, there were substantial retentions of 18F-AV-1451 in the motor cortex, corticospinal tract, and basal ganglia contralateral to the most affected body side. These patients could be clearly distinguished from patients with AD dementia or PSP using 18F-AV-1451. However, cortical atrophy was more widespread than the cortical retention of 18F-AV1451 in these CBS cases, and cortical AV-1451 uptake did not correlate with cortical thickness or glucose hypometabolism. These results are in sharp contrast to AD dementia, where 18F-AV-1451 retention was more widespread than cortical atrophy, and correlated well with cortical thickness and hypometabolism. Conclusions: Patients with CBS without typical AD pathology exhibited AV-1451 retention in the motor cortex, corticospinal tract, and basal ganglia contralateral to the affected body side, clearly different from controls and patients with AD dementia or PSP. However, cortical atrophy measured with MRI and decreased 18F-fluorodeoxyglucose uptake were more widespread than 18F-AV-1451 uptake and probably represent earlier, yet less specific, markers of CBS. Classification of evidence: This study provides Class III evidence that 18F-AV-1451 PET distinguishes between CBS and AD or PSP.
Collapse
Affiliation(s)
- Ruben Smith
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden.
| | - Michael Schöll
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Håkan Widner
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Danielle van Westen
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Per Svenningsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Douglas Hägerström
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Tomas Ohlsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Jonas Jögi
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Christer Nilsson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- From the Departments of Neurology (R.S., H.W., C.N.), Clinical Neurophysiology (D.H.), Radiation Physics (T.O.), and Clinical Physiology and Nuclear Medicine (J.J.), Skåne University Hospital (D.v.W.), Lund; Clinical Memory Research Unit (R.S., M.S., C.N., O.H.), Department of Clinical Sciences (D.v.W.), and Department of Diagnostic Radiology (D.v.W.), Lund University, Malmö; Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry (M.S.), University of Gothenburg; Department of Clinical Neuroscience (P.S.), CMM L8:01, Stockholm; and Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
47
|
Heurling K, Leuzy A, Jonasson M, Frick A, Zimmer ER, Nordberg A, Lubberink M. Quantitative positron emission tomography in brain research. Brain Res 2017; 1670:220-234. [PMID: 28652218 DOI: 10.1016/j.brainres.2017.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
The application of positron emission tomography (PET) in brain research has increased substantially during the past 20years, and is still growing. PET provides a unique insight into physiological and pathological processes in vivo. In this article we introduce the fundamentals of PET, and the methods available for acquiring quantitative estimates of the parameters of interest. A short introduction to different areas of application is also given, including basic research of brain function and in neurology, psychiatry, drug receptor occupancy studies, and its application in diagnostics of neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Our aim is to inform the unfamiliar reader of the underlying basics and potential applications of PET, hoping to inspire the reader into considering how the technique could be of benefit for his or her own research.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Antoine Leuzy
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - My Jonasson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas Frick
- Department of Psychology, Uppsala University, Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo R Zimmer
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Agneta Nordberg
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden; Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
48
|
Mossine AV, Brooks AF, Henderson BD, Hockley BG, Frey KA, Scott PJH. An updated radiosynthesis of [ 18F]AV1451 for tau PET imaging. EJNMMI Radiopharm Chem 2017; 2:7. [PMID: 29503848 PMCID: PMC5824695 DOI: 10.1186/s41181-017-0027-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 05/29/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND [18F]AV1451 is a commonly used radiotracer for imaging tau deposits in Alzheimer's disease (AD) and related non-AD tauopathies. Existing radiosyntheses of [18F]AV1451 require complex purifications to provide doses suitable for use in clinical imaging studies. To address this issue, we have modified the synthesis of [18F]AV1451 to use only 0.5 mg precursor, optimized the Boc-deprotection step and developed a simplified method for HPLC purification of the radiotracer. RESULTS An optimized [18F]AV1451 synthesis using a TRACERLab FXFN module led to high radiochemical yield (202 ± 57 mCi per synthesis) and doses with excellent radiochemical purity (98 ± 1%) and good specific activity (2521 ± 623 Ci/mmol). CONCLUSION An updated and operationally simple synthesis of [18F]AV1451 has been developed that is fully automated and prepares radiotracer doses suitable for use in clinical tau PET studies.
Collapse
Affiliation(s)
- Andrew V. Mossine
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Allen F. Brooks
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Bradford D. Henderson
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Brian G. Hockley
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Kirk A. Frey
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Peter J. H. Scott
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
49
|
Saint-Aubert L, Lemoine L, Chiotis K, Leuzy A, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging: present and future directions. Mol Neurodegener 2017; 12:19. [PMID: 28219440 PMCID: PMC5319037 DOI: 10.1186/s13024-017-0162-3] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Abnormal aggregation of tau in the brain is a major contributing factor in various neurodegenerative diseases. The role of tau phosphorylation in the pathophysiology of tauopathies remains unclear. Consequently, it is important to be able to accurately and specifically target tau deposits in vivo in the brains of patients. The advances of molecular imaging in the recent years have now led to the recent development of promising tau-specific tracers for positron emission tomography (PET), such as THK5317, THK5351, AV-1451, and PBB3. These tracers are now available for clinical assessment in patients with various tauopathies, including Alzheimer's disease, as well as in healthy subjects. Exploring the patterns of tau deposition in vivo for different pathologies will allow discrimination between neurodegenerative diseases, including different tauopathies, and monitoring of disease progression. The variety and complexity of the different types of tau deposits in the different diseases, however, has resulted in quite a challenge for the development of tau PET tracers. Extensive work remains in order to fully characterize the binding properties of the tau PET tracers, and to assess their usefulness as an early biomarker of the underlying pathology. In this review, we summarize recent findings on the most promising tau PET tracers to date, discuss what has been learnt from these findings, and offer some suggestions for the next steps that need to be achieved in a near future.
Collapse
Affiliation(s)
- Laure Saint-Aubert
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Laetitia Lemoine
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Konstantinos Chiotis
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Antoine Leuzy
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Elena Rodriguez-Vieitez
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Agneta Nordberg
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden. .,Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|