203 results on '"Tau imaging"'
Search Results
2. Is tau in the absence of amyloid on the Alzheimer’s continuum?: A study of discordant PET positivity
- Author
-
Weigand, Alexandra J, Bangen, Katherine J, Thomas, Kelsey R, Delano-Wood, Lisa, Gilbert, Paul E, Brickman, Adam M, Bondi, Mark W, and Initiative, Alzheimer’s Disease Neuroimaging
- Subjects
Biological Psychology ,Psychology ,Alzheimer's Disease Related Dementias (ADRD) ,Alzheimer's Disease ,Biomedical Imaging ,Neurodegenerative ,Clinical Research ,Prevention ,Aging ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Behavioral and Social Science ,Dementia ,Acquired Cognitive Impairment ,Basic Behavioral and Social Science ,Brain Disorders ,2.1 Biological and endogenous factors ,4.1 Discovery and preclinical testing of markers and technologies ,Neurological ,tau imaging ,amyloid imaging ,Alzheimer's disease ,mild cognitive impairment ,biomarkers ,Alzheimer’s Disease Neuroimaging Initiative ,Alzheimer’s disease ,Clinical sciences ,Biological psychology - Abstract
The amyloid cascade model of Alzheimer's disease posits the primacy of amyloid beta deposition preceding tau-mediated neurofibrillary tangle formation. The amyloid-tau-neurodegeneration biomarker-only diagnostic framework similarly requires the presence of amyloid beta for a diagnosis on the Alzheimer's continuum. However, medial temporal lobe tau pathology in the absence of amyloid beta is frequently observed at autopsy in cognitively normal individuals, a phenomenon that may reflect a consequence of aging and has been labelled 'primary age-related tauopathy'. Alternatively, others argue that this tauopathy reflects an early stage of the developmental continuum leading to Alzheimer's disease. We used positron emission tomography imaging to investigate amyloid beta and tau positivity and associations with cognition to better inform the conceptualization of biomarker changes in Alzheimer's pathogenesis. Five hundred twenty-three individuals from the Alzheimer's Disease Neuroimaging Initiative who had undergone flortaucipir positron emission tomography imaging were selected to derive positron emission tomography positivity thresholds using conditional inference decision tree regression. A subsample of 301 individuals without dementia (i.e. those with normal cognition or mild cognitive impairment) had also undergone florbetapir positron emission tomography imaging within 12 months and were categorized into one of the four groups based on cortical amyloid and Braak stage I/II tau positivity: A-/T-, A+/T-, A-/T+, or A+/T+. Tau positivity in the absence of amyloid beta positivity (i.e. A-/T+) comprised the largest group, representing 45% of the sample. In contrast, only 6% of the sample was identified as A+/T-, and the remainder of the sample fell into A-/T- (22%) or A+/T+ (27%) categories. A-/T- and A+/T- groups had the best cognitive performances across memory, language and executive function; the A-/T+ group showed small-to-moderate relative decreases in cognition; and the A+/T+ group had the worst cognitive performances. Furthermore, there were negative associations between Braak stage I/II tau values and all cognitive domains only in the A-/T+ and A+/T+ groups, with strongest associations for the A+/T+ group. Among our sample of older adults across the Alzheimer's pathological spectrum, 7-fold fewer individuals have positron emission tomography evidence of amyloid beta pathology in the absence of tau pathology than the converse, challenging prevailing models of amyloid beta's primacy in Alzheimer's pathogenesis. Given that cognitive performance in the A-/T+ group was poorer than in individuals without either pathology, our results suggest that medial temporal lobe tau without cortical amyloid beta may reflect an early stage on the Alzheimer's pathological continuum.
- Published
- 2020
3. 18F-flortaucipir (AV-1451) tau PET in frontotemporal dementia syndromes
- Author
-
Tsai, Richard M, Bejanin, Alexandre, Lesman-Segev, Orit, LaJoie, Renaud, Visani, Adrienne, Bourakova, Viktoriya, O’Neil, James P, Janabi, Mustafa, Baker, Suzanne, Lee, Suzee E, Perry, David C, Bajorek, Lynn, Karydas, Anna, Spina, Salvatore, Grinberg, Lea T, Seeley, William W, Ramos, Eliana M, Coppola, Giovanni, Gorno-Tempini, Maria Luisa, Miller, Bruce L, Rosen, Howard J, Jagust, William, Boxer, Adam L, and Rabinovici, Gil D
- Subjects
Biomedical and Clinical Sciences ,Health Sciences ,Brain Disorders ,Aging ,Frontotemporal Dementia (FTD) ,Aphasia ,Neurodegenerative ,Biomedical Imaging ,Acquired Cognitive Impairment ,Dementia ,Rare Diseases ,Neurosciences ,Alzheimer's Disease ,Alzheimer's Disease Related Dementias (ADRD) ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Genetics ,2.1 Biological and endogenous factors ,4.2 Evaluation of markers and technologies ,Neurological ,Adult ,Aged ,Aged ,80 and over ,Carbolines ,Cohort Studies ,Contrast Media ,Female ,Frontotemporal Dementia ,Humans ,Male ,Middle Aged ,Positron-Emission Tomography ,Young Adult ,tau Proteins ,Biomarkers ,Frontotemporal dementia ,Tau imaging ,Neuropathology ,Tau ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundThe tau positron emission tomography (PET) ligand 18F-flortaucipir binds to paired helical filaments of tau in aging and Alzheimer's disease (AD), but its utility in detecting tau aggregates in frontotemporal dementia (FTD) is uncertain.MethodsWe performed 18F-flortaucipir imaging in patients with the FTD syndromes (n = 45): nonfluent variant primary progressive aphasia (nfvPPA) (n = 11), corticobasal syndrome (CBS) (n = 10), behavioral variant frontotemporal dementia (bvFTD) (n = 10), semantic variant primary progressive aphasia (svPPA) (n = 2) and FTD associated pathogenic genetic mutations microtubule-associated protein tau (MAPT) (n = 6), chromosome 9 open reading frame 72 (C9ORF72) (n = 5), and progranulin (GRN) (n = 1). All patients underwent MRI and β-amyloid biomarker testing via 11C-PiB or cerebrospinal fluid. 18F-flortaucipir uptake in patients was compared to 53 β-amyloid negative normal controls using voxelwise and pre-specified region of interest approaches.ResultsOn qualitative assessment, patients with nfvPPA showed elevated 18F-flortacupir binding in the left greater than right inferior frontal gyrus. Patients with CBS showed elevated binding in frontal white matter, with higher cortical gray matter uptake in a subset of β-amyloid-positive patients. Five of ten patients with sporadic bvFTD demonstrated increased frontotemporal binding. MAPT mutation carriers had elevated 18F-flortaucipir retention primarily, but not exclusively, in mutations with Alzheimer's-like neurofibrillary tangles. However, tracer retention was also seen in patients with svPPA, and the mutations C9ORF72, GRN predicted to have TDP-43 pathology. Quantitative region-of-interest differences between patients and controls were seen only in inferior frontal gyrus in nfvPPA and left insula and bilateral temporal poles in MAPT carriers. No significant regional differences were found in CBS or sporadic bvFTD. Two patients underwent postmortem neuropathological examination. A patient with C9ORF72, TDP-43-type B pathology, and incidental co-pathology of scattered neurofibrillary tangles in the middle frontal, inferior temporal gyrus showed corresponding mild 18F-flortaucipir retention without additional uptake matching the widespread TDP-43 type B pathology. A patient with sporadic bvFTD demonstrated punctate inferior temporal and hippocampus tracer retention, corresponding to the area of severe argyrophilic grain disease pathology.Conclusions18F-flortaucipir in patients with FTD and predicted tauopathy or TDP-43 pathology demonstrated limited sensitivity and specificity. Further postmortem pathological confirmation and development of FTD tau-specific ligands are needed.
- Published
- 2019
4. Preliminary mechanistic insights of a brain-penetrant microtubule imaging PET ligand in a tau-knockout mouse model
- Author
-
Naresh Damuka, Miranda E. Orr, Avinash H. Bansode, Ivan Krizan, Mack Miller, Jillian Lee, Shannon L. Macauley, Christopher T. Whitlow, Akiva Mintz, Suzanne Craft, and Kiran Kumar Solingapuram Sai
- Subjects
Positron emission tomography (PET) ,Microtubules ,Alzheimer’s disease ,Biomarker ,Tau imaging ,And Biodistribution ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Background Microtubules (MTs) are critical for cell structure, function, and survival. MT instability may contribute to Alzheimer’s disease (AD) pathogenesis as evidenced by persistent negative regulation (phosphorylation) of the neuronal microtubule-associated protein tau. Hyperphosphorylated tau, not bound to MTs, forms intraneuronal pathology that correlates with dementia and can be tracked using positron emission tomography (PET) imaging. The contribution of MT instability in AD remains unknown, though it may be more proximal to neuronal dysfunction than tau accumulation. Our lab reported the first brain-penetrant MT-based PET ligand, [11C]MPC-6827, and its PET imaging with this ligand in normal rodents and non-human primates demonstrated high brain uptake and excellent pharmacokinetics. Target engagement and mechanism of action using in vitro, in vivo, and ex vivo methods were evaluated here. Methods In vitro cell uptake assay was performed in SH-SY5Y neuronal cells with [11C]MPC-6827, with various MT stabilizing and destabilizing agents. To validate the in vitro results, wild type (WT) mice (n = 4) treated with a brain-penetrant MT stabilizing drug (EpoD) underwent microPET/CT brain imaging with [11C]MPC-6827. To determine the influence of tau protein on radiotracer binding in the absence of protein accumulation, we utilized tau knockout (KO) mice. In vivo microPET imaging, ex vivo biodistribution, and autoradiography studies were performed in tau KO and WT mice (n = 6/group) with [11C]MPC-6827. Additionally, α, β, and acetylated tubulin levels in both brain samples were determined using commercially available cytoskeleton-based MT kit and capillary electrophoresis immunoblotting assays. Results Cell uptake demonstrated higher radioactive uptake with MT destabilizing agents and lower uptake with stabilizing agents compared to untreated cells. Similarly, acute treatment with EpoD in WT mice decreased [11C]MPC-6827 brain uptake, assessed with microPET/CT imaging. Compared to WT mice, tau KO mice expressed significantly lower β tubulin, which contains the MPC-6827 binding domain, and modestly lower levels of acetylated α tubulin, indicative of unstable MTs. In vivo imaging revealed significantly higher [11C]MPC-6827 uptake in tau KOs than WT, particularly in AD-relevant brain regions known to express high levels of tau. Ex vivo post-PET biodistribution and autoradiography confirmed the in vivo results. Conclusions Collectively, our data indicate that [11C]MPC-6827 uptake inversely correlates with MT stability and may better reflect the absence of tau than total tubulin levels. Given the radiotracer binding does not require the presence of aggregated tau, we hypothesize that [11C]MPC-6827 may be particularly useful in preclinical stages of AD prior to tau deposition. Our study provides immediate clarity on high uptake of the MT-based radiotracer in AD brains, which directly informs clinical utility in MT/tau-based PET imaging studies.
- Published
- 2022
- Full Text
- View/download PDF
5. Nuclear Imaging in Frontotemporal Dementia
- Author
-
Reesink, Fransje E., Stormezand, Gilles N., Dierckx, Rudi A. J. O., De Deyn, Peter Paul, Dierckx, Rudi A. J. O., editor, Otte, Andreas, editor, de Vries, Erik F. J., editor, van Waarde, Aren, editor, and Leenders, Klaus L., editor
- Published
- 2021
- Full Text
- View/download PDF
6. Local and distant relationships between amyloid, tau and neurodegeneration in Alzheimer's Disease
- Author
-
Iaccarino, Leonardo, Tammewar, Gautam, Ayakta, Nagehan, Baker, Suzanne L, Bejanin, Alexandre, Boxer, Adam L, Gorno-Tempini, Maria Luisa, Janabi, Mustafa, Kramer, Joel H, Lazaris, Andreas, Lockhart, Samuel N, Miller, Bruce L, Miller, Zachary A, O'Neil, James P, Ossenkoppele, Rik, Rosen, Howard J, Schonhaut, Daniel R, Jagust, William J, and Rabinovici, Gil D
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Acquired Cognitive Impairment ,Neurodegenerative ,Brain Disorders ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Aging ,Biomedical Imaging ,Neurosciences ,Dementia ,Alzheimer's Disease ,2.1 Biological and endogenous factors ,Neurological ,Aged ,Aged ,80 and over ,Alzheimer Disease ,Amyloid beta-Peptides ,Brain ,Female ,Humans ,Magnetic Resonance Imaging ,Male ,Middle Aged ,Neural Pathways ,Positron-Emission Tomography ,tau Proteins ,Amyloid imaging ,Tau imaging ,Brain atrophy ,Biological psychology ,Clinical and health psychology - Abstract
The relationships between β-amyloid (Aβ), tau and neurodegeneration within Alzheimer's Disease pathogenesis are not fully understood. To explore these associations in vivo, we evaluated 30 Aβ PET-positive patients (mean ± sd age 62.4 ± 8.3) with mild probable AD and 12 Aβ PET-negative healthy controls (HC) (mean ± sd age 77.3 ± 6.9) as comparison. All participants underwent 3 T MRI, 11C-PiB (Aβ) PET and 18F-AV1451 (tau) PET. Multimodal correlation analyses were run at both voxel- and region-of-interest levels. 11C-PiB retention in AD showed the most diffuse uptake pattern throughout association neocortex, whereas 18F-AV1451 and gray matter volume reduction (GMR) showed a progressive predilection for posterior cortices (p
- Published
- 2018
7. The Alzheimer's Disease Neuroimaging Initiative 3: Continued innovation for clinical trial improvement.
- Author
-
Weiner, Michael W, Veitch, Dallas P, Aisen, Paul S, Beckett, Laurel A, Cairns, Nigel J, Green, Robert C, Harvey, Danielle, Jack, Clifford R, Jagust, William, Morris, John C, Petersen, Ronald C, Salazar, Jennifer, Saykin, Andrew J, Shaw, Leslie M, Toga, Arthur W, Trojanowski, John Q, and Alzheimer's Disease Neuroimaging Initiative
- Subjects
Alzheimer's Disease Neuroimaging Initiative ,Brain ,Humans ,Alzheimer Disease ,Disease Progression ,Positron-Emission Tomography ,Magnetic Resonance Imaging ,Radionuclide Imaging ,Clinical Trials as Topic ,Neuroimaging ,Biomarkers ,Alzheimer's disease ,Amyloid phenotyping ,Brain Health Registry ,Centiloid method ,Clinical trial biomarkers ,Functional connectivity ,Tau imaging ,Geriatrics ,Neurosciences ,Clinical Sciences - Abstract
IntroductionThe overall goal of the Alzheimer's Disease Neuroimaging Initiative (ADNI) is to validate biomarkers for Alzheimer's disease (AD) clinical trials. ADNI-3, which began on August 1, 2016, is a 5-year renewal of the current ADNI-2 study.MethodsADNI-3 will follow current and additional subjects with normal cognition, mild cognitive impairment, and AD using innovative technologies such as tau imaging, magnetic resonance imaging sequences for connectivity analyses, and a highly automated immunoassay platform and mass spectroscopy approach for cerebrospinal fluid biomarker analysis. A Systems Biology/pathway approach will be used to identify genetic factors for subject selection/enrichment. Amyloid positron emission tomography scanning will be standardized using the Centiloid method. The Brain Health Registry will help recruit subjects and monitor subject cognition.ResultsMultimodal analyses will provide insight into AD pathophysiology and disease progression.DiscussionADNI-3 will aim to inform AD treatment trials and facilitate development of AD disease-modifying treatments.
- Published
- 2017
8. SPECT and PET of the Brain
- Author
-
Kimura, Yasuyuki, Kato, Takashi, Ito, Kengo, Ichise, Masanori, Ahmadzadehfar, Hojjat, editor, Biersack, Hans-Jürgen, editor, Freeman, Leonard M., editor, and Zuckier, Lionel S., editor
- Published
- 2020
- Full Text
- View/download PDF
9. Preliminary mechanistic insights of a brain-penetrant microtubule imaging PET ligand in a tau-knockout mouse model.
- Author
-
Damuka, Naresh, Orr, Miranda E., Bansode, Avinash H., Krizan, Ivan, Miller, Mack, Lee, Jillian, Macauley, Shannon L., Whitlow, Christopher T., Mintz, Akiva, Craft, Suzanne, and Solingapuram Sai, Kiran Kumar
- Subjects
TAU proteins ,POSITRON emission tomography ,CAPILLARY electrophoresis ,LABORATORY mice ,MICROTUBULES ,COMPUTED tomography ,ANIMAL disease models ,PHOSPHORYLATION - Abstract
Background: Microtubules (MTs) are critical for cell structure, function, and survival. MT instability may contribute to Alzheimer's disease (AD) pathogenesis as evidenced by persistent negative regulation (phosphorylation) of the neuronal microtubule-associated protein tau. Hyperphosphorylated tau, not bound to MTs, forms intraneuronal pathology that correlates with dementia and can be tracked using positron emission tomography (PET) imaging. The contribution of MT instability in AD remains unknown, though it may be more proximal to neuronal dysfunction than tau accumulation. Our lab reported the first brain-penetrant MT-based PET ligand, [
11 C]MPC-6827, and its PET imaging with this ligand in normal rodents and non-human primates demonstrated high brain uptake and excellent pharmacokinetics. Target engagement and mechanism of action using in vitro, in vivo, and ex vivo methods were evaluated here. Methods: In vitro cell uptake assay was performed in SH-SY5Y neuronal cells with [11 C]MPC-6827, with various MT stabilizing and destabilizing agents. To validate the in vitro results, wild type (WT) mice (n = 4) treated with a brain-penetrant MT stabilizing drug (EpoD) underwent microPET/CT brain imaging with [11 C]MPC-6827. To determine the influence of tau protein on radiotracer binding in the absence of protein accumulation, we utilized tau knockout (KO) mice. In vivo microPET imaging, ex vivo biodistribution, and autoradiography studies were performed in tau KO and WT mice (n = 6/group) with [11 C]MPC-6827. Additionally, α, β, and acetylated tubulin levels in both brain samples were determined using commercially available cytoskeleton-based MT kit and capillary electrophoresis immunoblotting assays. Results: Cell uptake demonstrated higher radioactive uptake with MT destabilizing agents and lower uptake with stabilizing agents compared to untreated cells. Similarly, acute treatment with EpoD in WT mice decreased [11 C]MPC-6827 brain uptake, assessed with microPET/CT imaging. Compared to WT mice, tau KO mice expressed significantly lower β tubulin, which contains the MPC-6827 binding domain, and modestly lower levels of acetylated α tubulin, indicative of unstable MTs. In vivo imaging revealed significantly higher [11 C]MPC-6827 uptake in tau KOs than WT, particularly in AD-relevant brain regions known to express high levels of tau. Ex vivo post-PET biodistribution and autoradiography confirmed the in vivo results. Conclusions: Collectively, our data indicate that [11 C]MPC-6827 uptake inversely correlates with MT stability and may better reflect the absence of tau than total tubulin levels. Given the radiotracer binding does not require the presence of aggregated tau, we hypothesize that [11 C]MPC-6827 may be particularly useful in preclinical stages of AD prior to tau deposition. Our study provides immediate clarity on high uptake of the MT-based radiotracer in AD brains, which directly informs clinical utility in MT/tau-based PET imaging studies. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
10. A dual-time-window protocol to reduce acquisition time of dynamic tau PET imaging using [18F]MK-6240
- Author
-
Guilherme D. Kolinger, David Vállez García, Talakad G. Lohith, Eric D. Hostetler, Cyrille Sur, Arie Struyk, Ronald Boellaard, and Michel Koole
- Subjects
[18F]MK-6240 ,Dual-time-window ,PET quantification ,Reference logan ,Tau imaging ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Background [18F]MK-6240 is a PET tracer with sub-nanomolar affinity for neurofibrillary tangles. Therefore, tau quantification is possible with [18F]MK-6240 PET/CT scans, and it can be used for assessment of Alzheimer’s disease. However, long acquisition scans are required to provide fully quantitative estimates of pharmacokinetic parameters. Therefore, on the present study, dual-time-window (DTW) acquisitions was simulated to reduce PET/CT acquisition time, while taking into consideration perfusion changes and possible scanning protocol non-compliance. To that end, time activity curves (TACs) representing a 120-min acquisition (TAC120) were simulated using a two-tissue compartment model with metabolite corrected arterial input function from 90-min dynamic [18F]MK-6240 PET scans of three healthy control subjects and five subjects with mild cognitive impairment or Alzheimer’s disease. Therefore, TACs corresponding to different levels of specific binding were generated and then various perfusion changes were simulated. Next, DTW acquisitions were simulated consisting of an acquisition starting at tracer injection, a break and a second acquisition starting at 90 min post-injection. Finally, non-compliance with the PET/CT scanning protocol were simulated to assess its impact on quantification. All TACs were quantified using reference Logan’s distribution volume ratio (DVR) and standardized uptake value ratio (SUVR90) using the cerebellar cortex as reference region. Results It was found that DVR from a DTW protocol with a 60-min break between two 30-min dynamic scans closely approximates the DVR from the uninterrupted TAC120, with a regional bias smaller than 2.5%. Moreover, SUVR90 estimates were more susceptible (regional bias ≤ 19%) to changes in perfusion compared to DVR from a DTW TAC (regional bias ≤ 10%). Similarly, SUVR90 was affected by late-time scanning protocol delays reaching an increase of 8% for a 20-min delay, while DVR was not affected (regional bias
- Published
- 2021
- Full Text
- View/download PDF
11. Colocalization of Tau but Not β-Amyloid with Cortical Superficial Siderosis in a Case with Probable CAA
- Author
-
Matthias Brendel, Cihan Catak, Leonie Beyer, Jennifer Linn, Hannes Wahl, Daniel Janowitz, Axel Rominger, Marianne Patt, Henryk Barthel, Osama Sabri, Peter Bartenstein, and Frank Arne Wollenweber
- Subjects
cortical superficial siderosis ,tau imaging ,amyloid imaging ,cerebral amyloid angiopathy ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Cortical superficial siderosis (cSS) is a common feature in patients with cerebral amyloid angiopathy (CAA). The correlation between β-amyloid and/or tau pathology and the occurrence of cSS is unclear. We report on an 80-year-old male patient who was diagnosed with probable CAA according to modified Boston criteria and underwent longitudinal magnetic resonance imaging, amyloid positron emission tomography (PET), and additional tau PET imaging. Amyloid deposition presented predominantly in the contralateral hemisphere not affected by cSS. In contrast, tau deposition was predominantly overlapping with brain regions affected by cSS. Amyloid deposition was not different in the vicinity of cSS whereas tau depositions were elevated in the vicinity of CSS-affected regions compared to non-cSS-affected brain regions. This case of probable CAA suggests that cSS may be associated with a locally elevated tau pathology but not with increased fibrillary amyloid deposition.
- Published
- 2020
- Full Text
- View/download PDF
12. Plasma p217+tau versus NAV4694 amyloid and MK6240 tau PET across the Alzheimer's continuum.
- Author
-
Doré, Vincent, Doecke, James D., Saad, Ziad S., Triana‐Baltzer, Gallen, Slemmon, Randy, Krishnadas, Natasha, Bourgeat, Pierrick, Huang, Kun, Burnham, Samantha, Fowler, Christopher, Rainey‐Smith, Stephanie R., Bush, Ashley I., Ward, Larry, Robertson, Jo, Martins, Ralph N., Masters, Colin L., Villemagne, Victor L., Fripp, Jurgen, Kolb, Hartmuth C., and Rowe, Christopher C.
- Subjects
ALZHEIMER'S disease ,TAU proteins ,POSITRON emission tomography ,AMYLOID - Abstract
Introduction: We evaluated a new Simoa plasma assay for phosphorylated tau (P‐tau) at aa217 enhanced by additional p‐tau sites (p217+tau). Methods: Plasma p217+tau levels were compared to 18F‐NAV4694 amyloid beta (Aβ) positron emission tomography (PET) and 18F‐MK6240 tau PET in 174 cognitively impaired (CI) and 223 cognitively unimpaired (CU) participants. Results: Compared to Aβ− CU, the plasma levels of p217+tau increased 2‐fold in Aβ+ CU and 3.5‐fold in Aβ+ CI. In Aβ− the p217+tau levels did not differ significantly between CU and CI. P217+tau correlated with Aβ centiloids P =.67 (CI, P =.64; CU, P =.45) and tau SUVRMTP =.63 (CI, P =.69; CU, P =.34). Area under curve (AUC) for Alzheimer's disease (AD) dementia versus Aβ− CU was 0.94, for AD dementia versus other dementia was 0.93, for Aβ+ versus Aβ− PET was 0.89, and for tau+ versus tau− PET was 0.89. Discussion: Plasma p217+tau levels elevate early in the AD continuum and correlate well with Aβ and tau PET. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Impact of the Alzheimer's Disease Neuroimaging Initiative, 2004 to 2014
- Author
-
Weiner, Michael W, Veitch, Dallas P, Aisen, Paul S, Beckett, Laurel A, Cairns, Nigel J, Cedarbaum, Jesse, Donohue, Michael C, Green, Robert C, Harvey, Danielle, Jack, Clifford R, Jagust, William, Morris, John C, Petersen, Ronald C, Saykin, Andrew J, Shaw, Leslie, Thompson, Paul M, Toga, Arthur W, Trojanowski, John Q, and Initiative, Alzheimer's Disease Neuroimaging
- Subjects
Biomedical and Clinical Sciences ,Biological Psychology ,Clinical Sciences ,Neurosciences ,Psychology ,Prevention ,Brain Disorders ,Clinical Research ,Aging ,Acquired Cognitive Impairment ,Dementia ,Alzheimer's Disease ,Clinical Trials and Supportive Activities ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Neurodegenerative ,4.1 Discovery and preclinical testing of markers and technologies ,2.1 Biological and endogenous factors ,Neurological ,Alzheimer Disease ,Biomarkers ,Clinical Trials as Topic ,Databases ,Bibliographic ,Disease Progression ,Humans ,Longitudinal Studies ,Neuroimaging ,Alzheimer's disease ,Data-sharing ,Amyloid phenotyping ,Clinical trial biomarkers ,Tau imaging ,AD biomarker signature ,Worldwide ADNI ,Alzheimer's Disease Neuroimaging Initiative ,Geriatrics ,Clinical sciences ,Biological psychology - Abstract
IntroductionThe Alzheimer's Disease Neuroimaging Initiative (ADNI) was established in 2004 to facilitate the development of effective treatments for Alzheimer's disease (AD) by validating biomarkers for AD clinical trials.MethodsWe searched for ADNI publications using established methods.ResultsADNI has (1) developed standardized biomarkers for use in clinical trial subject selection and as surrogate outcome measures; (2) standardized protocols for use across multiple centers; (3) initiated worldwide ADNI; (4) inspired initiatives investigating traumatic brain injury and post-traumatic stress disorder in military populations, and depression, respectively, as an AD risk factor; (5) acted as a data-sharing model; (6) generated data used in over 600 publications, leading to the identification of novel AD risk alleles, and an understanding of the relationship between biomarkers and AD progression; and (7) inspired other public-private partnerships developing biomarkers for Parkinson's disease and multiple sclerosis.DiscussionADNI has made myriad impacts in its first decade. A competitive renewal of the project in 2015 would see the use of newly developed tau imaging ligands, and the continued development of recruitment strategies and outcome measures for clinical trials.
- Published
- 2015
14. In vivo characterization of chronic traumatic encephalopathy using [F-18]FDDNP PET brain imaging
- Author
-
Barrio, Jorge R, Small, Gary W, Wong, Koon-Pong, Huang, Sung-Cheng, Liu, Jie, Merrill, David A, Giza, Christopher C, Fitzsimmons, Robert P, Omalu, Bennet, Bailes, Julian, and Kepe, Vladimir
- Subjects
Biological Psychology ,Psychology ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Biomedical Imaging ,Dementia ,Acquired Cognitive Impairment ,Neurosciences ,Neurodegenerative ,Clinical Research ,Aging ,Basic Behavioral and Social Science ,Brain Disorders ,Traumatic Head and Spine Injury ,Behavioral and Social Science ,Traumatic Brain Injury (TBI) ,Physical Injury - Accidents and Adverse Effects ,Minority Health ,Alzheimer's Disease ,Neurological ,Adult ,Aged ,Aged ,80 and over ,Alzheimer Disease ,Amygdala ,Autopsy ,Brain ,Brain Injury ,Chronic ,Case-Control Studies ,Demography ,Humans ,Male ,Mesencephalon ,Middle Aged ,Nitriles ,Positron-Emission Tomography ,traumatic brain injury ,chronic traumatic encephalopathy ,[F-18]FDDNP PET ,tau imaging ,concussions - Abstract
Chronic traumatic encephalopathy (CTE) is an acquired primary tauopathy with a variety of cognitive, behavioral, and motor symptoms linked to cumulative brain damage sustained from single, episodic, or repetitive traumatic brain injury (TBI). No definitive clinical diagnosis for this condition exists. In this work, we used [F-18]FDDNP PET to detect brain patterns of neuropathology distribution in retired professional American football players with suspected CTE (n = 14) and compared results with those of cognitively intact controls (n = 28) and patients with Alzheimer's dementia (AD) (n = 24), a disease that has been cognitively associated with CTE. [F-18]FDDNP PET imaging results in the retired players suggested the presence of neuropathological patterns consistent with models of concussion wherein brainstem white matter tracts undergo early axonal damage and cumulative axonal injuries along subcortical, limbic, and cortical brain circuitries supporting mood, emotions, and behavior. This deposition pattern is distinctively different from the progressive pattern of neuropathology [paired helical filament (PHF)-tau and amyloid-β] in AD, which typically begins in the medial temporal lobe progressing along the cortical default mode network, with no or minimal involvement of subcortical structures. This particular [F-18]FDDNP PET imaging pattern in cases of suspected CTE also is primarily consistent with PHF-tau distribution observed at autopsy in subjects with a history of mild TBI and autopsy-confirmed diagnosis of CTE.
- Published
- 2015
15. Brain network alterations in individuals with and without mild cognitive impairment: parallel independent component analysis of AV1451 and AV45 positron emission tomography
- Author
-
Yuan Li, Zhijun Yao, Yue Yu, Ying Zou, Yu Fu, Bin Hu, and for the Alzheimer’s Disease Neuroimaging Initiative
- Subjects
Parallel independent component analysis, multivariate data analysis ,Amyloid imaging ,Tau imaging ,Networks ,Psychiatry ,RC435-571 - Abstract
Abstract Background Amyloid β (Aβ) and tau proteins are considered as critical factors that affect Alzheimer’s disease (AD) and mild cognitive impairment (MCI). Although many studies have conducted on these two proteins, little study has investigated the relationship between their spatial distributions. This study aims to explore the associations of spatial patterns between Aβ deposition and tau deposition in patients with MCI and normal control (NC). Methods We used multimodality positron emission tomography (PET) data from a clinically heterogeneous population of patients with MCI and NC. All data were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database containing information of 65 patients with MCI and 75 NC who both had undergone AV45 (Aβ) and AV1451 (tau) PET. To assess the spatial distribution of Aβ and tau deposition, we employed parallel independent component analysis (pICA), which enabled the joint analysis of multimodal imaging data. pICA was conducted to identify the significant difference and correlation relationship of brain networks between Aβ PET and tau PET in MCI and NC groups. Results Our results revealed the strongly correlated network between Aβ PET and tau PET were colocalized with the default-mode network (DMN). Simultaneously, in comparison of the spatial distribution between Aβ PET and tau PET, it was found that the significant differences between MCI and NC were mainly distributed in DMN, cognitive control network and visual networks. The altered brain networks obtained from pICA analysis are consistent with the abnormalities of brain network in MCI patients. Conclusions Findings suggested the abnormal spatial distribution regions of tau PET were correlated with the abnormal spatial distribution regions of Aβ PET, and both of which were located in DMN network. This study revealed that combining pICA with multimodal imaging data is an effective approach for distinguishing MCI patients from NC group.
- Published
- 2019
- Full Text
- View/download PDF
16. Relationship between amyloid and tau levels and its impact on tau spreading.
- Author
-
Doré, Vincent, Krishnadas, Natasha, Bourgeat, Pierrick, Huang, Kun, Li, Shenpeng, Burnham, Samantha, Masters, Colin L., Fripp, Jurgen, Villemagne, Victor L., and Rowe, Christopher C.
- Subjects
- *
ENTORHINAL cortex , *TEMPORAL lobe , *AMYLOID , *NEOCORTEX , *POSITRON emission tomography , *HIPPOCAMPUS (Brain) - Abstract
Purpose: Previous studies have shown that Aβ-amyloid (Aβ) likely promotes tau to spread beyond the medial temporal lobe. However, the Aβ levels necessary for tau to spread in the neocortex is still unclear. Methods: Four hundred sixty-six participants underwent tau imaging with [18F]MK6420 and Aβ imaging with [18F]NAV4694. Aβ scans were quantified on the Centiloid (CL) scale with a cut-off of 25 CL for abnormal levels of Aβ (A+). Tau scans were quantified in three regions of interest (ROI) (mesial temporal (Me); temporoparietal neocortex (Te); and rest of neocortex (R)) and four mesial temporal region (entorhinal cortex, amygdala, hippocampus, and parahippocampus). Regional tau thresholds were established as the 95%ile of the cognitively unimpaired A- subjects. The prevalence of abnormal tau levels (T+) along the Centiloid continuum was determined. Results: The plots of prevalence of T+ show earlier and greater increase along the Centiloid continuum in the medial temporal area compared to neocortex. Prevalence of T+ was low but associated with Aβ level between 10 and 40 CL reaching 23% in Me, 15% in Te, and 11% in R. Between 40 and 70 CL, the prevalence of T+ subjects per CL increased fourfold faster and at 70 CL was 64% in Me, 51% in Te, and 37% in R. In cognitively unimpaired, there were no T+ in R below 50 CL. The highest prevalence of T+ were found in the entorhinal cortex, reaching 40% at 40 CL and 80% at 60 CL. Conclusion: Outside the entorhinal cortex, abnormal levels of cortical tau on PET are rarely found with Aβ below 40 CL. Above 40 CL prevalence of T+ accelerates in all areas. Moderate Aβ levels are required before abnormal neocortical tau becomes detectable. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. A dual-time-window protocol to reduce acquisition time of dynamic tau PET imaging using [18F]MK-6240.
- Author
-
Kolinger, Guilherme D., Vállez García, David, Lohith, Talakad G., Hostetler, Eric D., Sur, Cyrille, Struyk, Arie, Boellaard, Ronald, and Koole, Michel
- Subjects
POSITRON emission tomography ,COMPUTED tomography ,PERFUSION ,CEREBELLAR cortex ,MILD cognitive impairment ,NEUROFIBRILLARY tangles ,ALZHEIMER'S disease - Abstract
Background: [
18 F]MK-6240 is a PET tracer with sub-nanomolar affinity for neurofibrillary tangles. Therefore, tau quantification is possible with [18 F]MK-6240 PET/CT scans, and it can be used for assessment of Alzheimer's disease. However, long acquisition scans are required to provide fully quantitative estimates of pharmacokinetic parameters. Therefore, on the present study, dual-time-window (DTW) acquisitions was simulated to reduce PET/CT acquisition time, while taking into consideration perfusion changes and possible scanning protocol non-compliance. To that end, time activity curves (TACs) representing a 120-min acquisition (TAC120 ) were simulated using a two-tissue compartment model with metabolite corrected arterial input function from 90-min dynamic [18 F]MK-6240 PET scans of three healthy control subjects and five subjects with mild cognitive impairment or Alzheimer's disease. Therefore, TACs corresponding to different levels of specific binding were generated and then various perfusion changes were simulated. Next, DTW acquisitions were simulated consisting of an acquisition starting at tracer injection, a break and a second acquisition starting at 90 min post-injection. Finally, non-compliance with the PET/CT scanning protocol were simulated to assess its impact on quantification. All TACs were quantified using reference Logan's distribution volume ratio (DVR) and standardized uptake value ratio (SUVR90 ) using the cerebellar cortex as reference region. Results: It was found that DVR from a DTW protocol with a 60-min break between two 30-min dynamic scans closely approximates the DVR from the uninterrupted TAC120 , with a regional bias smaller than 2.5%. Moreover, SUVR90 estimates were more susceptible (regional bias ≤ 19%) to changes in perfusion compared to DVR from a DTW TAC (regional bias ≤ 10%). Similarly, SUVR90 was affected by late-time scanning protocol delays reaching an increase of 8% for a 20-min delay, while DVR was not affected (regional bias < 1.5%) by DTW protocol non-compliance. Conclusions: Therefore, such DTW protocol has the potential to increase patient comfort and throughput without compromising quantitative accuracy and is more reliable against SUVR in terms of perfusion changes and protocol deviations, which could prove beneficial for drug effect assessment and patient follow-up using longitudinal [18 F]MK-6240 PET imaging. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
18. Characterization of MK6240, a tau PET tracer, in autopsy brain tissue from Alzheimer's disease cases.
- Author
-
Malarte, Mona-Lisa, Nordberg, Agneta, and Lemoine, Laetitia
- Subjects
- *
ALZHEIMER'S disease , *AUTOPSY , *BINDING site assay , *ALZHEIMER'S patients , *BRAIN abscess , *NEUROFIBRILLARY tangles , *BINDING sites - Abstract
Purpose: MK6240 is a second-generation tau PET tracer designed to detect the neurofibrillary tangles in the brains of patients with Alzheimer's disease (AD). The aim of the study was to characterize 3H-MK6240 in AD and control brain tissue and to compare its binding properties with those of first-generation tau PET tracers. Methods: Saturation binding assays with 3H-MK6240 were carried out in the temporal and parietal cortices of AD brains to determine the maximum number of binding sites (Bmax) and the dissociation constants (Kd) at these sites. Competitive binding assays were carried out between 3H-MK6240 and unlabelled MK6240, AV-1451 (aka T807, flortaucipir) and THK5117, and between 3H-THK5351 and unlabelled MK6240. Regional binding studies with 3H-MK6240 were carried out in homogenates from six AD and seven control brains and, using autoradiography, on large frozen sections from two AD brains and one control brain. Results: The saturation binding assays gave Bmax and Kd values of 59.2 fmol/mg and 0.32 nM in the temporal cortex and 154.7 fmol/mg and 0.15 nM in the parietal cortex. The competitive binding assays revealed two binding sites with affinities in the picomolar and nanomolar range shared by 3H-MK6240 and all the tested unlabelled compounds. There were no binding sites in common between 3H-THK5351 and unlabelled MK6240. Regional binding of 3H-MK6240 was significantly higher in AD brain tissue than in controls. Binding in brain tissue from AD patients with early-onset AD was significantly higher than in brain tissue from patients with late-onset AD. Binding of 3H-MK6240 was not observed in off-target regions. Autoradiography showed high regional cortical binding in the two AD brains and very low binding in the control brain. Conclusions: 3H-MK6240 has a high binding affinity for tau deposits in AD brain tissue but also has different binding characteristics from those of the first-generation tau tracers. This confirms the complexity of tau tracer binding on tau deposits with different binding affinities for different binding sites. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. Relationships Between Cognition and Neuropathological Tau in Alzheimer's Disease Assessed by 18F Flortaucipir PET.
- Author
-
Devous Sr., Michael D., Fleisher, Adam S., Pontecorvo, Michael J., Lu, Ming, Siderowf, Andrew, Navitsky, Michael, Kennedy, Ian, Southekal, Sudeepti, Harris, Thomas S., Mintun, Mark A., Devous, Michael D, and Devous, Michael D Sr
- Subjects
- *
ALZHEIMER'S disease , *TAU proteins , *POSITRON emission tomography , *TRAIL Making Test , *COGNITION disorders , *PYRIDINE , *RESEARCH , *NEURONS , *NERVE tissue proteins , *CLINICAL trials , *WECHSLER Memory Scale , *RESEARCH methodology , *COGNITION , *MEDICAL cooperation , *EVALUATION research , *COMPARATIVE studies , *QUESTIONNAIRES - Abstract
Background: Tau neurofibrillary tangle burden increases with Alzheimer's disease (AD) stage and correlates with degree of cognitive impairment. Tau PET imaging could facilitate understanding the relationship between tau pathology and cognitive impairment.Objective: Evaluate the relationship between 18F flortaucipir uptake patterns and cognition across multiple cognitive domains.Methods: We acquired flortaucipir PET scans in 84 amyloid-positive control, mild cognitive impairment (MCI), and AD subjects. Flortaucipir standardized uptake value ratio (SUVr) values were obtained from a neocortical volume of interest (VOI), a precuneus VOI, and VOIs defined by the correlation between flortaucipir SUVr images and domain-specific cognitive tests. Cognitive assessments included Mini-Mental State Exam (MMSE), Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-cog), and a neuropsychological test battery (i.e., Wechsler Memory Scale-Revised Logical Memory (WMS-R), Trail Making Test, Boston Naming Test, Digit Symbol Substitution Test, Animal List Generation, WMS-R Digit Span, American National Adult Reading Test, Clock Drawing Test, Judgment of Line Orientation, and WMS-R Logical Memory II (Delayed Recall)) and the Functional Activities Questionnaire (FAQ). Correlation analyses compared regional and voxel-wise VOIs to cognitive scores.Results: Subjects included 5 controls, 47 MCI, and 32 AD subjects. Significant correlations were seen between both flortaucipir and florbetapir SUVrs and MMSE, ADAS-Cog, and FAQ. Cognitive impairment was associated with increased flortaucipir uptake in regionally specific patterns consistent with the neuroanatomy underlying specific cognitive tests.Conclusion: Flortaucipir SUVr values demonstrated significant inverse correlations with cognitive scores in domain-specific patterns. Findings support the hypothesis that PET imaging of neuropathologic tau deposits may reflect underlying neurodegeneration in AD. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
20. 18F-flortaucipir (AV-1451) tau PET in frontotemporal dementia syndromes
- Author
-
Richard M. Tsai, Alexandre Bejanin, Orit Lesman-Segev, Renaud LaJoie, Adrienne Visani, Viktoriya Bourakova, James P. O’Neil, Mustafa Janabi, Suzanne Baker, Suzee E. Lee, David C. Perry, Lynn Bajorek, Anna Karydas, Salvatore Spina, Lea T. Grinberg, William W. Seeley, Eliana M. Ramos, Giovanni Coppola, Maria Luisa Gorno-Tempini, Bruce L. Miller, Howard J. Rosen, William Jagust, Adam L. Boxer, and Gil D. Rabinovici
- Subjects
Biomarkers ,Frontotemporal dementia ,Tau imaging ,Neuropathology ,Tau ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The tau positron emission tomography (PET) ligand 18F-flortaucipir binds to paired helical filaments of tau in aging and Alzheimer’s disease (AD), but its utility in detecting tau aggregates in frontotemporal dementia (FTD) is uncertain. Methods We performed 18F-flortaucipir imaging in patients with the FTD syndromes (n = 45): nonfluent variant primary progressive aphasia (nfvPPA) (n = 11), corticobasal syndrome (CBS) (n = 10), behavioral variant frontotemporal dementia (bvFTD) (n = 10), semantic variant primary progressive aphasia (svPPA) (n = 2) and FTD associated pathogenic genetic mutations microtubule-associated protein tau (MAPT) (n = 6), chromosome 9 open reading frame 72 (C9ORF72) (n = 5), and progranulin (GRN) (n = 1). All patients underwent MRI and β-amyloid biomarker testing via 11C-PiB or cerebrospinal fluid. 18F-flortaucipir uptake in patients was compared to 53 β-amyloid negative normal controls using voxelwise and pre-specified region of interest approaches. Results On qualitative assessment, patients with nfvPPA showed elevated 18F-flortacupir binding in the left greater than right inferior frontal gyrus. Patients with CBS showed elevated binding in frontal white matter, with higher cortical gray matter uptake in a subset of β-amyloid-positive patients. Five of ten patients with sporadic bvFTD demonstrated increased frontotemporal binding. MAPT mutation carriers had elevated 18F-flortaucipir retention primarily, but not exclusively, in mutations with Alzheimer’s-like neurofibrillary tangles. However, tracer retention was also seen in patients with svPPA, and the mutations C9ORF72, GRN predicted to have TDP-43 pathology. Quantitative region-of-interest differences between patients and controls were seen only in inferior frontal gyrus in nfvPPA and left insula and bilateral temporal poles in MAPT carriers. No significant regional differences were found in CBS or sporadic bvFTD. Two patients underwent postmortem neuropathological examination. A patient with C9ORF72, TDP-43-type B pathology, and incidental co-pathology of scattered neurofibrillary tangles in the middle frontal, inferior temporal gyrus showed corresponding mild 18F-flortaucipir retention without additional uptake matching the widespread TDP-43 type B pathology. A patient with sporadic bvFTD demonstrated punctate inferior temporal and hippocampus tracer retention, corresponding to the area of severe argyrophilic grain disease pathology. Conclusions 18F-flortaucipir in patients with FTD and predicted tauopathy or TDP-43 pathology demonstrated limited sensitivity and specificity. Further postmortem pathological confirmation and development of FTD tau-specific ligands are needed.
- Published
- 2019
- Full Text
- View/download PDF
21. Neuroinflammation and protein aggregation co-localize across the frontotemporal dementia spectrum.
- Author
-
Bevan-Jones, W Richard, Cope, Thomas E, Jones, P Simon, Kaalund, Sanne S, Passamonti, Luca, Allinson, Kieren, Green, Oliver, Hong, Young T, Fryer, Tim D, Arnold, Robert, Coles, Jonathan P, Aigbirhio, Franklin I, Larner, Andrew J, Patterson, Karalyn, O'Brien, John T, and Rowe, James B
- Subjects
- *
CELL metabolism , *PROTEINS , *PYRIDINE , *RESEARCH , *INFLAMMATION , *RESEARCH methodology , *ISOQUINOLINE , *CASE-control method , *RADIOISOTOPES , *EVALUATION research , *MEDICAL cooperation , *COMPARATIVE studies , *DNA-binding proteins , *RESEARCH funding , *FRONTOTEMPORAL dementia , *NEURODEGENERATION , *DISEASE complications - Abstract
The clinical syndromes of frontotemporal dementia are clinically and neuropathologically heterogeneous, but processes such as neuroinflammation may be common across the disease spectrum. We investigated how neuroinflammation relates to the localization of tau and TDP-43 pathology, and to the heterogeneity of clinical disease. We used PET in vivo with (i) 11C-PK-11195, a marker of activated microglia and a proxy index of neuroinflammation; and (ii) 18F-AV-1451, a radioligand with increased binding to pathologically affected regions in tauopathies and TDP-43-related disease, and which is used as a surrogate marker of non-amyloid-β protein aggregation. We assessed 31 patients with frontotemporal dementia (10 with behavioural variant, 11 with the semantic variant and 10 with the non-fluent variant), 28 of whom underwent both 18F-AV-1451 and 11C-PK-11195 PET, and matched control subjects (14 for 18F-AV-1451 and 15 for 11C-PK-11195). We used a univariate region of interest analysis, a paired correlation analysis of the regional relationship between binding distributions of the two ligands, a principal component analysis of the spatial distributions of binding, and a multivariate analysis of the distribution of binding that explicitly controls for individual differences in ligand affinity for TDP-43 and different tau isoforms. We found significant group-wise differences in 11C-PK-11195 binding between each patient group and controls in frontotemporal regions, in both a regions-of-interest analysis and in the comparison of principal spatial components of binding. 18F-AV-1451 binding was increased in semantic variant primary progressive aphasia compared to controls in the temporal regions, and both semantic variant primary progressive aphasia and behavioural variant frontotemporal dementia differed from controls in the expression of principal spatial components of binding, across temporal and frontotemporal cortex, respectively. There was a strong positive correlation between 11C-PK-11195 and 18F-AV-1451 uptake in all disease groups, across widespread cortical regions. We confirmed this association with post-mortem quantification in 12 brains, demonstrating strong associations between the regional densities of microglia and neuropathology in FTLD-TDP (A), FTLD-TDP (C), and FTLD-Pick's. This was driven by amoeboid (activated) microglia, with no change in the density of ramified (sessile) microglia. The multivariate distribution of 11C-PK-11195 binding related better to clinical heterogeneity than did 18F-AV-1451: distinct spatial modes of neuroinflammation were associated with different frontotemporal dementia syndromes and supported accurate classification of participants. These in vivo findings indicate a close association between neuroinflammation and protein aggregation in frontotemporal dementia. The inflammatory component may be important in shaping the clinical and neuropathological patterns of the diverse clinical syndromes of frontotemporal dementia. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. An updated radiosynthesis of [18F]AV1451 for tau PET imaging
- Author
-
Andrew V. Mossine, Allen F. Brooks, Bradford D. Henderson, Brian G. Hockley, Kirk A. Frey, and Peter J. H. Scott
- Subjects
Tau imaging ,Brain PET ,[18F]T807 ,Flortaucipir F18 ,Fluorine-18 radiochemistry ,Automated radiosynthesis ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
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.
- Published
- 2017
- Full Text
- View/download PDF
23. Parametric methods for [ 18 F]flortaucipir PET.
- Author
-
Golla, Sandeep SV, Wolters, Emma E, Timmers, Tessa, Ossenkoppele, Rik, van der Weijden, Chris WJ, Scheltens, Philip, Schwarte, Lothar, Mintun, Mark A, Devous Sr, Michael D, Schuit, Robert C, Windhorst, Albert D, Lammertsma, Adriaan A, Yaqub, Maqsood, van Berckel, Bart NM, and Boellaard, Ronald
- 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) (r 2 = 0.92; slope = 0.99) derived VT correlated well with NLR-derived VT. RPM (r 2 = 0.95; slope = 0.98) derived BPND correlated well with NLR-derived DVR. Although SUVr80–100 min correlated well with NLR-derived DVR (r 2 = 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. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
24. Amyloid and tau imaging biomarkers explain cognitive decline from late middle-age.
- Author
-
Betthauser, Tobey J, Koscik, Rebecca L, Jonaitis, Erin M, Allison, Samantha L, Cody, Karly A, Erickson, Claire M, Rowley, Howard A, Stone, Charles K, Mueller, Kimberly D, Clark, Lindsay R, Carlsson, Cynthia M, Chin, Nathaniel A, Asthana, Sanjay, Christian, Bradley T, and Johnson, Sterling C
- Subjects
- *
AMYLOID , *NEUROFIBRILLARY tangles , *ALZHEIMER'S disease , *ENTORHINAL cortex , *COGNITIVE testing , *PET therapy , *COGNITIVE Abilities Test - Abstract
This study investigated differences in retrospective cognitive trajectories between amyloid and tau PET biomarker stratified groups in initially cognitively unimpaired participants sampled from the Wisconsin Registry for Alzheimer's Prevention. One hundred and sixty-seven initially unimpaired individuals (baseline age 59 ± 6 years; 115 females) were stratified by elevated amyloid-β and tau status based on 11C-Pittsburgh compound B (PiB) and 18F-MK-6240 PET imaging. Mixed effects models were used to determine if longitudinal cognitive trajectories based on a composite of cognitive tests including memory and executive function differed between biomarker groups. Secondary analyses investigated group differences for a variety of cross-sectional health and cognitive tests, and associations between 18F-MK-6240, 11C-PiB, and age. A significant group × age interaction was observed with post hoc comparisons indicating that the group with both elevated amyloid and tau pathophysiology were declining approximately three times faster in retrospective cognition compared to those with just one or no elevated biomarkers. This result was robust against various thresholds and medial temporal lobe regions defining elevated tau. Participants were relatively healthy and mostly did not differ between biomarker groups in health factors at the beginning or end of study, or most cognitive measures at study entry. Analyses investigating association between age, MK-6240 and PiB indicated weak associations between age and 18F-MK-6240 in tangle-associated regions, which were negligible after adjusting for 11C-PiB. Strong associations, particularly in entorhinal cortex, hippocampus and amygdala, were observed between 18F-MK-6240 and global 11C-PiB in regions associated with Braak neurofibrillary tangle stages I–VI. These results suggest that the combination of pathological amyloid and tau is detrimental to cognitive decline in preclinical Alzheimer's disease during late middle-age. Within the Alzheimer's disease continuum, middle-age health factors likely do not greatly influence preclinical cognitive decline. Future studies in a larger preclinical sample are needed to determine if and to what extent individual contributions of amyloid and tau affect cognitive decline. 18F-MK-6240 shows promise as a sensitive biomarker for detecting neurofibrillary tangles in preclinical Alzheimer's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
25. Az Alzheimer-kór molekuláris képi megjelenítése.
- Author
-
Albert, Annamária and Borbély, Katalin
- Abstract
Copyright of Hungarian Medical Journal / Orvosi Hetilap is the property of Akademiai Kiado and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2019
- Full Text
- View/download PDF
26. Imaging of tau deposits in adults with Niemann-Pick type C disease: a case-control study.
- Author
-
Bozinoski, S., Villemagne, Victor L., Rowe, C. C., Doré, V., Masters, C. L., Walterfang, Mark, and Velakoulis, D.
- Subjects
- *
NIEMANN-Pick diseases , *AMYLOID , *MILD cognitive impairment , *SEVERITY of illness index , *LYSOSOMES - Abstract
Purpose: Niemann-Pick type C (NPC) is a cholesterol storage disease characterized by disruption in the endosomal–lysosomal transport system that leads to the accumulation of cholesterol and glycolipids in lysosomes. Developmental cognitive delay and progressive motor and cognitive impairment are characteristic of the disease. Tau accumulation has been reported in some NPC patients. We investigated the presence of tau and Aβ-amyloid deposits in a group of NPC patients and for comparison in age-matched healthy controls (HC). Methods: Eight NPC patients and seven HC were included in the study. Participants underwent tau imaging with 18F-AV1451 and amyloid imaging with 11C-PiB. Both 18F-AV1451 and 11C-PiB standardized uptake value ratios were generated using the cerebellar cortex as the reference region. Associations between imaging results, and clinical and neurocognitive parameters were assessed through nonparametric analyses. Results: All participants were Aβ-negative. Four NPC patients presented with high tau burden in the brain. A 21-year-old female patient and a 40-year-old male patient showed high neocortical tau burden in a pattern different from that observed in patients with Alzheimer's disease, while the same 40-year-old male patient, a 40-year-old female patient and a 50-year-old female patient showed high regional tau burden in the mesial temporal cortex. Spearman's correlation analysis showed an association between tau burden in the mesial temporal lobe and age (p = 0.022), and age at symptom onset (p = 0.009), and between frontotemporal tau and duration of symptoms (p = 0.027). There were no correlations between global and regional tau and cognitive parameters. Conclusion: Four of eight NPC patients showed tau deposition in the brain. The results of our exploratory study suggest that while tau deposits do not affect cognitive performance, tau deposits are associated with measures of disease onset and progression. Further studies in a larger cohort of NPC patients are needed to confirm these initial findings. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. In vivo binding of a tau imaging probe, [11 C]PBB3, in patients with progressive supranuclear palsy.
- Author
-
Endo, Hironobu, Shimada, Hitoshi, Sahara, Naruhiko, Ono, Maiko, Koga, Shunsuke, Kitamura, Soichiro, Niwa, Fumitoshi, Hirano, Shigeki, Kimura, Yasuyuki, Ichise, Masanori, Shinotoh, Hitoshi, Zhang, Ming Rong, Kuwabara, Satoshi, Dickson, Dennis W., Toda, Tatsushi, Suhara, Tetsuya, and Higuchi, Makoto
- Subjects
- *
AMINES , *BRAIN , *COMPARATIVE studies , *RESEARCH methodology , *MEDICAL cooperation , *NERVE tissue proteins , *PROGRESSIVE supranuclear palsy , *RADIOGRAPHY , *RADIOISOTOPES , *RESEARCH , *THIAZOLES , *POSITRON emission tomography , *EVALUATION research , *CASE-control method ,BRAIN metabolism - Abstract
Background: [11 C]pyridinyl-butadienyl-benzothiazole 3 is a PET imaging agent designed for capturing pathological tau aggregates in diverse neurodegenerative disorders, and would be of clinical utility for neuropathological investigations of PSP.Objectives: To explore the usefulness of [11 C]pyridinyl-butadienyl-benzothiazole 3/PET in assessing characteristic distributions of tau pathologies and their association with clinical symptoms in the brains of living PSP patients.Methods: We assessed 13 PSP patients and 13 age-matched healthy control subjects. Individuals negative for amyloid β PET with [11 C]Pittsburgh compound B underwent clinical scoring, MR scans, and [11 C]pyridinyl-butadienyl-benzothiazole 3/PET.Results: There were significant differences in binding potential for [11 C]pyridinyl-butadienyl-benzothiazole 3 between PSP patients and healthy control subjects (P = 0.02). PSP patients exhibited greater radioligand retention than healthy control subjects in multiple brain regions, including frontoparietal white matter, parietal gray matter, globus pallidus, STN, red nucleus, and cerebellar dentate nucleus. [11 C]pyridinyl-butadienyl-benzothiazole 3 deposition in frontoparietal white matter, but not gray matter, was correlated with general severity of parkinsonian and PSP symptoms, whereas both gray matter and white matter [11 C]pyridinyl-butadienyl-benzothiazole 3 accumulations in the frontoparietal cortices were associated with nonverbal cognitive impairments. Autoradiographic and fluorescence labeling with pyridinyl-butadienyl-benzothiazole 3 was observed in gray matter and white matter of PSP motor cortex tissues.Conclusions: Our findings support the in vivo detectability of tau fibrils characteristic of PSP by [11 C]pyridinyl-butadienyl-benzothiazole 3/PET, and imply distinct and synergistic contributions of gray matter and white matte tau pathologies to clinical symptoms. [11 C]pyridinyl-butadienyl-benzothiazole 3/PET potentially provides a neuroimaging-based index for the evolution of PSP tau pathologies promoting the deterioration of motor and cognitive functions. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
28. PET imaging of tau protein targets: a methodology perspective.
- Author
-
Lois, Cristina, Gonzalez, Ivan, Johnson, Keith A., and Price, Julie C.
- 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. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
29. Tau-mediated Neurodegeneration and Potential Implications in Diagnosis and Treatment of Alzheimer's Disease
- Author
-
Xi-Lin Wu, Juan Piña-Crespo, Yun-Wu Zhang, Xiao-Chun Chen, and Hua-Xi Xu
- Subjects
Alzheimer's Disease ,Biomarker ,Immunotherapy ,Tau Imaging ,Tau Protein ,Transcellular Propagation ,Medicine - Abstract
Objective: To review recent research advances on tau, a major player in Alzheimer's disease (AD) pathogenesis, a biomarker for AD onset, and potential target for AD therapy. Data Sources: This review was based on a comprehensive search using online literature databases, including PubMed, Web of Science, and Google Scholar. Study Selection: Literature search was based on the following keywords: Alzheimer's disease, tau protein, biomarker, cerebrospinal fluid (CSF), therapeutics, plasma, imaging, propagation, spreading, seeding, prion, conformational templating, and posttranslational modification. Relevant articles were carefully reviewed, with no exclusions applied to study design and publication type. Results: Amyloid plaques enriched with extracellular amyloid beta (Aβ) and intracellular neurofibrillary tangles comprised of hyperphosphorylated tau proteins are the two main pathological hallmarks of AD. Although the Aβ hypothesis has dominated AD research for many years, clinical Aβ-targeting strategies have consistently failed to effectively treat AD or prevent AD onset. The research focus in AD has recently shifted to the role of tau in AD. In addition to phosphorylation, tau is acetylated and proteolytically cleaved, which also contribute to its physiological and pathological functions. Emerging evidence characterizing pathological tau propagation and spreading provides new avenues for research into the molecular and cellular mechanisms underlying AD pathogenesis. Techniques to detect tau at minute levels in CSF and blood have been developed, and improved tracers have facilitated tau imaging in the brain. These advances have potential to accurately determine tau levels at early diagnostic stages in AD. Given that tau is a potential therapeutic target, anti-tau immunotherapy may potentially be a viable treatment strategy in AD intervention. Conclusion: Detecting changes in tau and targeting tau pathology represent a promising lead in the diagnosis and treatment of AD.
- Published
- 2017
- Full Text
- View/download PDF
30. Untangling tau imaging
- Author
-
Victor L. Villemagne, Nobuyuki Okamura, and Christopher C. Rowe
- Subjects
PET ,Tau imaging ,Alzheimer's disease ,Positron emission tomography ,Tauopathies ,Dementia ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract In vivo imaging of tau deposits is providing a better understanding of the temporal and spatial tau deposition in the brain, allowing a more comprehensive insight into the causes, diagnoses, and potentially treatment of tauopathies such as Alzheimer's disease, progressive supranuclear palsy, corticobasal syndrome, chronic traumatic encephalopathy, and some variants of frontotemporal lobar degeneration. The assessment of tau deposition in the brain over time will allow a deeper understanding of the relationship between tau and other variables such as cognition, genotype, and neurodegeneration, as well as assessing the role tau plays in ageing. Preliminary human studies suggest that tau imaging could also be used as a diagnostic, prognostic, and theranostic biomarker, as well as a surrogate marker for target engagement, patient recruitment, and efficacy monitoring for disease‐specific therapeutic trials.
- Published
- 2016
- Full Text
- View/download PDF
31. Amyloid and tau imaging biomarkers explain cognitive decline from late middle-age.
- Author
-
Betthauser, Tobey J, Koscik, Rebecca L, Jonaitis, Erin M, Allison, Samantha L, Cody, Karly A, Erickson, Claire M, Rowley, Howard A, Stone, Charles K, Mueller, Kimberly D, Clark, Lindsay R, Carlsson, Cynthia M, Chin, Nathaniel A, Asthana, Sanjay, Christian, Bradley T, and Johnson, Sterling C
- Abstract
This study investigated differences in retrospective cognitive trajectories between amyloid and tau PET biomarker stratified groups in initially cognitively unimpaired participants sampled from the Wisconsin Registry for Alzheimer's Prevention. One hundred and sixty-seven initially unimpaired individuals (baseline age 59 ± 6 years; 115 females) were stratified by elevated amyloid-β and tau status based on 11C-Pittsburgh compound B (PiB) and 18F-MK-6240 PET imaging. Mixed effects models were used to determine if longitudinal cognitive trajectories based on a composite of cognitive tests including memory and executive function differed between biomarker groups. Secondary analyses investigated group differences for a variety of cross-sectional health and cognitive tests, and associations between 18F-MK-6240, 11C-PiB, and age. A significant group × age interaction was observed with post hoc comparisons indicating that the group with both elevated amyloid and tau pathophysiology were declining approximately three times faster in retrospective cognition compared to those with just one or no elevated biomarkers. This result was robust against various thresholds and medial temporal lobe regions defining elevated tau. Participants were relatively healthy and mostly did not differ between biomarker groups in health factors at the beginning or end of study, or most cognitive measures at study entry. Analyses investigating association between age, MK-6240 and PiB indicated weak associations between age and 18F-MK-6240 in tangle-associated regions, which were negligible after adjusting for 11C-PiB. Strong associations, particularly in entorhinal cortex, hippocampus and amygdala, were observed between 18F-MK-6240 and global 11C-PiB in regions associated with Braak neurofibrillary tangle stages I-VI. These results suggest that the combination of pathological amyloid and tau is detrimental to cognitive decline in preclinical Alzheimer's disease during late middle-age. Within the Alzheimer's disease continuum, middle-age health factors likely do not greatly influence preclinical cognitive decline. Future studies in a larger preclinical sample are needed to determine if and to what extent individual contributions of amyloid and tau affect cognitive decline. 18F-MK-6240 shows promise as a sensitive biomarker for detecting neurofibrillary tangles in preclinical Alzheimer's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
32. Aerobic glycolysis and tau deposition in preclinical Alzheimer's disease.
- Author
-
Vlassenko, Andrei G., Gordon, Brian A., Goyal, Manu S., Su, Yi, Blazey, Tyler M., Durbin, Tony J., Couture, Lars E., Christensen, Jon J., Jafri, Hussain, Morris, John C., Raichle, Marcus E., and Benzinger, Tammie L.-S.
- Subjects
- *
GLYCOLYSIS , *ALZHEIMER'S disease , *TAU proteins , *OXIDATIVE phosphorylation ,BRAIN metabolism - Abstract
Research of the human brain metabolism in vivo has largely focused on total glucose use (via fluorodeoxyglucose positron emission tomography) and, until recently, did not examine the use of glucose outside oxidative phosphorylation, which is known as aerobic glycolysis (AG). AG supports important functions including biosynthesis and neuroprotection but decreases dramatically with aging. This multitracer positron emission tomography study evaluated the relationship between AG, total glucose use (CMRGlc), oxygen metabolism (CMRO 2 ), tau, and amyloid deposition in 42 individuals, including those at preclinical and symptomatic stages of Alzheimer's disease. Our findings demonstrate that in individuals with amyloid burden, lower AG is associated with higher tau deposition. No such correlation was observed for CMRGlc or CMRO 2 . We suggest that aging-related loss of AG leading to decreased synaptic plasticity and neuroprotection may accelerate tauopathy in individuals with amyloid burden. Longitudinal AG and Alzheimer's disease pathology studies are needed to verify causality. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. Longitudinal uncoupling of cerebral perfusion, glucose metabolism, and tau deposition in Alzheimer's disease.
- Author
-
Leuzy, Antoine, Rodriguez‐Vieitez, Elena, Saint‐Aubert, Laure, Chiotis, Konstantinos, Almkvist, Ove, Savitcheva, Irina, Jonasson, My, Lubberink, Mark, Wall, Anders, Antoni, Gunnar, and Nordberg, Agneta
- Abstract
Introduction: Cross‐sectional findings using the tau tracer [18F]THK5317 (THK5317) have shown that [18F]fluorodeoxyglucose (FDG) positron emission tomography (PET) data can be approximated using perfusion measures (early‐frame standardized uptake value ratio; ratio of tracer delivery in target to reference regions). In this way, a single PET study can provide both functional and molecular information. Methods: We included 16 patients with Alzheimer's disease who completed follow‐up THK5317 and FDG studies 17 months after baseline investigations. Linear mixed‐effects models and annual percentage change maps were used to examine longitudinal change. Results: Limited spatial overlap was observed between areas showing declines in THK5317 perfusion measures and FDG. Minimal overlap was seen between areas showing functional change and those showing increased retention of THK5317. Discussion: Our findings suggest a spatiotemporal offset between functional changes and tau pathology and a partial uncoupling between perfusion and metabolism, possibly as a function of Alzheimer's disease severity. Highlights: Overlap between changes in perfusion, metabolism, and tau was assessed.Declines in perfusion and metabolism showed only partial spatial overlap.Minimal spatial overlap was found between declines in perfusion and increased tau.Perfusion measures related differently to FDG PET findings in terms of overlap. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
34. Comparative binding properties of the tau PET tracers THK5117, THK5351, PBB3, and T807 in postmortem Alzheimer brains.
- Author
-
Lemoine, Laetitia, Gillberg, Per-Göran, Svedberg, Marie, Stepanov, Vladimir, Zhisheng Jia, Jinghai Huang, Sangram Nag, He Tian, Ghetti, Bernardino, Nobuyuki Okamura, Makoto Higuchi, Halldin, Christer, and Nordberg, Agneta
- Subjects
- *
ALZHEIMER'S disease , *AMYLOID beta-protein - Abstract
Background: The aim of this study was to compare the binding properties of several tau positron emission tomography tracers-THK5117, THK5351, T807 (also known as AV1451; flortaucipir), and PBB3-head to head in the same human brain tissue. Methods: Binding assays were performed to compare the regional distribution of 3H-THK5117 and 3H-THK5351 in postmortem tissue from three Alzheimer's disease (AD) cases and three control subjects in frontal and temporal cortices as well as in the hippocampus. Competition binding assays between THK5351, THK5117, PBB3, and T807, as well as off-target binding of THK5117 and T807 toward monoamine oxidase B (MAO-B), were performed using binding assays in brain homogenates and autoradiography of three AD cases. Results: Regional binding of 3H-THK5117 and 3H-THK5351 was similar, except in the temporal cortex, which showed higher 3H-THK5117 binding. Saturation studies demonstrated two binding sites for 3H-THK5351 (Kd1 = 5.6 nM, Bmax = 76 pmol/g; Kd2 = 1 nM, Bmax = 40 pmol/g). Competition studies in the hippocampus between 3HTHK5351 and unlabeled THK5351, THK5117, and T807 revealed super-high-affinity sites for all three tracers (THK5351 Ki = 0.1 pM; THK5117 Ki = 0.3 pM; T807 Ki = 0.2 pM) and an additional high-affinity site (THK5351 Ki = 16 nM; THK5117 Ki = 20 nM; T807 Ki = 78nM). 18F-T807, 11C-THK5351, and 11C-PBB3 autoradiography of large frozen sections from three AD brains showed similar regional binding for the three tracers, with lower binding intensity for 11C-PBB3. Unlabeled THK5351 and T807 displaced 11C-THK5351 to a similar extent and a lower extent, respectively, compared with 11C-PBB3. Competition with the MAO-B inhibitor 3H-L-deprenyl was observed for THK5117 and T807 in the hippocampus (THK5117 Ki = 286 nM; T807 Ki = 227 nM) and the putamen (THK5117 Ki = 148 nM; T807 Ki = 135 nM). 3H-THK5351 binding was displaced using autoradiography competition with unlabeled THK5351 and T807 in cortical areas by 70-80% and 60-77%, respectively, in the basal ganglia, whereas unlabeled deprenyl displaced 3HTHK5351 binding by 40% in the frontal cortex and 50% in the basal ganglia.Conclusions: THK5351, THK5117, and T807 seem to target similar binding sites, but with different affinities, whereas PBB3 seems to target its own binding site. Both THK5117 and T807 demonstrated off-target binding in the hippocampus and putamen with a ten times lower binding affinity to the MAO-B inhibitor deprenyl compared with 3H-THK5351. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
35. Quantification of Tau Load Using [18F]AV1451 PET.
- Author
-
Golla, Sandeep, Timmers, Tessa, Ossenkoppele, Rik, Groot, Colin, Verfaillie, Sander, Scheltens, Philip, Flier, Wiesje, Schwarte, Lothar, Mintun, Mark, Devous, Michael, Schuit, Robert, Windhorst, Albert, Lammertsma, Adriaan, Boellaard, Ronald, Berckel, Bart, Yaqub, Maqsood, Golla, Sandeep S V, van der Flier, Wiesje M, Mintun, Mark A, and Schuit, Robert C
- Subjects
- *
ALZHEIMER'S disease diagnosis , *LIGAND analysis , *BIOMARKERS , *TRACERS (Biology) , *POSITRON emission tomography , *EMISSION tomography equipment , *ALZHEIMER'S disease , *DYNAMICS , *NERVE tissue proteins , *PYRIDINE , *CASE-control method - Abstract
Purpose: The tau tracer [18F]AV1451, also known as flortaucipir, is a promising ligand for imaging tau accumulation in Alzheimer's disease (AD). Most of the previous studies have quantified tau load using standardized uptake value ratios (SUVr) derived from a static [18F]AV1451 scan. SUVr may, however, be flow dependent and, especially for longitudinal studies, should be validated against a fully quantitative approach. The objective of this study was to identify the optimal tracer kinetic model for measuring tau load using [18F]AV1451.Procedures: Following intravenous injection of 225 ± 16 MBq [18F]AV1451, 130 min dynamic PET scans were performed in five biomarker confirmed AD patients and five controls. Arterial blood sampling was performed to obtain a metabolite-corrected plasma input function. Next, regional time-activity curves were generated using PVElab software. These curves were analysed using several pharmacokinetic models.Results: The reversible single tissue compartment model (1T2k_VB) was the preferred model for all but one control. For AD patients, however, model preference shifted towards a reversible two tissue compartmental model (2T4k_VB). The simplified reference tissue model (SRTM) derived binding potential (BPND) showed good correlation (AD: r 2 = 0.87, slope = 1.06; controls: r 2 = 0.87, slope = 0.86) with indirect plasma input binding (distribution volume ratio-1). Standardized uptake value ratios (80-100 min) correlated well with DVR (r 2 = 0.93, slope = 1.07) and SRTM-derived BPND (r 2 = 0.84, slope = 0.95). In addition, regional differences in tracer binding between subject groups in different tau-specific regions were observed.Conclusions: Model preference of [18F]AV1451 appears to depend on subject status and, in particular, VT. The relationship between model preference and VT suggests that (higher) tau load may be reflected by a second tissue compartment. Nevertheless, consistent results can be obtained using a 2T4k_VB model. In addition, SRTM can be used to derive BPND. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
36. An updated radiosynthesis of [F]AV1451 for tau PET imaging.
- Author
-
Mossine, Andrew, Brooks, Allen, Henderson, Bradford, Hockley, Brian, Frey, Kirk, and Scott, Peter
- Subjects
ALZHEIMER'S disease diagnosis ,NEUROFIBRILLARY tangles ,HIGH performance liquid chromatography ,DIAGNOSTIC imaging ,FRONTOTEMPORAL dementia ,DIAGNOSIS - Abstract
Background: [F]AV1451 is a commonly used radiotracer for imaging tau deposits in Alzheimer's disease (AD) and related non-AD tauopathies. Existing radiosyntheses of [F]AV1451 require complex purifications to provide doses suitable for use in clinical imaging studies. To address this issue, we have modified the synthesis of [F]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 [F]AV1451 synthesis using a TRACERLab FX 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 [F]AV1451 has been developed that is fully automated and prepares radiotracer doses suitable for use in clinical tau PET studies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
37. The Alzheimer's Disease Neuroimaging Initiative 3: Continued innovation for clinical trial improvement.
- Author
-
Weiner, Michael W., Veitch, Dallas P., Aisen, Paul S., Beckett, Laurel A., Cairns, Nigel J., Green, Robert C., Harvey, Danielle, Jr.Jack, Clifford R., Jagust, William, Morris, John C., Petersen, Ronald C., Salazar, Jennifer, Saykin, Andrew J., Shaw, Leslie M., Toga, Arthur W., and Trojanowski, John Q.
- Abstract
Introduction The overall goal of the Alzheimer's Disease Neuroimaging Initiative (ADNI) is to validate biomarkers for Alzheimer's disease (AD) clinical trials. ADNI-3, which began on August 1, 2016, is a 5-year renewal of the current ADNI-2 study. Methods ADNI-3 will follow current and additional subjects with normal cognition, mild cognitive impairment, and AD using innovative technologies such as tau imaging, magnetic resonance imaging sequences for connectivity analyses, and a highly automated immunoassay platform and mass spectroscopy approach for cerebrospinal fluid biomarker analysis. A Systems Biology/pathway approach will be used to identify genetic factors for subject selection/enrichment. Amyloid positron emission tomography scanning will be standardized using the Centiloid method. The Brain Health Registry will help recruit subjects and monitor subject cognition. Results Multimodal analyses will provide insight into AD pathophysiology and disease progression. Discussion ADNI-3 will aim to inform AD treatment trials and facilitate development of AD disease-modifying treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
38. Tau-PET Binding Distinguishes Patients With Early-stage Posterior Cortical Atrophy From Amnestic Alzheimer Disease Dementia.
- Author
-
Day, Gregory S., Gordon, Brian A., Jackson, Kelley, Christensen, Jon J., Ponisio, Maria Rosana, Yi Su, Ances, Beau M., Benzinger, Tammie L. S., Morris, John C., Rosana Ponisio, Maria, and Su, Yi
- Abstract
Background: Flortaucipir (tau) positron emission tomography (PET) binding distinguishes individuals with clinically well-established posterior cortical atrophy (PCA) due to Alzheimer disease (AD) from cognitively normal (CN) controls. However, it is not known whether tau-PET binding patterns differentiate individuals with PCA from those with amnestic AD, particularly early in the symptomatic stages of disease.Methods: Flortaucipir and florbetapir (β-amyloid) PET imaging were performed in individuals with early-stage PCA (N=5), amnestic AD dementia (N=22), and CN controls (N=47). Average tau and β-amyloid deposition were quantified using standard uptake value ratios and compared at a voxelwise level, controlling for age.Results: PCA patients [median age-at-onset, 59 (51 to 61) years] were younger at symptom onset than similarly staged individuals with amnestic AD [75 (60 to 85) years] or CN controls [73 (61 to 90) years; P=0.002]. Flortaucipir uptake was higher in individuals with early-stage symptomatic PCA versus those with early-stage amnestic AD or CN controls, and greatest in posterior regions. Regional elevations in florbetapir were observed in areas of greatest tau deposition in PCA patients.Conclusions and Relevance: Flortaucipir uptake distinguished individuals with PCA and amnestic AD dementia early in the symptomatic course. The posterior brain regions appear to be uniquely vulnerable to tau deposition in PCA, aligning with clinical deficits that define this disease subtype. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
39. Development of [11C]/[3H]THK-5351 – A potential novel carbon-11 tau imaging PET radioligand.
- Author
-
Stepanov, Vladimir, Svedberg, Marie, Jia, Zhisheng, Krasikova, Raisa, Lemoine, Laetitia, Okamura, Nobujuki, Furumoto, Shozo, Mitsios, Nicholas, Mulder, Jan, Långström, Bengt, Nordberg, Agneta, and Halldin, Christer
- Subjects
- *
DEMENTIA , *POSITRON emission tomography , *AUTORADIOGRAPHY , *CARBAMATES , *TRIFLUOROACETIC acid - Abstract
Introduction Due to the rise in the number of patients with dementia the imperative for finding new diagnostic and treatment options becomes ever more pressing. While significant progress has been made in PET imaging of Aβ aggregates both in vitro and in vivo , options for imaging tau protein aggregates selectively are still limited. Based on the work previously published by researchers from the Tohoku University, Japan, that resulted in the development of [ 18 F]THK-5351, we have undertaken an effort to develop a carbon-11 version of the identical structure - [ 11 C]THK-5351. In parallel, THK-5351 was also labeled with tritium ([ 3 H]THK-5351) for use in in vitro autoradiography (ARG). Methods The carbon-11 labeling was performed starting with di-protected enantiomeric pure precursor - tert -butyl 5-(6-((2S)-3-fluoro-2-(tetrahydro-2 H -pyran-2-yloxy)propoxy)quinolin-2-yl)pyridin-2-yl carbamate, which was reacted with [ 11 C]MeI, using DMF as the solvent and NaH as base, followed by deprotection with trifluoroacetic acid/water mixture, resulting in enantiomerically pure carbon-11 radioligand, [ 11 C]THK-5351 - (S)-1-fluoro-3-(2-(6-([ 11 C]methylamino)pyridin-3-yl)quinolin-6-yloxy)propan-2-ol. Tritium labeling and purification of [ 3 H]THK-5351 were undertaken using similar approach, resulting in [ 3 H]THK-5351 with RCP >99.8% and specific radioactivity of 1.3 GBq/μmol. Results [ 11 C]THK-5351 was produced in good yield (1900 ± 355 MBq), specific radioactivity (SRA) (361 ± 119 GBq/μmol at EOS + 20 min) and radiochemical purity (RCP) (>99.8%), with enantiomeric purity of 98.7%. [ 3 H]THK-5351 was evaluated for ARG of tau binding in post-mortem human brain tissue using cortical sections from one AD patient and one control subject. [ 3 H]THK-5351 binding density was higher in the AD patient compared to the control subject, the binding was displaced by unlabeled THK-5351 confirming specific [ 3 H]THK-5351 binding. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
40. Relationship between amyloid and tau levels and its impact on tau spreading
- Author
-
Christopher C. Rowe, Shenpeng Li, Jurgen Fripp, Pierrick Bourgeat, Vincent Dore, Colin L. Masters, Natasha Krishnadas, Samantha C. Burnham, Victor L. Villemagne, and Kun Huang
- Subjects
0301 basic medicine ,Aβ-amyloid imaging ,Positron emission tomography ,Amyloid ,medicine.medical_specialty ,Epidemiology ,Hippocampus ,tau Proteins ,Amygdala ,Temporal lobe ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Neurodegeneration ,Tau imaging ,Amyloid beta-Peptides ,Neocortex ,Chemistry ,Health Policy ,General Medicine ,Entorhinal cortex ,medicine.disease ,Magnetic Resonance Imaging ,Psychiatry and Mental health ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Tauopathies ,Positron-Emission Tomography ,Original Article ,Neurology (clinical) ,Alzheimer's disease ,Geriatrics and Gerontology ,Alzheimer’s disease ,030217 neurology & neurosurgery - Abstract
Purpose Previous studies have shown that Aβ-amyloid (Aβ) likely promotes tau to spread beyond the medial temporal lobe. However, the Aβ levels necessary for tau to spread in the neocortex is still unclear. Methods Four hundred sixty-six participants underwent tau imaging with [18F]MK6420 and Aβ imaging with [18F]NAV4694. Aβ scans were quantified on the Centiloid (CL) scale with a cut-off of 25 CL for abnormal levels of Aβ (A+). Tau scans were quantified in three regions of interest (ROI) (mesial temporal (Me); temporoparietal neocortex (Te); and rest of neocortex (R)) and four mesial temporal region (entorhinal cortex, amygdala, hippocampus, and parahippocampus). Regional tau thresholds were established as the 95%ile of the cognitively unimpaired A- subjects. The prevalence of abnormal tau levels (T+) along the Centiloid continuum was determined. Results The plots of prevalence of T+ show earlier and greater increase along the Centiloid continuum in the medial temporal area compared to neocortex. Prevalence of T+ was low but associated with Aβ level between 10 and 40 CL reaching 23% in Me, 15% in Te, and 11% in R. Between 40 and 70 CL, the prevalence of T+ subjects per CL increased fourfold faster and at 70 CL was 64% in Me, 51% in Te, and 37% in R. In cognitively unimpaired, there were no T+ in R below 50 CL. The highest prevalence of T+ were found in the entorhinal cortex, reaching 40% at 40 CL and 80% at 60 CL. Conclusion Outside the entorhinal cortex, abnormal levels of cortical tau on PET are rarely found with Aβ below 40 CL. Above 40 CL prevalence of T+ accelerates in all areas. Moderate Aβ levels are required before abnormal neocortical tau becomes detectable.
- Published
- 2021
- Full Text
- View/download PDF
41. Tau Imaging in Alzheimer's Disease Diagnosis and Clinical Trials.
- Author
-
Brosch, Jared, Farlow, Martin, Risacher, Shannon, Apostolova, Liana, Brosch, Jared R, Farlow, Martin R, Risacher, Shannon L, and Apostolova, Liana G
- Subjects
BRAIN metabolism ,ALZHEIMER'S disease ,NERVE tissue proteins ,RADIOPHARMACEUTICALS ,POSITRON emission tomography - Abstract
In vivo imaging of the tau protein has the potential to aid in quantitative diagnosis of Alzheimer's disease, corroborate or dispute the amyloid hypothesis, and demonstrate biomarker engagement in clinical drug trials. A host of tau positron emission tomography agents have been designed, validated, and tested in humans. Several agents have characteristics approaching the ideal imaging tracer with some limitations, primarily regarding off-target binding. Dozens of clinical trials evaluating imaging techniques and several pharmaceutical trials have begun to integrate tau imaging into their protocols. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
42. Regional tau deposition measured by [18F]THK5317 positron emission tomography is associated to cognition via glucose metabolism in Alzheimer's disease.
- Author
-
Saint-Aubert, Laure, Almkvist, Ove, Chiotis, Konstantinos, Almeida, Rita, Wall, Anders, and Nordberg, Agneta
- Subjects
- *
POSITRON emission tomography , *GLUCOSE metabolism , *MILD cognitive impairment , *TAU leptons - Abstract
Background: The recent development of tau-specific positron emission tomography (PET) tracers has allowed in vivo quantification of regional tau deposition and offers the opportunity to monitor the progression of tau pathology along with cognitive impairment. In this study, we investigated the relationships of cerebral tau deposition ([18F]THK5317-PET) and metabolism ([18F]FDG-PET) with concomitant cognitive function in patients with probable Alzheimer's disease (AD). Methods: Nine patients diagnosed with AD dementia and 11 with prodromal AD (mild cognitive impairment, amyloid-positive on [11C]PiB-PET) were included in this study. All patients underwent PET scans using each tracer, as well as episodic memory and global cognition assessment. Linear models were used to investigate the association of regional [18F]THK5317 retention and [18F]FDG uptake with cognition. The possible mediating effect of local metabolism on the relationship between tau deposition and cognitive performance was investigated using mediation analyses. Results: Significant negative associations were found between [18F]THK5317 regional retention, mainly in temporal regions, and both episodic memory and global cognition. Significant positive associations were found between [18F]FDG regional uptake and cognition. The association of [18F]FDG with global cognition was regionally more extensive than that of [18F]THK5317, while the opposite was observed with episodic memory, suggesting that [18F]THK5317 retention might be more sensitive than [18F]FDG regional uptake to early cognitive impairment. Finally, [18F]FDG uptake had a mediating effect on the relationship between [18F]THK5317 retention in temporal regions and global cognition. Conclusions: These findings suggest a mediating role for local glucose metabolism in the observed association between in vivo tau deposition and concomitant cognitive impairment in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
43. The relationship between cerebrospinal fluid markers of Alzheimer pathology and positron emission tomography tau imaging.
- Author
-
Gordon, Brian A., Friedrichsen, Karl, Brier, Matthew, Blazey, Tyler, Yi Su, Christensen, Jon, Aldea, Patricia, McConathy, Jonathan, Holtzman, David M., Cairns, Nigel J., Morris, John C., Fagan, Anne M., Ances, Beau M., Benzinger, Tammie L. S., and Su, Yi
- Subjects
- *
CEREBROSPINAL fluid , *ALZHEIMER'S disease , *POSITRON emission tomography , *NEUROFIBRILLARY tangles , *BIOMARKERS , *LUMBAR puncture , *MILD cognitive impairment , *NERVE tissue proteins , *PEPTIDES , *PYRIDINE , *RESEARCH funding - Abstract
The two primary molecular pathologies in Alzheimer's disease are amyloid-β plaques and tau-immunoreactive neurofibrillary tangles. Investigations into these pathologies have been restricted to cerebrospinal fluid assays, and positron emission tomography tracers that can image amyloid-β plaques. Tau tracers have recently been introduced into the field, although the utility of the tracer and its relationship to other Alzheimer biomarkers are still unknown. Here we examined tau deposition in 41 cognitively normal and 11 cognitively impaired older adults using the radioactive tau ligand (18)F-AV-1451 (previously known as T807) who also underwent a lumbar puncture to assess cerebrospinal fluid levels of total tau (t-tau), phosphorylated tau181 (p-tau181) and amyloid-β42 Voxel-wise statistical analyses examined spatial patterns of tau deposition associated with cognitive impairment. We then related the amount of tau tracer uptake to levels of cerebrospinal fluid biomarkers. All analyses controlled for age and gender and, when appropriate, the time between imaging and lumbar puncture assessments. Symptomatic individuals (Clinical Dementia Rating > 0) demonstrated markedly increased levels of tau tracer uptake. This elevation was most prominent in the temporal lobe and temporoparietal junction, but extended more broadly into parietal and frontal cortices. In the entire cohort, there were significant relationships among all cerebrospinal fluid biomarkers and tracer uptake, notably for tau-related cerebrospinal fluid markers. After controlling for levels of amyloid-β42, the correlations with tau uptake were r = 0.490 (P < 0.001) for t-tau and r = 0.492 (P < 0.001) for p-tau181 Within the cognitively normal cohort, levels of amyloid-β42, but not t-tau or p-tau181, were associated with elevated tracer binding that was confined primarily to the medial temporal lobe and adjacent neocortical regions. AV-1451 tau binding in the medial temporal, parietal, and frontal cortices is correlated with tau-related cerebrospinal fluid measures. In preclinical Alzheimer's disease, there is focal tauopathy in the medial temporal lobes and adjacent cortices. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
44. Tau imaging in neurodegenerative diseases.
- Author
-
Dani, M., Brooks, D., and Edison, P.
- Subjects
- *
TAU proteins , *NEURODEGENERATION , *PATHOLOGICAL physiology , *POSITRON emission tomography , *BIOMARKERS , *TRACERS (Chemistry) - Abstract
Aggregated tau protein is a major neuropathological substrate central to the pathophysiology of neurodegenerative diseases such as Alzheimer's disease (AD), frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration and chronic traumatic encephalopathy. In AD, it has been shown that the density of hyperphosphorylated tau tangles correlates closely with neuronal dysfunction and cell death, unlike β-amyloid. Until now, diagnostic and pathologic information about tau deposition has only been available from invasive techniques such as brain biopsy or autopsy. The recent development of selective in-vivo tau PET imaging ligands including [F]THK523, [F]THK5117, [F]THK5105 and [F]THK5351, [F]AV1451(T807) and [C]PBB3 has provided information about the role of tau in the early phases of neurodegenerative diseases, and provided support for diagnosis, prognosis, and imaging biomarkers to track disease progression. Moreover, the spatial and longitudinal relationship of tau distribution compared with β - amyloid and other pathologies in these diseases can be mapped. In this review, we discuss the role of aggregated tau in tauopathies, the challenges posed in developing selective tau ligands as biomarkers, the state of development in tau tracers, and the new clinical information that has been uncovered, as well as the opportunities for improving diagnosis and designing clinical trials in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
45. Cryptic Sites in Tau Fibrils Explain the Preferential Binding of the AV-1451 PET Tracer toward Alzheimer's Tauopathy
- Author
-
Murugan, N. Arul, Nordberg, Agneta, Ågren, Hans, Murugan, N. Arul, Nordberg, Agneta, and Ågren, Hans
- Abstract
Tauopathies are a subclass of neurodegenerative diseases characterized by an accumulation of microtubule binding tau fibrils in brain regions. Diseases such as Alzheimer's (AD), chronic traumatic encephalopathy (CTE), Pick's disease (PiD), and corticobasal degeneration (CBD) belong to this subclass. Development of tracers which can visualize and discriminate between different tauopathies is of clinical importance in the diagnosis of various tauopathies. Currently, several tau tracers are available for in vivo imaging using a positron emission tomography (PET) technique. Among these tracers, PBB3 is reported to bind to various types of tau fibrils with comparable binding affinities. In contrast, tau tracer AV-1451 is reported to bind to specific types of tau fibrils (in particular to AD-associated and CTE) with higher binding affinity and only show nonspecific or weaker binding toward tau fibrils dominant with 3R isoforms (associated with PiD). The tau fibrils associated with different tauopathies can adopt different microstructures with different binding site microenvironments. By using detailed studies of the binding profiles of tau tracers for different types of tau fibrils, it may be possible to design tracers with high selectivity toward a specific tauopathy. The microstructures for the tau fibrils from patients with AD, PiD, and CTE have recently been demonstrated by cryogenic electron microscopy (cryo-EM) measurements allowing structure-based in silico simulations. In the present study, we have performed a multiscale computational study involving molecular docking, molecular dynamics, free energy calculations, and QM fragmentation calculations to understand the binding profiles of tau tracer AV-1451 and its potential use for diagnosis of AD, CTE, and PiD tauopathies. Our computational study reveals that different affinity binding sites exist for AV-1451 in the tau fibrils associated with different tauopathies. The binding affinity of this tracer toward differe
- Published
- 2021
- Full Text
- View/download PDF
46. Evaluation of [
- Author
-
Toshiki, Tezuka, Keisuke, Takahata, Morinobu, Seki, Hajime, Tabuchi, Yuki, Momota, Mika, Shiraiwa, Natsumi, Suzuki, Ayaka, Morimoto, Tadaki, Nakahara, Yu, Iwabuchi, Eisuke, Miura, Yasuharu, Yamamoto, Yasunori, Sano, Kei, Funaki, Bun, Yamagata, Ryo, Ueda, Takahito, Yoshizaki, Kyoko, Mashima, Mamoru, Shibata, Munenori, Oyama, Kensuke, Okada, Masahito, Kubota, Hajime, Okita, Masaki, Takao, Masahiro, Jinzaki, Jin, Nakahara, Masaru, Mimura, and Daisuke, Ito
- Subjects
Tau imaging ,AcademicSubjects/SCI01870 ,tauopathy ,Original Article ,AcademicSubjects/MED00310 ,progressive supranuclear palsy ,Alzheimer’s disease ,corticobasal degeneration - Abstract
Tau aggregates represent a key pathologic feature of Alzheimer’s disease and other neurodegenerative diseases. Recently, PET probes have been developed for in vivo detection of tau accumulation; however, they are limited because of off-target binding and a reduced ability to detect tau in non-Alzheimer’s disease tauopathies. The novel tau PET tracer, [18F]PI-2620, has a high binding affinity and specificity for aggregated tau; therefore, it was hypothesized to have desirable properties for the visualization of tau accumulation in Alzheimer’s disease and non-Alzheimer’s disease tauopathies. To assess the ability of [18F]PI-2620 to detect regional tau burden in non-Alzheimer’s disease tauopathies compared with Alzheimer’s disease, patients with progressive supranuclear palsy (n = 3), corticobasal syndrome (n = 2), corticobasal degeneration (n = 1) or Alzheimer’s disease (n = 8), and healthy controls (n = 7) were recruited. All participants underwent MRI, amyloid β assessment and [18F]PI-2620 PET (Image acquisition at 60–90 min post-injection). Cortical and subcortical tau accumulations were assessed by calculating standardized uptake value ratios using [18F]PI-2620 PET. For pathologic validation, tau pathology was assessed using tau immunohistochemistry and compared with [18F]PI-2620 retention in an autopsied case of corticobasal degeneration. In Alzheimer’s disease, focal retention of [18F]PI-2620 was evident in the temporal and parietal lobes, precuneus, and cingulate cortex. Standardized uptake value ratio analyses revealed that patients with non-Alzheimer’s disease tauopathies had elevated [18F]PI-2620 uptake only in the globus pallidus, as compared to patients with Alzheimer’s disease, but not healthy controls. A head-to-head comparison of [18F]PI-2620 and [18F]PM-PBB3, another tau PET probe for possibly visualizing the four-repeat tau pathogenesis in non-Alzheimer’s disease, revealed different retention patterns in one subject with progressive supranuclear palsy. Imaging-pathology correlation analysis of the autopsied patient with corticobasal degeneration revealed no significant correlation between [18F]PI-2620 retention in vivo. High [18F]PI-2620 uptake at 60–90 min post-injection in the globus pallidus may be a sign of neurodegeneration in four-repeat tauopathy, but not necessarily practical for diagnosis of non-Alzheimer’s disease tauopathies. Collectively, this tracer is a promising tool to detect Alzheimer’s disease-tau aggregation. However, late acquisition PET images of [18F]PI-2620 may have limited utility for reliable detection of four-repeat tauopathy because of lack of correlation between post-mortem tau pathology and different retention pattern than the non-Alzheimer’s disease-detectable tau radiotracer, [18F]PM-PBB3. A recent study reported that [18F]PI-2620 tracer kinetics curves in four-repeat tauopathies peak earlier (within 30 min) than Alzheimer’s disease; therefore, further studies are needed to determine appropriate PET acquisition times that depend on the respective interest regions and diseases., This study reports that the novel tau-tracer, [18F]PI-2620 (late time frame), is a promising tool for detecting Alzheimer’s disease-tau deposits. However, this may have limited utility for the detection of four-repeat tau-pathology due to the lack of correlation with post-mortem pathology and different distribution with the non-Alzheimer’s disease tau-tracer, [18F]PM-PBB3., Graphical Abstract Graphical Abstract
- Published
- 2021
47. Cryptic Sites in Tau Fibrils Explain the Preferential Binding of the AV-1451 PET Tracer toward Alzheimer's Tauopathy
- Author
-
N. Arul Murugan, Agneta Nordberg, and Hans Ågren
- Subjects
Gene isoform ,Physiology ,Cognitive Neuroscience ,In silico ,tau Proteins ,Molecular Dynamics Simulation ,Fibril ,Biochemistry ,Pick's disease ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Humans ,Corticobasal degeneration ,chronic traumatic encephalopathy ,Pick’s disease ,Binding site ,030304 developmental biology ,0303 health sciences ,Tau imaging ,Chemistry ,Neurosciences ,Brain ,Cell Biology ,General Medicine ,Alzheimer's disease ,medicine.disease ,multiscale modeling ,Molecular Docking Simulation ,Chronic traumatic encephalopathy ,Tauopathies ,neurofibrillary tangles ,Positron-Emission Tomography ,Biophysics ,Tauopathy ,Alzheimer’s disease ,QM fragmentation scheme ,030217 neurology & neurosurgery ,Neurovetenskaper ,Carbolines ,Research Article - Abstract
Tauopathies are a subclass of neurodegenerative diseases characterized by an accumulation of microtubule binding tau fibrils in brain regions. Diseases such as Alzheimer's (AD), chronic traumatic encephalopathy (CTE), Pick's disease (PiD), and corticobasal degeneration (CBD) belong to this subclass. Development of tracers which can visualize and discriminate between different tauopathies is of clinical importance in the diagnosis of various tauopathies. Currently, several tau tracers are available for in vivo imaging using a positron emission tomography (PET) technique. Among these tracers, PBB3 is reported to bind to various types of tau fibrils with comparable binding affinities. In contrast, tau tracer AV-1451 is reported to bind to specific types of tau fibrils (in particular to AD-associated and CTE) with higher binding affinity and only show nonspecific or weaker binding toward tau fibrils dominant with 3R isoforms (associated with PiD). The tau fibrils associated with different tauopathies can adopt different microstructures with different binding site microenvironments. By using detailed studies of the binding profiles of tau tracers for different types of tau fibrils, it may be possible to design tracers with high selectivity toward a specific tauopathy. The microstructures for the tau fibrils from patients with AD, PiD, and CTE have recently been demonstrated by cryogenic electron microscopy (cryo-EM) measurements allowing structure-based in silico simulations. In the present study, we have performed a multiscale computational study involving molecular docking, molecular dynamics, free energy calculations, and QM fragmentation calculations to understand the binding profiles of tau tracer AV-1451 and its potential use for diagnosis of AD, CTE, and PiD tauopathies. Our computational study reveals that different affinity binding sites exist for AV-1451 in the tau fibrils associated with different tauopathies. The binding affinity of this tracer toward different tau fibrils goes in this order: PiD > AD > CTE. The interaction energies for different tau fibril-tracer complexes using the QM fragmentation scheme also showed the same trend. However, by carrying out molecular dynamics simulations for the AD-derived tau fibrils in organic solvents, we found additional high affinity binding sites for AV-1451. The AV-1451 binding profile in these cryptic sites correctly explains the preferential binding of this tracer toward the AD fibrils when compared with the PiD fibrils. This study clearly demonstrates having a cryo-EM structure is still not sufficient for the structure-based tracer discovery for certain targets, as they may have "potential but hidden" high affinity binding sites, and we need additional strategies to identify them.
- Published
- 2021
48. A dual-time-window protocol to reduce acquisition time of dynamic tau PET imaging using [
- Author
-
Guilherme D, Kolinger, David, Vállez García, Talakad G, Lohith, Eric D, Hostetler, Cyrille, Sur, Arie, Struyk, Ronald, Boellaard, and Michel, Koole
- Subjects
Tau imaging ,Reference logan ,[18F]MK-6240 ,PET quantification ,Dual-time-window ,Original Research - Abstract
Background [18F]MK-6240 is a PET tracer with sub-nanomolar affinity for neurofibrillary tangles. Therefore, tau quantification is possible with [18F]MK-6240 PET/CT scans, and it can be used for assessment of Alzheimer’s disease. However, long acquisition scans are required to provide fully quantitative estimates of pharmacokinetic parameters. Therefore, on the present study, dual-time-window (DTW) acquisitions was simulated to reduce PET/CT acquisition time, while taking into consideration perfusion changes and possible scanning protocol non-compliance. To that end, time activity curves (TACs) representing a 120-min acquisition (TAC120) were simulated using a two-tissue compartment model with metabolite corrected arterial input function from 90-min dynamic [18F]MK-6240 PET scans of three healthy control subjects and five subjects with mild cognitive impairment or Alzheimer’s disease. Therefore, TACs corresponding to different levels of specific binding were generated and then various perfusion changes were simulated. Next, DTW acquisitions were simulated consisting of an acquisition starting at tracer injection, a break and a second acquisition starting at 90 min post-injection. Finally, non-compliance with the PET/CT scanning protocol were simulated to assess its impact on quantification. All TACs were quantified using reference Logan’s distribution volume ratio (DVR) and standardized uptake value ratio (SUVR90) using the cerebellar cortex as reference region. Results It was found that DVR from a DTW protocol with a 60-min break between two 30-min dynamic scans closely approximates the DVR from the uninterrupted TAC120, with a regional bias smaller than 2.5%. Moreover, SUVR90 estimates were more susceptible (regional bias ≤ 19%) to changes in perfusion compared to DVR from a DTW TAC (regional bias ≤ 10%). Similarly, SUVR90 was affected by late-time scanning protocol delays reaching an increase of 8% for a 20-min delay, while DVR was not affected (regional bias
- Published
- 2020
49. In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease.
- Author
-
Villemagne, Victor, Furumoto, Shozo, Fodero-Tavoletti, Michelle, Mulligan, Rachel, Hodges, John, Harada, Ryuichi, Yates, Paul, Piguet, Olivier, Pejoska, Svetlana, Doré, Vincent, Yanai, Kazuhiko, Masters, Colin, Kudo, Yukitsuka, Rowe, Christopher, and Okamura, Nobuyuki
- Subjects
- *
ALZHEIMER'S disease , *DISEASES in older people , *POSITRON emission tomography , *ELECTRONIC collimation , *POSITRON emission , *RADIONUCLIDE imaging , *MEDICAL imaging systems , *MEDICAL equipment - Abstract
Purpose: Diagnosis of tauopathies such as Alzheimer's disease (AD) still relies on post-mortem examination of the human brain. A non-invasive method of determining brain tau burden in vivo would allow a better understanding of the pathophysiology of tauopathies. The purpose of the study was to evaluate F-THK523 as a potential tau imaging tracer. Methods: Ten healthy elderly controls, three semantic dementia (SD) and ten AD patients underwent neuropsychological examination, MRI as well as F-THK523 and C-Pittsburgh compound B (PIB) positron emission tomography (PET) scans. Composite memory and non-memory scores, global and hippocampal brain volume, and partial volume-corrected tissue ratios for F-THK523 and C-PIB were estimated for all participants. Correlational analyses were performed between global and regional F-THK523, C-PIB, cognition and brain volumetrics. Results: F-THK523 presented with fast reversible kinetics. Significantly higher F-THK523 retention was observed in the temporal, parietal, orbitofrontal and hippocampi of AD patients when compared to healthy controls and SD patients. White matter retention was significantly higher than grey matter retention in all participants. The pattern of cortical F-THK523 retention did not correlate with Aβ distribution as assessed by C-PIB and followed the known distribution of tau in the AD brain, being higher in temporal and parietal areas than in the frontal region. Unlike C-PIB, hippocampal F-THK523 retention was correlated with several cognitive parameters and with hippocampal atrophy. Conclusion: F-THK523 does not bind to Aβ in vivo, while following the known distribution of paired helical filaments (PHF)-tau in the brain. Significantly higher cortical F-THK523 retention in AD patients as well as the association of hippocampal F-THK523 retention with cognitive parameters and hippocampal volume suggests F-THK523 selectively binds to tau in AD patients. Unfortunately, the very high F-THK523 retention in white matter precludes simple visual inspection of the images, preventing its use in research or clinical settings. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
50. Neuroinflammation and protein aggregation co-localize across the frontotemporal dementia spectrum
- Author
-
W. Richard Bevan-Jones, 1, Thomas E. Cope, 2, 3, P. Simon Jones, 2 Sanne S. Kaalund, 2 Luca Passamonti, 4 Kieren Allinson, 5 Oliver Green, 4 Young T. Hong, 6 Tim D. Fryer, 6 Robert Arnold, 1 Jonathan P. Coles, 7 Franklin I. Aigbirhio, 6 Andrew J. Larner, 8 Karalyn Patterson, 3 John T. O'Brien1, James B. Rowe2, Cope, Thomas [0000-0002-4751-1786], Jones, Simon [0000-0001-9695-0702], Kaalund, Sanne [0000-0002-8975-1825], Passamonti, Luca [0000-0002-7937-0615], Coles, Jonathan [0000-0003-4013-679X], Aigbirhio, Franklin [0000-0001-9453-5257], Patterson, Karalyn [0000-0003-1927-7424], Rowe, James [0000-0001-7216-8679], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Semantic dementia ,Disease ,Neuropathology ,Biology ,Primary progressive aphasia ,03 medical and health sciences ,Protein Aggregates ,0302 clinical medicine ,Semantic Dementia ,Microglia ,neuropathology ,primary progressive aphasia ,semantic dementia ,tau imaging ,mental disorders ,medicine ,Radioligand ,Humans ,Tau Imaging ,Carbon Radioisotopes ,Neuroinflammation ,Aged ,Inflammation ,Original Articles ,Middle Aged ,medicine.disease ,Isoquinolines ,Primary Progressive Aphasia ,DNA-Binding Proteins ,Editor's Choice ,030104 developmental biology ,medicine.anatomical_structure ,Tauopathies ,Case-Control Studies ,Frontotemporal Dementia ,Positron-Emission Tomography ,Female ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery ,Frontotemporal dementia ,Carbolines ,Protein Binding - Abstract
Bevan-Jones, Cope et al. report that neuroinflammation co-localizes with protein aggregation in all major types of frontotemporal dementia, both in vivo with positron emission tomography, and at post mortem. In vivo neuroinflammation patterns are disease-specific and can accurately classify patients into groups., The clinical syndromes of frontotemporal dementia are clinically and neuropathologically heterogeneous, but processes such as neuroinflammation may be common across the disease spectrum. We investigated how neuroinflammation relates to the localization of tau and TDP-43 pathology, and to the heterogeneity of clinical disease. We used PET in vivo with (i) 11C-PK-11195, a marker of activated microglia and a proxy index of neuroinflammation; and (ii) 18F-AV-1451, a radioligand with increased binding to pathologically affected regions in tauopathies and TDP-43-related disease, and which is used as a surrogate marker of non-amyloid-β protein aggregation. We assessed 31 patients with frontotemporal dementia (10 with behavioural variant, 11 with the semantic variant and 10 with the non-fluent variant), 28 of whom underwent both 18F-AV-1451 and 11C-PK-11195 PET, and matched control subjects (14 for 18F-AV-1451 and 15 for 11C-PK-11195). We used a univariate region of interest analysis, a paired correlation analysis of the regional relationship between binding distributions of the two ligands, a principal component analysis of the spatial distributions of binding, and a multivariate analysis of the distribution of binding that explicitly controls for individual differences in ligand affinity for TDP-43 and different tau isoforms. We found significant group-wise differences in 11C-PK-11195 binding between each patient group and controls in frontotemporal regions, in both a regions-of-interest analysis and in the comparison of principal spatial components of binding. 18F-AV-1451 binding was increased in semantic variant primary progressive aphasia compared to controls in the temporal regions, and both semantic variant primary progressive aphasia and behavioural variant frontotemporal dementia differed from controls in the expression of principal spatial components of binding, across temporal and frontotemporal cortex, respectively. There was a strong positive correlation between 11C-PK-11195 and 18F-AV-1451 uptake in all disease groups, across widespread cortical regions. We confirmed this association with post-mortem quantification in 12 brains, demonstrating strong associations between the regional densities of microglia and neuropathology in FTLD-TDP (A), FTLD-TDP (C), and FTLD-Pick's. This was driven by amoeboid (activated) microglia, with no change in the density of ramified (sessile) microglia. The multivariate distribution of 11C-PK-11195 binding related better to clinical heterogeneity than did 18F-AV-1451: distinct spatial modes of neuroinflammation were associated with different frontotemporal dementia syndromes and supported accurate classification of participants. These in vivo findings indicate a close association between neuroinflammation and protein aggregation in frontotemporal dementia. The inflammatory component may be important in shaping the clinical and neuropathological patterns of the diverse clinical syndromes of frontotemporal dementia.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.