101
|
Peretti DE, Vállez García D, Reesink FE, Doorduin J, de Jong BM, De Deyn PP, Dierckx RAJO, Boellaard R. Diagnostic performance of regional cerebral blood flow images derived from dynamic PIB scans in Alzheimer's disease. EJNMMI Res 2019; 9:59. [PMID: 31273465 PMCID: PMC6609664 DOI: 10.1186/s13550-019-0528-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/20/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND In clinical practice, visual assessment of glucose metabolism images is often used for the diagnosis of Alzheimer's disease (AD) through 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) scans. However, visual assessment of the characteristic AD hypometabolic pattern relies on the expertise of the reader. Therefore, user-independent pipelines are preferred to evaluate the images and to classify the subjects. Moreover, glucose consumption is highly correlated with cerebral perfusion. Regional cerebral blood flow (rCBF) images can be derived from dynamic 11C-labelled Pittsburgh Compound B PET scans, which are also used for the assessment of the deposition of amyloid-β plaques on the brain, a fundamental characteristic of AD. The aim of this study was to explore whether these rCBF PIB images could be used for diagnostic purposes through the PMOD Alzheimer's Discrimination Tool. RESULTS Both tracer relative cerebral flow (R1) and early PIB (ePIB) (20-130 s) uptake presented a good correlation when compared to FDG standardized uptake value ratio (SUVR), while ePIB (1-8 min) showed a worse correlation. All receiver operating characteristic curves exhibited a similar shape, with high area under the curve values, and no statistically significant differences were found between curves. However, R1 and ePIB (1-8 min) had the highest sensitivity, while FDG SUVR had the highest specificity. CONCLUSION rCBF images were suggested to be a good surrogate for FDG scans for diagnostic purposes considering an adjusted threshold value.
Collapse
Affiliation(s)
- Débora E. Peretti
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Fransje E. Reesink
- Department of Neurology, Alzheimer Centrum Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bauke M. de Jong
- Department of Neurology, Alzheimer Centrum Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Peter P. De Deyn
- Department of Neurology, Alzheimer Centrum Groningen, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, University of Antwerp, Universiteitsplein 1, 2610 Antwerpen, Belgium
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
102
|
Kim MJ, McGwier M, Jenko KJ, Snow J, Morse C, Zoghbi SS, Pike VW, Innis RB, Kreisl WC. Neuroinflammation in frontotemporal lobar degeneration revealed by 11 C-PBR28 PET. Ann Clin Transl Neurol 2019; 6:1327-1331. [PMID: 31353865 PMCID: PMC6649481 DOI: 10.1002/acn3.50802] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022] Open
Abstract
This study used 11 C-PBR28 positron emission tomography (PET) imaging to determine whether levels of 18-kDa translocator protein (TSPO), an inflammation-specific biomarker, are increased in frontotemporal lobar degeneration (FTLD) patients. 11 C-PBR28, 18 F-FDG, and 11 C-PIB brain PET scans, as well as magnetic resonance imaging (MRI), were conducted in four FTLD patients and 22 healthy controls. 11 C-PBR28 scans revealed that all FTLD patients showed increased TSPO binding versus controls. Significantly greater increases in TSPO were observed in the frontal, lateral temporal, parietal, and occipital cortices, topographically consistent with individual clinical phenotypes and with brain MRI and 18 F-FDG PET. Amyloid burden was not increased.
Collapse
Affiliation(s)
- Min-Jeong Kim
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Meghan McGwier
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Kimberly J. Jenko
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Joseph Snow
- Office of the Clinical DirectorNational Institute of Mental HealthBethesdaMaryland
| | - Cheryl Morse
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Sami S. Zoghbi
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Victor W. Pike
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - Robert B. Innis
- Molecular Imaging BranchNational Institute of Mental HealthBethesdaMaryland
| | - William C. Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew York
| |
Collapse
|
103
|
Peroxide-mediated oxidative coupling of primary alcohols and disulfides: Synthesis of 2-substituted benzothiazoles. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
104
|
Adams JN, Lockhart SN, Li L, Jagust WJ. Relationships Between Tau and Glucose Metabolism Reflect Alzheimer's Disease Pathology in Cognitively Normal Older Adults. Cereb Cortex 2019; 29:1997-2009. [PMID: 29912295 PMCID: PMC6458898 DOI: 10.1093/cercor/bhy078] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/22/2018] [Accepted: 03/20/2018] [Indexed: 12/26/2022] Open
Abstract
Tau is associated with hypometabolism in patients with Alzheimer's disease. In normal aging, the association between tau and glucose metabolism is not fully characterized. We used [18F] AV-1451, [18F] Fluorodeoxyglucose, and [11C] Pittsburgh Compound-B (PiB) PET to measure associations between tau and glucose metabolism in cognitively normal older adults (N = 49). Participants were divided into amyloid-negative (PiB-, n = 28) and amyloid-positive (PiB+, n = 21) groups to determine effects of amyloid-β. We assessed both local and across-brain regional tau-glucose metabolism associations separately in PiB-/PiB+ groups using correlation matrices and sparse canonical correlations. Relationships between tau and glucose metabolism differed by amyloid status, and were primarily spatially distinct. In PiB- subjects, tau was associated with broad regions of increased glucose metabolism. In PiB+ subjects, medial temporal lobe tau was associated with widespread hypometabolism, while tau outside of the medial temporal lobe was associated with decreased and increased glucose metabolism. We further found that regions with earlier tau spread were associated with stronger negative correlations with glucose metabolism. Our findings indicate that in normal aging, low levels of tau are associated with a phase of increased metabolism, while high levels of tau in the presence of amyloid-β are associated with hypometabolism at downstream sites.
Collapse
Affiliation(s)
- Jenna N Adams
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Samuel N Lockhart
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Lexin Li
- Department of Biostatistics, University of California, Berkeley, CA, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
105
|
Erickson CM, Schultz SA, Oh JM, Darst BF, Ma Y, Norton D, Betthauser T, Gallagher CL, Carlsson CM, Bendlin BB, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Engelman CD, Christian BT, Johnson SC, Dubal DB, Okonkwo OC. KLOTHO heterozygosity attenuates APOE4-related amyloid burden in preclinical AD. Neurology 2019; 92:e1878-e1889. [PMID: 30867273 PMCID: PMC6550504 DOI: 10.1212/wnl.0000000000007323] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To examine whether the KLOTHO gene variant KL-VS attenuates APOE4-associated β-amyloid (Aβ) accumulation in a late-middle-aged cohort enriched with Alzheimer disease (AD) risk factors. METHODS Three hundred nine late-middle-aged adults from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center were genotyped to determine KL-VS and APOE4 status and underwent CSF sampling (n = 238) and/or 11C-Pittsburgh compound B (PiB)-PET imaging (n = 183). Covariate-adjusted regression analyses were used to investigate whether APOE4 exerted expected effects on Aβ burden. Follow-up regression analyses stratified by KL-VS genotype (i.e., noncarrier vs heterozygous; there were no homozygous individuals) evaluated whether the influence of APOE4 on Aβ was different among KL-VS heterozygotes compared to noncarriers. RESULTS APOE4 carriers exhibited greater Aβ burden than APOE4-negative participants. This effect was stronger in CSF (t = -5.12, p < 0.001) compared with PiB-PET (t = 3.93, p < 0.001). In the stratified analyses, this APOE4 effect on Aβ load was recapitulated among KL-VS noncarriers (CSF: t = -5.09, p < 0.001; PiB-PET: t = 3.77, p < 0 .001). In contrast, among KL-VS heterozygotes, APOE4-positive individuals did not exhibit higher Aβ burden than APOE4-negative individuals (CSF: t = -1.03, p = 0.308; PiB-PET: t = 0.92, p = 0.363). These differential APOE4 effects remained after KL-VS heterozygotes and noncarriers were matched on age and sex. CONCLUSION In a cohort of at-risk late-middle-aged adults, KL-VS heterozygosity was associated with an abatement of APOE4-associated Aβ aggregation, suggesting KL-VS heterozygosity confers protections against APOE4-linked pathways to disease onset in AD.
Collapse
Affiliation(s)
- Claire M Erickson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Stephanie A Schultz
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Jennifer M Oh
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Burcu F Darst
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Yue Ma
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Derek Norton
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Tobey Betthauser
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Catherine L Gallagher
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Cynthia M Carlsson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Barbara B Bendlin
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sanjay Asthana
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bruce P Hermann
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Mark A Sager
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Kaj Blennow
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Henrik Zetterberg
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Corinne D Engelman
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bradley T Christian
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sterling C Johnson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Dena B Dubal
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Ozioma C Okonkwo
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco.
| |
Collapse
|
106
|
Lois C, Gonzalez I, Johnson KA, Price JC. PET imaging of tau protein targets: a methodology perspective. Brain Imaging Behav 2019; 13:333-344. [PMID: 29497982 PMCID: PMC6119534 DOI: 10.1007/s11682-018-9847-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The two neuropathological hallmarks of Alzheimer's disease (AD) are amyloid-[Formula: see text] plaques and neurofibrillary tangles of tau protein. Fifteen years ago, Positron Emission Tomography (PET) with Pittsburgh Compound B (11C-PiB) enabled selective in-vivo visualization of amyloid-[Formula: see text] plaque deposits and has since provided valuable information about the role of amyloid-[Formula: see text] deposition in AD. The progression of tau deposition has been shown to be highly associated with neuronal loss, neurodegeneration, and cognitive decline. Until recently it was not possible to visualize tau deposition in-vivo, but several tau PET tracers are now available in different stages of clinical development. To date, no tau tracer has been approved by the Food and Drug Administration for use in the evaluation of AD or other tauopathies, despite very active research efforts. In this paper we review the recent developments in tau PET imaging with a focus on in-vivo findings in AD and discuss the challenges associated with tau tracer development, the status of development and validation of different tau tracers, and the clinical information these provide.
Collapse
Affiliation(s)
- Cristina Lois
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA.
| | - Ivan Gonzalez
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Athinoula A. Martinos Center for Biomedical Research, Department of Radiology, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA
| |
Collapse
|
107
|
Wu M, Thurston RC, Tudorascu DL, Karim HT, Mathis CA, Lopresti BJ, Kamboh MI, Cohen AD, Snitz BE, Klunk WE, Aizenstein HJ. Amyloid deposition is associated with different patterns of hippocampal connectivity in men versus women. Neurobiol Aging 2019; 76:141-150. [PMID: 30711677 PMCID: PMC6584958 DOI: 10.1016/j.neurobiolaging.2018.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 10/04/2018] [Accepted: 11/18/2018] [Indexed: 01/26/2023]
Abstract
Compared to men, women are disproportionally affected by Alzheimer's disease (AD) and have an accelerated trajectory of cognitive decline and disease progression. Neurobiological factors underlying gender differences in AD remain unclear. This study investigated brain beta-amyloid (Aβ)-related neural system differences in cognitively normal older men and women (N = 61; 41 females, 65-93 years old). We found that men and women showed different associations between Aβ load and hippocampal functional connectivity. During associative memory encoding, in men greater Aβ burden was accompanied by greater hippocampus-prefrontal connectivity (i.e., more synchronized activities), whereas in women hippocampal connectivity did not vary by Aβ burden. For resting-state data, the interaction of gender × Aβ on hippocampal connectivity did not survive multiple comparison in the whole-brain analyses. In the region of interest-based analyses, resting-state hippocampal-prefrontal connectivity was positively correlated with Aβ load in men and was negatively correlated with Aβ load in women. The observed Aβ-related neural differences may explain the accelerated trajectory of cognitive decline and AD progression in women.
Collapse
Affiliation(s)
- Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Departments of Epidemiology and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Departments of Medicine and Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Helmet T Karim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
108
|
Heeman F, Yaqub M, Lopes Alves I, Heurling K, Berkhof J, Gispert JD, Bullich S, Foley C, Lammertsma AA. Optimized dual-time-window protocols for quantitative [ 18F]flutemetamol and [ 18F]florbetaben PET studies. EJNMMI Res 2019; 9:32. [PMID: 30919133 PMCID: PMC6437225 DOI: 10.1186/s13550-019-0499-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background A long dynamic scanning protocol may be required to accurately measure longitudinal changes in amyloid load. However, such a protocol results in a lower patient comfort and scanning efficiency compared to static scans. A compromise can be achieved by implementing dual-time-window protocols. This study aimed to optimize these protocols for quantitative [18F]flutemetamol and [18F]florbetaben studies. Methods Rate constants for subjects across the Alzheimer’s disease spectrum (i.e., non-displaceable binding potential (BPND) in the range 0.02–0.77 and 0.02–1.04 for [18F]flutemetamol and [18F]florbetaben, respectively) were established based on clinical [18F]flutemetamol (N = 6) and [18F]florbetaben (N = 20) data, and used to simulate tissue time-activity curves (TACs) of 110 min using a reference tissue and plasma input model. Next, noise was added (N = 50) and data points corresponding to different intervals were removed from the TACs, ranging from 0 (i.e., 90–90 = full-kinetic curve) to 80 (i.e., 10–90) minutes, creating a dual-time-window. Resulting TACs were fitted using the simplified reference tissue method (SRTM) to estimate the BPND, outliers (≥ 1.5 × BPND max) were removed and the bias was assessed using the distribution volume ratio (DVR = BPND + 1). To this end, acceptability curves, which display the fraction of data below a certain bias threshold, were generated and the area under those curves were calculated. Results [18F]Flutemetamol and [18F]florbetaben data demonstrated an increased bias in amyloid estimate for larger intervals and higher noise levels. An acceptable bias (≤ 3.1%) in DVR could be obtained with all except the 10–90 and 20–90-min intervals. Furthermore, a reduced fraction of acceptable data and most outliers were present for these two largest intervals (maximum percentage outliers 48 and 32 for [18F]flutemetamol and [18F]florbetaben, respectively). Conclusions The length of the interval inversely correlates with the accuracy of the BPND estimates. Consequently, a dual-time-window protocol of 0–30 and 90–110 min (=maximum of 60 min interval) allows for accurate estimation of BPND values for both tracers. [18F]flutemetamol: EudraCT 2007-000784-19, registered 8 February 2007, [18F]florbetaben: EudraCT 2006-003882-15, registered 2006. Electronic supplementary material The online version of this article (10.1186/s13550-019-0499-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fiona Heeman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands.
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Isadora Lopes Alves
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Johannes Berkhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Epidemiology and Biostatistics, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Carrer de Wellington, 30, 08005, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.,Universitat Pompeu Fabra, Plaça de la Mercè, 10, 08002, Barcelona, Spain
| | - Santiago Bullich
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353, Berlin, Germany
| | | | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan, 1117, Amsterdam, Netherlands
| | | |
Collapse
|
109
|
Zou R, Kuang G, Ågren H, Nordberg A, Långström B, Tu Y. Free Energy Profile for Penetration of Pittsburgh Compound-B into the Amyloid β Fibril. ACS Chem Neurosci 2019; 10:1783-1790. [PMID: 30698013 DOI: 10.1021/acschemneuro.8b00662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The amyloid β (Aβ) fibril is a hallmark of Alzheimer's disease (AD) and has therefore served as an important target for early diagnosis of AD. The Pittsburgh Compound-B (PiB) is one of the most famous positron emission tomography (PET) tracers commonly used for in vivo detection of Aβ fibrils. Many theoretical studies have predicted the existence of various core binding sites with different microenvironments for probes binding to the Aβ fibril. However, little attention has been devoted to how the probes actually penetrate into the different core binding sites. In this study, an integrated molecular modeling scheme is used to study the penetration of PiB into the core binding sites of the Aβ1-42 fibril structure recently obtained by cryogenic electron microscopy. We find that there are two core binding sites for PiB with dramatic differences in cavity size and microenvironment properties, and furthermore that the penetration of PiB into site-1 is energetically prohibitive, whereas the penetration into site-2 is much more favorable. Therefore, the binding capacity at site-2 may be larger than that at site-1 despite its lower binding affinity. Our results thus suggest that site-2 may be a major binding site for PiB binding to Aβ fibril and emphasize the importance to adopt a full dynamical picture when studying tracer-fibril binding problems in general, something that in turn can be used to guide the development of tracers with higher affinity and selectivity for the Aβ fibril.
Collapse
Affiliation(s)
- Rongfeng Zou
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Guanglin Kuang
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Hans Ågren
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center of Alzheimer Research, Clinical Geriatrics, Neo and Theme Aging, Karolinska University Hospital, Karolinska Institute, 141 83 Huddinge, Sweden
| | - Bengt Långström
- Department of Chemistry—BMC, Physical Organic Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Yaoquan Tu
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| |
Collapse
|
110
|
Yamauchi H, Kagawa S, Takahashi M, Oishi N, Ono M, Higashi T. Misery perfusion and amyloid deposition in atherosclerotic major cerebral artery disease. NEUROIMAGE-CLINICAL 2019; 22:101762. [PMID: 30884364 PMCID: PMC6424140 DOI: 10.1016/j.nicl.2019.101762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/08/2019] [Accepted: 03/10/2019] [Indexed: 11/18/2022]
Abstract
Although experimental studies have shown that global cerebral hypoperfusion leads to amyloid deposition in the hemisphere with carotid artery occlusion in rodents, the results of such occurrence are controversial in humans. Hence, we aim to determine whether global cerebral hypoperfusion leading to decreased blood flow relative to metabolic demand [increased oxygen extraction fraction (OEF), misery perfusion] is associated with increases in amyloid deposition in the hemisphere with atherosclerotic major cerebral artery disease in patients. We evaluated the distribution of β-amyloid plaques using positron emission tomography and a [18F]-pyridylbenzofuran derivative (18F-FPYBF-2) in 13 patients with unilateral atherosclerotic disease of the internal carotid artery (ICA) or middle cerebral artery (MCA) disease and no cortical infarction. The distribution volume ratio (DVR) of 18F- FPYBF-2 was calculated using dynamic data and Logan graphical analysis with reference tissue and was correlated with the cerebral blood flow (CBF), cerebral metabolic rate of oxygen (CMRO2), and OEF, obtained from 15O-gas PET. The mean cortical value was calculated as the mean value within the frontal, posterior cingulate, precuneus, parietal, and lateral temporal cortical regions. Significant reductions in CBF and CMRO2 and increases in OEF were found in the hemisphere ipsilateral to the arterial lesion compared with the contralateral hemisphere. There was no significant difference for 18F-FPYBF-2 DVR between hemispheres. The ipsilateral to contralateral ratio of the 18F- FPYBF-2 DVR was increased in 3 patients, while the ipsilateral to contralateral OEF ratio was increased in 4 patients. The incidence of an increased hemispheric DVR ratio was significantly higher in patients with an increased hemispheric OEF ratio (3/4) than in patients without (0/9) (p < 0.02). Although the 18F- FPYBF-2 DVR in the ipsilateral hemisphere was positively correlated with OEF after adjustment for the 18F- FPYBF-2 DVR in the contralateral hemisphere using multiple regression analysis (p < 0.05), the contribution rate of OEF was small (R2 = 5.5%). Only one of the 4 patients with an increased hemispheric OEF ratio showed amyloid positivity based on the DVR value. In atherosclerotic major cerebral artery disease, misery perfusion accompanied only small increases of amyloid deposition at best. Misery perfusion was not associated with amyloid positivity. Misery perfusion accompanied only small increases of amyloid deposition at best. Relative oxygen extraction fraction correlated with relative amyloid deposition. Misery perfusion was not associated with amyloid positivity.
Collapse
Affiliation(s)
- Hiroshi Yamauchi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan.
| | - Shinya Kagawa
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan
| | - Masaaki Takahashi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan
| | - Naoya Oishi
- Human Brain Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Tatsuya Higashi
- Division of PET Imaging, Shiga Medical Centre Research Institute, Moriyama, Japan; National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
111
|
Cysouw MCF, Golla SVS, Frings V, Smit EF, Hoekstra OS, Kramer GM, Boellaard R. Partial-volume correction in dynamic PET-CT: effect on tumor kinetic parameter estimation and validation of simplified metrics. EJNMMI Res 2019; 9:12. [PMID: 30715647 PMCID: PMC6362178 DOI: 10.1186/s13550-019-0483-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/25/2019] [Indexed: 12/27/2022] Open
Abstract
Background Partial-volume effects generally result in an underestimation of tumor tracer uptake on PET-CT for small lesions, necessitating partial-volume correction (PVC) for accurate quantification. However, investigation of PVC in dynamic oncological PET studies to date is scarce. The aim of this study was to investigate PVC’s impact on tumor kinetic parameter estimation from dynamic PET-CT acquisitions and subsequent validation of simplified semi-quantitative metrics. Ten patients with EGFR-mutated non-small cell lung cancer underwent dynamic 18F-fluorothymidine PET-CT before, 7 days after, and 28 days after commencing treatment with a tyrosine kinase inhibitor. Parametric PVC was applied using iterative deconvolution without and with highly constrained backprojection (HYPR) denoising, respectively. Using an image-derived input function with venous parent plasma calibration, we estimated full kinetic parameters VT, K1, and k3/k4 (BPND) using a reversible two-tissue compartment model, and simplified metrics (SUV and tumor-to-blood ratio) at 50–60 min post-injection. Results PVC had a non-linear effect on measured activity concentrations per timeframe. PVC significantly changed each kinetic parameter, with a median increase in VT of 11.8% (up to 25.1%) and 10.8% (up to 21.7%) without and with HYPR, respectively. Relative changes in kinetic parameter estimates vs. simplified metrics after applying PVC were poorly correlated (correlations 0.36–0.62; p < 0.01). PVC increased correlations between simplified metrics and VT from 0.82 and 0.81 (p < 0.01) to 0.90 and 0.88 (p < 0.01) for SUV and TBR, respectively, albeit non-significantly. PVC also increased correlations between treatment-induced changes in simplified metrics vs. VT at 7 (SUV) and 28 (SUV and TBR) days after treatment start non-significantly. Delineation on partial-volume corrected PET images resulted in a median decrease in metabolic tumor volume of 14.3% (IQR − 22.1 to − 7.5%), and increased the effect of PVC on kinetic parameter estimates. Conclusion PVC has a significant impact on tumor kinetic parameter estimation from dynamic PET-CT data, which differs from its effect on simplified metrics. However, it affected validation of these simplified metrics both as single measurements and as biomarkers of treatment response only to a small extent. Future dynamic PET studies should preferably incorporate PVC. Trial registration Dutch Trial Register, NTR3557. Electronic supplementary material The online version of this article (10.1186/s13550-019-0483-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M C F Cysouw
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - S V S Golla
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - V Frings
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - E F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands
| | - O S Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - G M Kramer
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - R Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | | |
Collapse
|
112
|
Peretti DE, Vállez García D, Reesink FE, van der Goot T, De Deyn PP, de Jong BM, Dierckx RAJO, Boellaard R. Relative cerebral flow from dynamic PIB scans as an alternative for FDG scans in Alzheimer's disease PET studies. PLoS One 2019; 14:e0211000. [PMID: 30653612 PMCID: PMC6336325 DOI: 10.1371/journal.pone.0211000] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/04/2019] [Indexed: 11/29/2022] Open
Abstract
In Alzheimer's Disease (AD) dual-tracer positron emission tomography (PET) studies with 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) and 11C-labelled Pittsburgh Compound B (PIB) are used to assess metabolism and cerebral amyloid-β deposition, respectively. Regional cerebral metabolism and blood flow (rCBF) are closely coupled, both providing an index for neuronal function. The present study compared PIB-derived rCBF, estimated by the ratio of tracer influx in target regions relative to reference region (R1) and early-stage PIB uptake (ePIB), to FDG scans. Fifteen PIB positive (+) patients and fifteen PIB negative (-) subjects underwent both FDG and PIB PET scans to assess the use of R1 and ePIB as a surrogate for FDG. First, subjects were classified based on visual inspection of the PIB PET images. Then, discriminative performance (PIB+ versus PIB-) of rCBF methods were compared to normalized regional FDG uptake. Strong positive correlations were found between analyses, suggesting that PIB-derived rCBF provides information that is closely related to what can be seen on FDG scans. Yet group related differences between method's distributions were seen as well. Also, a better correlation with FDG was found for R1 than for ePIB. Further studies are needed to validate the use of R1 as an alternative for FDG studies in clinical applications.
Collapse
Affiliation(s)
- Débora E. Peretti
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - Fransje E. Reesink
- Department of Neurology, Alzheimer Research Centre, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - Tim van der Goot
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - Peter P. De Deyn
- Department of Neurology, Alzheimer Research Centre, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
- Institute Born-Bunge, Laboratory of Neurochemistry and Behaviour, University of Antwerp, Antwerp, Antwerp, Belgium
| | - Bauke M. de Jong
- Department of Neurology, Alzheimer Research Centre, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Groningen, The Netherlands
| |
Collapse
|
113
|
Harrison TM, La Joie R, Maass A, Baker SL, Swinnerton K, Fenton L, Mellinger TJ, Edwards L, Pham J, Miller BL, Rabinovici GD, Jagust WJ. Longitudinal tau accumulation and atrophy in aging and alzheimer disease. Ann Neurol 2019; 85:229-240. [PMID: 30597624 PMCID: PMC6579738 DOI: 10.1002/ana.25406] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/26/2018] [Accepted: 12/22/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine the rate of tau accumulation in healthy older adults (OA) and patients with Alzheimer disease (AD), as well as the relationship of tau accumulation to cortical atrophy. METHODS Two longitudinal flortaucipir (FTP) positron emission tomography (PET) and magnetic resonance imaging (MRI) scans were acquired from 42 OA (21 Pittsburg compound B [PiB]+ , age = 77.6 ± 4.6 years, 25 female [F]/17 male [M]) and 19 PiB+ patients with AD (age = 63.1 ± 10.3 years, 12 F/7 M) over 1 to 3 years of follow-up. FTP change, structural MRI measures of atrophy, and cross-modal correlations were examined on a voxelwise level. Regional annual percentage change in FTP was also calculated. RESULTS Voxelwise FTP change in AD showed the greatest increases in lateral and medial frontal lobes. Atrophy over the same interval was more widespread and included posteromedial cortical areas, where tau accumulation rates were lower. In OA, FTP binding increased in bilateral temporal lobe and retrosplenial cortex, accompanied by atrophy in the same regions. There were no associations between voxelwise change in FTP and sex, PiB, or APOE. Regional FTP significantly increased at follow-up in OA and patients with AD. Mixed effects models showed greater FTP increases in AD compared to OA, and no differences within OA based on PiB status. INTERPRETATION Our findings indicate that tau accumulates even in amyloid-negative healthy OA and this process can be measured with in vivo tau-PET. In OA, tau accumulation and atrophy share a similar topography. In AD, tau increases more rapidly and accumulation occurs in frontal regions that are not yet undergoing significant atrophy. Ann Neurol 2019; 1-12 ANN NEUROL 2019;85:229-240.
Collapse
Affiliation(s)
- Theresa M Harrison
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Anne Maass
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA.,German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | | | - Kaitlin Swinnerton
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | - Laura Fenton
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA
| | - Taylor J Mellinger
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Lauren Edwards
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Julie Pham
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Bruce L Miller
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA.,Lawrence Berkeley National Laboratory, Berkeley, CA
| |
Collapse
|
114
|
Abstract
PURPOSE Longitudinal studies into the variability of F-Flutemetamol uptake are lacking. METHODS/PATIENTS Therefore, the current study examined change in F-Flutemetamol uptake in 19 nondemented older adults (65 to 82 y old) who were either cognitively intact or had Mild Cognitive Impairment (MCI) who were scanned twice across 3.6 years. RESULTS Baseline and follow-up composite SUVRs were significantly correlated (0.96, P<0.001). Significant increases in the composite SUVR from baseline to follow-up were observed (P=0.002). For the total sample, the average difference over this time period when using the composite SUVR was 6.8%. Similar results were seen in subsets of the total sample (MCI vs. cognitively intact, amyloid positive vs. negative). Finally, a Reliable Change Index that exceeded ±0.046 SUVR units would indicate a significant change of F-Flutemetamol. CONCLUSIONS The current results extend the limited literature on longitudinal variability of F-Flutemetamol uptake across 3.6 years, which should give clinicians and researchers more confidence in the stability of this amyloid imaging agent in longer therapeutic and prevention trials in cognitive decline in MCI and Alzheimer disease.
Collapse
|
115
|
Oeltzschner G, Wijtenburg SA, Mikkelsen M, Edden RAE, Barker PB, Joo JH, Leoutsakos JMS, Rowland LM, Workman CI, Smith GS. Neurometabolites and associations with cognitive deficits in mild cognitive impairment: a magnetic resonance spectroscopy study at 7 Tesla. Neurobiol Aging 2019; 73:211-218. [PMID: 30390554 PMCID: PMC6294473 DOI: 10.1016/j.neurobiolaging.2018.09.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/05/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022]
Abstract
The levels of several brain metabolites were investigated in the anterior cingulate cortex (ACC) and posterior cingulate cortex (PCC) in 13 healthy controls (HC) and 13 patients with mild cognitive impairment (MCI) using single-voxel magnetic resonance spectroscopy at 7T. Levels of γ-aminobutyric acid (GABA), glutamate (Glu), glutathione (GSH), N-acetylaspartylglutamate (NAAG), N-acetylaspartate (NAA), and myo-inositol (mI) were quantified relative to total creatine (tCr). The effect of diagnosis on metabolite levels, and relationships between metabolite levels and memory and executive function, correcting for age, were investigated. MCI patients showed significantly decreased GABA/tCr (ACC, PCC), Glu/tCr (PCC), and NAA/tCr (PCC), and significantly increased mI/tCr (ACC). In the combined group, worse episodic verbal memory performance was correlated with lower Glu/tCr (PCC), lower NAA/tCr (PCC), and higher mI/tCr (ACC, PCC). Worse verbal fluency performance was correlated with lower GSH/tCr (PCC). In summary, MCI is associated with decreased GABA and Glu, most consistently in the PCC. Further studies in larger patient samples should be undertaken to determine the utility of 7T magnetic resonance spectroscopy in detecting MCI-related neurochemical changes.
Collapse
Affiliation(s)
- Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - S Andrea Wijtenburg
- Department of Psychiatry, Neuroimaging Research Program, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Peter B Barker
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Jin Hui Joo
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeannie-Marie S Leoutsakos
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura M Rowland
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Psychiatry, Neuroimaging Research Program, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Clifford I Workman
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gwenn S Smith
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
116
|
Tudorascu DL, Anderson SJ, Minhas DS, Yu Z, Comer D, Lao P, Hartley S, Laymon CM, Snitz BE, Lopresti BJ, Johnson S, Price JC, Mathis CA, Aizenstein HJ, Klunk WE, Handen BL, Christian BT, Cohen AD. Comparison of longitudinal Aβ in nondemented elderly and Down syndrome. Neurobiol Aging 2019; 73:171-176. [PMID: 30359879 PMCID: PMC6251757 DOI: 10.1016/j.neurobiolaging.2018.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/21/2018] [Accepted: 09/23/2018] [Indexed: 12/31/2022]
Abstract
Down syndrome (DS) predisposes individuals to early Alzheimer's disease (AD). Using Pittsburgh Compound B ([11C]PiB), a pattern of striatal amyloid beta (Aβ) that is elevated relative to neocortical binding has been reported, similar to that of nondemented autosomal dominant AD mutation carriers. However, it is not known whether changes in striatal and neocortical [11C]PiB retention differ over time in a nondemented DS population when compared to changes in a nondemented elderly (NDE) population. The purpose of this work was to assess longitudinal changes in trajectories of Aβ in a nondemented DS compared to an NDE cohort. The regional trajectories for anterior ventral striatum (AVS), frontal cortex, and precuneus [11C]PiB retention were explored over time using linear mixed effects models with fixed effects of time, cohort, and time-by-cohort interactions and subject as random effects. Significant differences between DS and NDE cohort trajectories for all 3 region of interests were observed (p < 0.05), with the DS cohort showing a faster accumulation in the AVS and slower accumulation in the frontal cortex and precuneus compared to the NDE cohort. These data add to the previously reported distinct pattern of early striatal deposition not commonly seen in sporadic AD by demonstrating that individuals with DS may also accumulate Aβ at a rate faster in the AVS when compared to NDE subjects.
Collapse
Affiliation(s)
- Dana L Tudorascu
- Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA; Department of Biostatistics, University of Pittsburgh, Graduate School of Public Health, Pittsburgh, PA, USA
| | - Stewart J Anderson
- Department of Biostatistics, University of Pittsburgh, Graduate School of Public Health, Pittsburgh, PA, USA
| | - Davneet S Minhas
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zheming Yu
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diane Comer
- Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Patrick Lao
- Department of Medical Physics, University of Wisconsin, Madison School of Medicine, Madison, WI, USA; Department of Waisman Center, University of Wisconsin, Madison School of Medicine, Madison, WI, USA
| | - Sigan Hartley
- Department of Waisman Center, University of Wisconsin, Madison School of Medicine, Madison, WI, USA; Departments of Human Development and Family Studies, University of Wisconsin, Madison School of Medicine, Madison, WI, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Biongeenering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sterling Johnson
- Department of Medical Physics, University of Wisconsin, Madison School of Medicine, Madison, WI, USA; Department of Medicine and Geriatrics, University of Wisconsin, Madison School of Medicine, Madison, WI, USA
| | - Julie C Price
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Massachusetts General Hospital, A. A. Martinos Center for Biomedical Imaging, Cambridge, MA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Brad T Christian
- Department of Medical Physics, University of Wisconsin, Madison School of Medicine, Madison, WI, USA; Department of Waisman Center, University of Wisconsin, Madison School of Medicine, Madison, WI, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
117
|
Lao PJ, Handen BL, Betthauser TJ, Cody KA, Cohen AD, Tudorascu DL, Stone CK, Price JC, Johnson SC, Klunk WE, Christian BT. Imaging neurodegeneration in Down syndrome: brain templates for amyloid burden and tissue segmentation. Brain Imaging Behav 2019; 13:345-353. [PMID: 29752653 PMCID: PMC6230506 DOI: 10.1007/s11682-018-9888-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The focus of Alzheimer's disease (AD) neuroimaging research has shifted towards an investigation of the earliest stages of AD pathogenesis, which manifests in every young adult with Down syndrome (DS; trisomy 21) resulting from a deterministic genetic predisposition to amyloid precursor protein overproduction. Due to morphological differences in brain structure in the DS population, special consideration must be given to processing pipelines and the use of normative atlases developed for the non-DS population. Further, the use of typical MRI to MRI template spatial normalization is less desirable in this cohort due to a greater presence of motion artefacts in MRI images. The diffuse nature of PiB uptake and comparatively lower spatial resolution of the PET image permits the purposing of this modality as a template for spatial normalization, which can substantially improve the robustness of this procedure in the cases of MRI images with motion. The aim of this work was to establish standardized methods for spatial normalization and tissue type segmentation using DS specific templates in order to perform voxel-wise analyses. A total of 72 adults with DS underwent [11C]PiB PET to assess brain amyloid burden and volumetric MRI imaging. A DS specific PiB template for spatial normalization and a set of DS specific prior probability templates were created with two-pass methods. With implementation of this DS specific PiB template, no participants were excluded due to poor spatial normalization, thus maximizing the sample size for PiB analyses in standardized space. In addition, difference images between prior probability templates created from the general population and the DS population reflected known morphological differences, particularly in the frontal cortex. In conclusion, DS specific templates that account for unique challenges improve spatial normalization and tissue type segmentation, and provide a framework for reliable voxel-wise analysis of AD biomarkers in this atypical population.
Collapse
Affiliation(s)
- Patrick J. Lao
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705 USA ,Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705 USA
| | - Ben L. Handen
- Department of Psychiatry, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213 USA ,Department of Pediatrics, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224 USA ,Department of Psychology, University of Pittsburgh, 201 South Bouquet Street, Pittsburgh, PA 15260 USA ,Department of Instruction and Learning, University of Pittsburgh, 230 South Bouquet Street, Pittsburgh, PA 15260 USA
| | - Tobey J. Betthauser
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705 USA ,Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705 USA
| | - Karly A. Cody
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705 USA
| | - Annie D. Cohen
- Department of Psychiatry, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213 USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213 USA ,Department of Internal Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,Department of Biostatistics, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Charles K. Stone
- Department of Cardiovascular Medicine, University of Wisconsin-Madison, 1 South Park Street, Madison, WI 53715 USA
| | - Julie C. Price
- Department of Radiology, University of Pittsburgh, 3600 Forbes @ Meyran Avenues, Pittsburgh, PA 15213 USA ,Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129 USA
| | - Sterling C. Johnson
- Department of Medicine-Geriatrics, University of Wisconsin-Madison, 1685 Highland Ave, Madison, WI 53705 USA
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213 USA ,Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Bradley T. Christian
- Department of Medical Physics, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705 USA ,Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705 USA ,Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Blvd, Madison, WI 53719 USA
| |
Collapse
|
118
|
d'Oleire Uquillas F, Jacobs HIL, Biddle KD, Properzi M, Hanseeuw B, Schultz AP, Rentz DM, Johnson KA, Sperling RA, Donovan NJ. Regional tau pathology and loneliness in cognitively normal older adults. Transl Psychiatry 2018; 8:282. [PMID: 30563962 PMCID: PMC6299114 DOI: 10.1038/s41398-018-0345-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Loneliness is a perception of social and emotional isolation that increases in prevalence among older adults during the eighth decade of life. Loneliness has been associated with higher brain amyloid-β deposition, a biologic marker of Alzheimer's disease, in cognitively normal older adults, suggesting a link with preclinical Alzheimer's disease pathophysiology. This study examined whether greater loneliness was associated with tau pathology, the other defining feature of Alzheimer's disease, in 117 cognitively normal older adults. Using flortaucipir positron emission tomography, we measured tau pathology in the entorhinal cortex, a region of initial accumulation in aging adults with or without elevated amyloid-β, and in the inferior temporal cortex, a region of early accumulation typically associated with elevated amyloid-β and memory impairment. Loneliness was measured by self-report using the 3-item UCLA-loneliness scale. We found that higher tau pathology in the right entorhinal cortex was associated with greater loneliness, controlling for age, sex, and apolipoprotein E ε4, the Alzheimer's disease genetic risk marker. This association remained significant after further adjustment for socioeconomic status, social network, depression and anxiety scores, and memory performance. There was no association of inferior temporal cortical or left entorhinal tau pathology with loneliness. Exploratory whole-brain surface maps supported these findings and identified additional clusters correlating loneliness and tau in the right fusiform gyrus. These results provide further support for loneliness as a socioemotional symptom in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
| | - Heidi I L Jacobs
- Division of Nuclear Medicine, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- School for Mental Health and Neuroscience, Alzheimer Centre, Limburg, Maastricht University, Maastricht, The Netherlands
| | - Kelsey D Biddle
- Department of Psychiatry, Division of Geriatric Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Bernard Hanseeuw
- Division of Nuclear Medicine, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Neurology, Saint-Luc University Hospital, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Psychiatry, Division of Geriatric Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Division of Nuclear Medicine, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nancy J Donovan
- Department of Psychiatry, Division of Geriatric Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
119
|
Cohen AD, Landau SM, Snitz BE, Klunk WE, Blennow K, Zetterberg H. Fluid and PET biomarkers for amyloid pathology in Alzheimer's disease. Mol Cell Neurosci 2018; 97:3-17. [PMID: 30537535 DOI: 10.1016/j.mcn.2018.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid plaques and tau pathology (neurofibrillary tangles and neuropil threads). Amyloid plaques are primarily composed of aggregated and oligomeric β-amyloid (Aβ) peptides ending at position 42 (Aβ42). The development of fluid and PET biomarkers for Alzheimer's disease (AD), has allowed for detection of Aβ pathology in vivo and marks a major advancement in understanding the role of Aβ in Alzheimer's disease (AD). In the recent National Institute on Aging and Alzheimer's Association (NIA-AA) Research Framework, AD is defined by the underlying pathology as measured in patients during life by biomarkers (Jack et al., 2018), while clinical symptoms are used for staging of the disease. Therefore, sensitive, specific and robust biomarkers to identify brain amyloidosis are central in AD research. Here, we discuss fluid and PET biomarkers for Aβ and their application.
Collapse
Affiliation(s)
- Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, United States of America.
| | - Susan M Landau
- Neurology Helen Wills Neuroscience Institute, University of California, Berkeley, United States of America; Lawrence Berkeley National Laboratory, Molecular Biophysics and Integrated Bioimaging Functional Imaging Department, Life Sciences Division, United States of America
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh School of Medicine, United States of America
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, United States of America
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, University College, London, United Kingdom of Great Britain and Northern Ireland
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, University College, London, United Kingdom of Great Britain and Northern Ireland; Department of Molecular Neuroscience, UCL Institute of Neurology, United Kingdom of Great Britain and Northern Ireland; UK Dementia Research Institute at UCL, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
120
|
Faria DDP, Duran FL, Squarzoni P, Coutinho AM, Garcez AT, Santos PP, Brucki SM, de Oliveira MO, Trés ES, Forlenza OV, Nitrini R, Buchpiguel CA, Busatto Filho G. Topography of 11C-Pittsburgh compound B uptake in Alzheimer's disease: a voxel-based investigation of cortical and white matter regions. ACTA ACUST UNITED AC 2018; 41:101-111. [PMID: 30540022 PMCID: PMC6781685 DOI: 10.1590/1516-4446-2017-0002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/06/2018] [Indexed: 01/09/2023]
Abstract
Objective: To compare results of positron emission tomography (PET) with carbon-11-labeled Pittsburgh compound B (11C-PIB) obtained with cerebellar or global brain uptake for voxel intensity normalization, describe the cortical sites with highest tracer uptake in subjects with mild Alzheimer’s disease (AD), and explore possible group differences in 11C-PIB binding to white matter. Methods: 11C-PIB PET scans were acquired from subjects with AD (n=17) and healthy elderly controls (n=19). Voxel-based analysis was performed with statistical parametric mapping (SPM). Results: Cerebellar normalization showed higher 11C-PIB uptake in the AD group relative to controls throughout the cerebral cortex, involving the lateral temporal, orbitofrontal, and superior parietal cortices. With global uptake normalization, greatest cortical binding was detected in the orbitofrontal cortex; decreased 11C-PIB uptake in white matter was found in the posterior hippocampal region, corpus callosum, pons, and internal capsule. Conclusion: The present case-control voxelwise 11C-PIB PET comparison highlighted the regional distribution of amyloid deposition in the cerebral cortex of mildly demented AD patients. Tracer uptake was highest in the orbitofrontal cortex. Decreased 11C-PIB uptake in white-matter regions in this patient population may be a marker of white-matter damage in AD.
Collapse
Affiliation(s)
- Daniele de P Faria
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil
| | - Fabio L Duran
- Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil.,Laboratório de Neuroimagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Paula Squarzoni
- Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil.,Laboratório de Neuroimagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Artur M Coutinho
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil
| | - Alexandre T Garcez
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil
| | - Pedro P Santos
- Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil.,Laboratório de Neuroimagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Sonia M Brucki
- Departamento de Neurologia, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Maira O de Oliveira
- Departamento de Neurologia, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Eduardo S Trés
- Departamento de Neurologia, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Orestes V Forlenza
- Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil.,Laboratório de Neurociências (LIM 27), Departamento de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Ricardo Nitrini
- Departamento de Neurologia, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Carlos A Buchpiguel
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil.,Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil
| | - Geraldo Busatto Filho
- Núcleo de Apoio à Pesquisa em Neurociência Aplicada (NAPNA), USP, São Paulo, SP, Brazil.,Laboratório de Neuroimagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| |
Collapse
|
121
|
Ponto LLB, Moser DJ, Menda Y, Harlynn EL, DeVries SD, Oleson JJ, Magnotta VA, Schultz SK. Early Phase PIB-PET as a Surrogate for Global and Regional Cerebral Blood Flow Measures. J Neuroimaging 2018; 29:85-96. [PMID: 30461110 DOI: 10.1111/jon.12582] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/01/2018] [Accepted: 11/05/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE To explore the potential for simplified measures of [11 C]PIB uptake to serve as a surrogate for cerebral blood flow (CBF) measures, thereby, providing both pathological and functional information in the same scan. METHODS Participants (N = 24, 16 M, 8 F, 57-87 years) underwent quantitative [15 O]water imaging and dynamic [11 C]PIB imaging. Time-activity curves were created for each participant's regional [11 C]PIB data scaled in standardized uptake values (SUVs). The frame in which maximal uptake occurred was defined for each subject (ie, "peak"). The concentration (SUV) for each region at the individual's peak, during the 3.5-4 minute time interval and for the initial 6 minute sum, was determined. R1 (ie, relative delivery using cerebellum as reference tissue) from the simplified reference tissue model 2 was determined for each region. PIB SUVs were compared to the absolute CBF global and regional values (in mL/minute/100 mL) and the R1 values were compared to the cerebellar-normalized rCBF. RESULTS Significant linear relationships were found for all SUV measures with measures of absolute global and regional CBF that were comparable to the relationship between normalized CBF and R1. The individual SUVpeak exhibited the strongest relationship both regionally and globally. All individuals and all regions had highly significant regression slopes. Age, gender, or amyloid burden did not influence the relationship. CONCLUSION Early PIB uptake has the potential to effectively serve as a surrogate for global and regional CBF measures. The simple and readily obtainable individual's SUVpeak value was the strongest predictor regionally and globally of CBF.
Collapse
Affiliation(s)
- Laura L Boles Ponto
- Department of Radiology, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| | - David J Moser
- Department of Psychiatry, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| | - Yusuf Menda
- Department of Radiology, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| | - Emily L Harlynn
- Department of Radiology, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA.,Department of Psychiatry, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| | - Sean D DeVries
- Department of Biostatistics, University of Iowa College of Public Health, Iowa City, IA
| | - Jacob J Oleson
- Department of Biostatistics, University of Iowa College of Public Health, Iowa City, IA
| | - Vincent A Magnotta
- Department of Radiology, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| | - Susan K Schultz
- Department of Psychiatry, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA
| |
Collapse
|
122
|
Nai YH, Watanuki S, Tashiro M, Okamura N, Watabe H. Investigation of the quantitative accuracy of low-dose amyloid and tau PET imaging. Radiol Phys Technol 2018; 11:451-459. [PMID: 30328073 DOI: 10.1007/s12194-018-0485-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Abstract
With the increasing incidence of dementia worldwide, the frequent use of amyloid and tau positron emission tomography imaging requires low-dose protocols for the differential diagnoses of various neurodegenerative diseases and the monitoring of disease progression. In this study, we investigated the feasibility to reduce the PET dose without a significant loss of quantitative accuracy in 3D dynamic row action maximum likelihood algorithm-reconstructed PET images using [11C]PIB and [18F]THK5351. Eighteen cognitively normal young controls, cognitively normal elderly controls, and patients with probable Alzheimer's disease (n = 6 each), were included. Reduced doses were simulated by randomly sampling half and quarter of the full counts in list mode data for one independent realization at each simulated dose. Bias was evaluated between the reduced dose from the full dose of standardized uptake value ratio (SUVR), distribution volume ratio (DVR) from reference Logan, and non-displaceable binding potential (BPND) from simplified reference tissue model (SRTM). DVR yielded the least bias at low dose compared to SUVR and BPND, and thus, is highly recommended. The dose of [18F]THK5351 and [11C]PIB can be reduced to a quarter of the full dose using DVR for evaluation, whereas the dose can only be reduced to half and a quarter of the full dose for [18F]THK5351 and [11C]PIB using SUVR. BPND showed inconsistent trend and large bias at low dose. The feasibility of dose reduction was dependent on the selected parameters of interest, reconstruction algorithms, reference regions, and to a lesser degree by motion effects.
Collapse
Affiliation(s)
- Ying-Hwey Nai
- Division of Radiation Informatics for Medical Imaging, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan.,Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Shoichi Watanuki
- Division of Cyclotron Nuclear Medicine, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Manabu Tashiro
- Division of Cyclotron Nuclear Medicine, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | | | - Hiroshi Watabe
- Division of Radiation Informatics for Medical Imaging, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan. .,Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
| |
Collapse
|
123
|
Amariglio RE, Buckley RF, Mormino EC, Marshall GA, Johnson KA, Rentz DM, Sperling RA. Amyloid-associated increases in longitudinal report of subjective cognitive complaints. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2018; 4:444-449. [PMID: 30258973 PMCID: PMC6153378 DOI: 10.1016/j.trci.2018.08.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction To investigate whether baseline subjective cognitive complaints (SCCs) predict longitudinal decline on neuropsychological testing and whether SCC increases longitudinally, in the setting of high levels of amyloid burden. Methods Two hundred seventy-nine clinically normal older participants (mean age = 73.7 ± 6.1 years) from the Harvard Aging Brain Study, a cohort of community-dwelling individuals, were followed longitudinally (4.27 ± 1.35 years) with annual subjective memory questionnaires and neuropsychological assessment. 11C Pittsburgh compound-B positron emission tomography was used to measure cortical amyloid and to classify status (Aβ+/Aβ−) at baseline. Results Higher baseline SCC predicted more rapid cognitive decline on neuropsychological measures among those with elevated amyloid (t = −2.18, P < .0001). In addition, longitudinal report of SCC significantly increased over time, with SCC progression most pronounced among Aβ+ individuals (t = 2.24, P = .0005). Discussion SCC may inform risk for future cognitive decline and track progression of self-perceived decline, particularly in those along the AD trajectory, providing potentially important indicators of clinical meaningfulness in AD prevention trials.
Collapse
Affiliation(s)
- Rebecca E. Amariglio
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author. Tel.: +(617) 732-8085. Fax: (617) 738-9122.
| | - Rachel F. Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Florey Institute, University of Melbourne, Parkville, Victoria, Australia
- Melbourne School of Psychological Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth C. Mormino
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Stanford Medical School, Palo Alto, CA, USA
| | - Gad A. Marshall
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A. Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorene M. Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A. Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
124
|
The Relationship of Current Cognitive Activity to Brain Amyloid Burden and Glucose Metabolism. Am J Geriatr Psychiatry 2018; 26:977-984. [PMID: 29885987 PMCID: PMC6482956 DOI: 10.1016/j.jagp.2018.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/20/2018] [Accepted: 03/29/2018] [Indexed: 11/20/2022]
Abstract
Several studies have investigated how lifetime cognitive engagement affects levels of amyloid-beta (Aβ) deposition in the brain. However, there has been some disagreement, leaving the relationship of cognitive activity (CA) to Aβ a largely open question. The present study investigated the relationship between CA, Aβ deposition, and glucose metabolism. One hundred nine cognitively normal participants underwent Pittsburgh Compound-B (PiB) and [18F]fluorodeoxyglucose-positron emission tomography and completed a questionnaire designed to measure current CA. Statistical analyses revealed significant differences in PiB retention between those in the high and low CA groups. Linear regression models revealed a significant negative relationship between PiB retention and CA and a significant positive relationship between glucose metabolism and CA. These data suggest that CA may have a direct beneficial effect on the pathophysiology of AD or reflect another underlying process that results in both higher CA and lower AD pathophysiology.
Collapse
|
125
|
Data driven diagnostic classification in Alzheimer's disease based on different reference regions for normalization of PiB-PET images and correlation with CSF concentrations of Aβ species. NEUROIMAGE-CLINICAL 2018; 20:603-610. [PMID: 30186764 PMCID: PMC6120605 DOI: 10.1016/j.nicl.2018.08.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/01/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Positron emission tomography (PET) neuroimaging with the Pittsburgh Compound_B (PiB) is widely used to assess amyloid plaque burden. Standard quantification approaches normalize PiB-PET by mean cerebellar gray matter uptake. Previous studies suggested similar pons and white-matter uptake in Alzheimer's disease (AD) and healthy controls (HC), but lack exhaustive comparison of normalization across the three regions, with data-driven diagnostic classification. We aimed to compare the impact of distinct reference regions in normalization, measured by data-driven statistical analysis, and correlation with cerebrospinal fluid (CSF) amyloid β (Aβ) species concentrations. 243 individuals with clinical diagnosis of AD, HC, mild cognitive impairment (MCI) and other dementias, from the Biomarkers for Alzheimer's/Parkinson's Disease (BIOMARKAPD) initiative were included. PiB-PET images and CSF concentrations of Aβ38, Aβ40 and Aβ42 were submitted to classification using support vector machines. Voxel-wise group differences and correlations between normalized PiB-PET images and CSF Aβ concentrations were calculated. Normalization by cerebellar gray matter and pons yielded identical classification accuracy of AD (accuracy-96%, sensitivity-96%, specificity-95%), and significantly higher than Aβ concentrations (best accuracy 91%). Normalization by the white-matter showed decreased extent of statistically significant multivoxel patterns and was the only method not outperforming CSF biomarkers, suggesting statistical inferiority. Aβ38 and Aβ40 correlated negatively with PiB-PET images normalized by the white-matter, corroborating previous observations of correlations with non-AD-specific subcortical changes in white-matter. In general, when using the pons as reference region, higher voxel-wise group differences and stronger correlation with Aβ42, the Aβ42/Aβ40 or Aβ42/Aβ38 ratios were found compared to normalization based on cerebellar gray matter. Direct multivariate comparison of distinct reference regions in normalization of PET amyloid markers Using the pons as ROI, higher voxel-wise group differences emerge Using the pons as ROIs stronger correlation with Aβ42, the Aβ42/Aβ40 or Aβ42/Aβ38 ratios were found. Evidence for statistical inferiority of CSF biomarkers Aβ38 and Aβ40 correlated negatively with PiB-PET white-matter normalized images.
Collapse
|
126
|
Chen Z, Jamadar SD, Li S, Sforazzini F, Baran J, Ferris N, Shah NJ, Egan GF. From simultaneous to synergistic MR-PET brain imaging: A review of hybrid MR-PET imaging methodologies. Hum Brain Mapp 2018; 39:5126-5144. [PMID: 30076750 DOI: 10.1002/hbm.24314] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022] Open
Abstract
Simultaneous Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) scanning is a recent major development in biomedical imaging. The full integration of the PET detector ring and electronics within the MR system has been a technologically challenging design to develop but provides capacity for simultaneous imaging and the potential for new diagnostic and research capability. This article reviews state-of-the-art MR-PET hardware and software, and discusses future developments focusing on neuroimaging methodologies for MR-PET scanning. We particularly focus on the methodologies that lead to an improved synergy between MRI and PET, including optimal data acquisition, PET attenuation and motion correction, and joint image reconstruction and processing methods based on the underlying complementary and mutual information. We further review the current and potential future applications of simultaneous MR-PET in both systems neuroscience and clinical neuroimaging research. We demonstrate a simultaneous data acquisition protocol to highlight new applications of MR-PET neuroimaging research studies.
Collapse
Affiliation(s)
- Zhaolin Chen
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | - Sharna D Jamadar
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| | - Shenpeng Li
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | | | - Jakub Baran
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszów, Rzeszów, Poland
| | - Nicholas Ferris
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Imaging, Monash Health, Clayton, Victoria, Australia
| | - Nadim Jon Shah
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum, Jülich, Germany
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
127
|
Pascoal TA, Shin M, Kang MS, Chamoun M, Chartrand D, Mathotaarachchi S, Bennacef I, Therriault J, Ng KP, Hopewell R, Bouhachi R, Hsiao HH, Benedet AL, Soucy JP, Massarweh G, Gauthier S, Rosa-Neto P. In vivo quantification of neurofibrillary tangles with [ 18F]MK-6240. ALZHEIMERS RESEARCH & THERAPY 2018; 10:74. [PMID: 30064520 PMCID: PMC6069775 DOI: 10.1186/s13195-018-0402-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/06/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Imaging agents capable of quantifying the brain's tau aggregates will allow a more precise staging of Alzheimer's disease (AD). The aim of the present study was to examine the in vitro properties as well as the in vivo kinetics, using gold standard methods, of the novel positron emission tomography (PET) tau imaging agent [18F]MK-6240. METHODS In vitro properties of [18F]MK-6240 were estimated with autoradiography in postmortem brain tissues of 14 subjects (seven AD patients and seven age-matched controls). In vivo quantification of [18F]MK-6240 binding was performed in 16 subjects (four AD patients, three mild cognitive impairment patients, six healthy elderly individuals, and three healthy young individuals) who underwent 180-min dynamic scans; six subjects had arterial sampling for metabolite correction. Simplified approaches for [18F]MK-6240 quantification were validated using full kinetic modeling with metabolite-corrected arterial input function. All participants also underwent amyloid-PET and structural magnetic resonance imaging. RESULTS In vitro [18F]MK-6240 uptake was higher in AD patients than in age-matched controls in brain regions expected to contain tangles such as the hippocampus, whereas no difference was found in the cerebellar gray matter. In vivo, [18F]MK-6240 displayed favorable kinetics with rapid brain delivery and washout. The cerebellar gray matter had low binding across individuals, showing potential for use as a reference region. A reversible two-tissue compartment model well described the time-activity curves across individuals and brain regions. Distribution volume ratios using the plasma input and standardized uptake value ratios (SUVRs) calculated after the binding approached equilibrium (90 min) were correlated and higher in mild cognitive impairment or AD dementia patients than in controls. Reliability analysis revealed robust SUVRs calculated from 90 to 110 min, while earlier time points provided inaccurate estimates. CONCLUSIONS This evaluation shows an [18F]MK-6240 distribution in concordance with postmortem studies and that simplified quantitative approaches such as the SUVR offer valid estimates of neurofibrillary tangle load 90 min post injection. [18F]MK-6240 is a promising tau tracer with the potential to be applied in the disease diagnosis and assessment of therapeutic interventions.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Mira Chamoun
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Daniel Chartrand
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Kok Pin Ng
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Robert Hopewell
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Reda Bouhachi
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Hung-Hsin Hsiao
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada. .,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada. .,Douglas Hospital, McGill University, 6875 La Salle Blvd-FBC room 3149, Montreal, QC, H4H 1R3, Canada.
| |
Collapse
|
128
|
Son HJ, Jeong YJ, Yoon HJ, Lee SY, Choi GE, Park JA, Kim MH, Lee KC, Lee YJ, Kim MK, Cho K, Kang DY. Assessment of brain beta-amyloid deposition in transgenic mouse models of Alzheimer's disease with PET imaging agents 18F-flutemetamol and 18F-florbetaben. BMC Neurosci 2018; 19:45. [PMID: 30053803 PMCID: PMC6063010 DOI: 10.1186/s12868-018-0447-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although amyloid beta (Aβ) imaging is widely used for diagnosing and monitoring Alzheimer's disease in clinical fields, paralleling comparison between 18F-flutemetamol and 18F-florbetaben was rarely attempted in AD mouse model. We performed a comparison of Aβ PET images between 18F-flutemetamol and 18F-florbetaben in a recently developed APPswe mouse model, C57BL/6-Tg (NSE-hAPPsw) Korl. RESULTS After an injection (0.23 mCi) of 18F-flutemetamol and 18F-florbetaben at a time interval of 2-3 days, we compared group difference of SUVR and kinetic parameters between the AD (n = 7) and control (n = 7) mice, as well as between 18F-flutemetamol and 18F-florbetaben image. In addition, bio-distribution and histopathology were conducted. With visual image and VOI-based SUVR analysis, the AD group presented more prominent uptake than did the control group in both the 18F-florbetaben and 18F-flutemetamol images. With kinetic analysis, the 18F-florbetaben images showed differences in K1 and k4 between the AD and control groups, although 18F-flutemetamol images did not show significant difference. 18F-florbetaben images showed more prominent cortical uptake and matched well to the thioflavin S staining images than did the 18F-flutemetamol image. In contrast, 18F-flutemetamol images presented higher K1, k4, K1/k2 values than those of 18F-florbetaben images. Also, 18F-flutemetamol images presented prominent uptake in the bowel and bladder, consistent with higher bio-distribution in kidney, lung, blood and heart. CONCLUSIONS Compared with 18F-flutemetamol images, 18F-florbetaben images showed prominent visual uptake intensity, SUVR, and higher correlations with the pathology. In contrast, 18F-flutemetamol was more actively metabolized than was 18F-florbetaben (Son et al. in J Nucl Med 58(Suppl 1):S278, 2017].
Collapse
Affiliation(s)
- Hye Joo Son
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Young Jin Jeong
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Hyun Jin Yoon
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Sang Yoon Lee
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Go-Eun Choi
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Ji-Ae Park
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Min Hwan Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yong Jin Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Mun Ki Kim
- Pohang Center of Evolution of Biomaterials, Pohang Technopark, Pohang, Korea
| | - Kook Cho
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Do-Young Kang
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| |
Collapse
|
129
|
Wilckens KA, Tudorascu DL, Snitz BE, Price JC, Aizenstein HJ, Lopez OL, Erickson KI, Lopresti BJ, Laymon CM, Minhas D, Mathis CA, Buysse DJ, Klunk WE, Cohen AD. Sleep moderates the relationship between amyloid beta and memory recall. Neurobiol Aging 2018; 71:142-148. [PMID: 30138767 DOI: 10.1016/j.neurobiolaging.2018.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/18/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022]
Abstract
Amyloid-β (Aβ) accumulation is a hallmark of Alzheimer's disease, although Aβ alone may be insufficient to cause impairments. Modifiable health factors, including sleep, may mitigate functional symptoms of neurodegeneration. We assessed whether sleep moderated the relationship between Aβ and cognitive performance in 41 older adults, mean age 83 years. Sleep measures included actigraphy-assessed wake after sleep onset and total sleep time. Cognitive performance was assessed with memory recall, cognitive flexibility, and verbal fluency. Memory recall was assessed with the Rey-Osterrieth Complex Figure task, cognitive flexibility with the Trail Making test, and verbal fluency with FAS word generation. Aβ was assessed with a global measure of Pittsburgh Compound B. Wake after sleep onset moderated the relationship between Aβ and memory, with a stronger positive association for Aβ and forgetting in those with poorer sleep. These results suggest a possible protective role of sleep in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Kristine A Wilckens
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Dana L Tudorascu
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie C Price
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Davneet Minhas
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daniel J Buysse
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
130
|
Zeydan B, Lowe VJ, Schwarz CG, Przybelski SA, Tosakulwong N, Zuk SM, Senjem ML, Gunter JL, Roberts RO, Mielke MM, Benarroch EE, Rodriguez M, Machulda MM, Lesnick TG, Knopman DS, Petersen RC, Jack CR, Kantarci K, Kantarci OH. Pittsburgh compound-B PET white matter imaging and cognitive function in late multiple sclerosis. Mult Scler 2018; 24:739-749. [PMID: 28474977 PMCID: PMC5665724 DOI: 10.1177/1352458517707346] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND There is growing interest in white matter (WM) imaging with positron emission tomography (PET). OBJECTIVES We studied the association of cognitive function in late multiple sclerosis (MS) with cortical and WM Pittsburgh compound-B PET (PiB-PET) binding. METHODS In the population-based Mayo Clinic Study of Aging, 24 of 4869 participants had MS (12 underwent PiB-PET). Controls were age and sex matched (5:1). We used automated or semi-automated processing for quantitative image analyses and conditional logistic regression for group differences. RESULTS MS patients had lower memory ( p = 0.03) and language ( p = 0.02) performance; smaller thalamic volumes ( p = 0.003); and thinner temporal ( p = 0.001) and frontal ( p = 0.045) cortices on magnetic resonance imaging (MRI) than controls. There was no difference in global cortical PiB standardized uptake value ratios between MS and controls ( p = 0.35). PiB uptake was lower in areas of WM hyperintensities compared to normal-appearing white matter (NAWM) in MS ( p = 0.0002). Reduced PiB uptake in both the areas of WM hyperintensities ( r = 0.65; p = 0.02) and NAWM ( r = 0.69; p = 0.01) was associated with decreased visuospatial performance in MS. CONCLUSION PiB uptake in the cortex in late MS is not different from normal age-matched controls. PiB uptake in the WM in late MS may be a marker of the large network structures' integrity such as those involved in visuospatial performance.
Collapse
Affiliation(s)
- Burcu Zeydan
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Val J. Lowe
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Christopher G. Schwarz
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Scott A. Przybelski
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Rochester, Minnesota, United States of America
| | - Nirubol Tosakulwong
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Rochester, Minnesota, United States of America
| | - Samantha M. Zuk
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Matthew L. Senjem
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
- Mayo Clinic College of Medicine, Department of Information Technology, Rochester, Minnesota, United States of America
| | - Jeffrey L. Gunter
- Mayo Clinic College of Medicine, Department of Information Technology, Rochester, Minnesota, United States of America
| | - Rosebud O. Roberts
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Rochester, Minnesota, United States of America
| | - Michelle M. Mielke
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Rochester, Minnesota, United States of America
| | - Eduardo E. Benarroch
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
| | - Moses Rodriguez
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
| | - Mary M. Machulda
- Mayo Clinic College of Medicine, Department of Psychiatry and Psychology, Rochester, Minnesota, United States of America
| | - Timothy G. Lesnick
- Mayo Clinic College of Medicine, Department of Health Sciences Research, Rochester, Minnesota, United States of America
| | - David S. Knopman
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
| | - Ronald C. Petersen
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
| | - Clifford R. Jack
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Kejal Kantarci
- Mayo Clinic College of Medicine, Department of Radiology, Rochester, Minnesota, United States of America
| | - Orhun H. Kantarci
- Mayo Clinic College of Medicine, Department of Neurology, Rochester, Minnesota, United States of America
| |
Collapse
|
131
|
Lopez OL, Becker JT, Chang Y, Klunk WE, Mathis C, Price J, Aizenstein HJ, Snitz B, Cohen AD, DeKosky ST, Ikonomovic M, Kamboh MI, Kuller LH. Amyloid deposition and brain structure as long-term predictors of MCI, dementia, and mortality. Neurology 2018; 90:e1920-e1928. [PMID: 29695596 DOI: 10.1212/wnl.0000000000005549] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 03/14/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES To test the hypothesis that brain structural integrity (i.e., hippocampal [HIP] volume), white matter lesions (WMLs), and β-amyloid deposition are associated with long-term increased risk of incident dementia and mortality in 183 cognitively normal individuals and patients with mild cognitive impairment (MCI) aged 80 years and older. METHODS All participants had a brain structural MRI scan and PET scan with 11C-labeled Pittsburgh compound B in 2009 and were reexamined yearly through 2015 (mean follow-up time 5.2 ± 1.3 years). RESULTS In the last evaluation through 2010-2015, 56 (31%) participants were cognitively normal, 67 (37%) had MCI, and 60 (33%) had dementia. Fifty-seven (31%) died during follow-up, and 20 (35%) developed dementia before their death. All 3 biomarkers were independent predictors of incident dementia in all participants. After adjusting for the risk of dying, amyloid deposition and WMLs remained strong predictors. Of the 60 participants with incident dementia, 54 (90%) had at least one imaging abnormality. Participants with no biomarker positivity had a very low risk of dementia (16%), while 75% of the participants with the 3 biomarkers progressed to dementia. HIP volume and β-amyloid deposition were associated with death only in participants with MCI. CONCLUSIONS This study showed the presence of more than one biomarker was a stronger long-term predictor of incident dementia than any biomarker alone. After adjusting for the risk of dying, amyloid deposition and WMLs were stronger predictors of dementia than HIP volume. The risk of dying during follow-up was associated with both neurodegeneration and amyloid deposition, especially in individuals with MCI.
Collapse
Affiliation(s)
- Oscar L Lopez
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville.
| | - James T Becker
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - YueFang Chang
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - William E Klunk
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Chester Mathis
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Julia Price
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Howard J Aizenstein
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Beth Snitz
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Ann D Cohen
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Steven T DeKosky
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Milos Ikonomovic
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - M Ilyas Kamboh
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| | - Lewis H Kuller
- From the Departments of Neurology (O.L.L., J.T.B., B.S., M.I.), Psychiatry (O.L.L., J.T.B., W.E.K., H.J.A., A.D.C.), Psychology (J.T.B.), Neurosurgery (Y.C.), Epidemiology, Graduate School of Public Health (Y.C., L.H.K.), Radiology (C.M., J.P.), Pharmaceutical Sciences (C.M.), and Genetics (M.I.K.), University of Pittsburgh, School of Medicine, PA; and Department of Neurology (S.T.D.), University of Florida, Gainesville
| |
Collapse
|
132
|
Nobili F, Cagnin A, Calcagni ML, Chincarini A, Guerra UP, Morbelli S, Padovani A, Paghera B, Pappatà S, Parnetti L, Sestini S, Schillaci O. Emerging topics and practical aspects for an appropriate use of amyloid PET in the current Italian context. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 63:83-92. [PMID: 29697220 DOI: 10.23736/s1824-4785.18.03069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In May 2017 some representatives of the Italian nuclear medicine and neurological communities spontaneously met to discuss the issues emerged during the first two years of routine application of amyloid PET with fluorinated radiopharmaceuticals in the real world. The limitations of a binary classification of scans, the possibility to obtain early images as a surrogate marker of regional cerebral bloos flow, the need for (semi-)quantification and, thus, the opportunity of ranking brain amyloidosis, the correlation with Aβ42 levels in the cerebrospinal fluid, the occurrence and biological meaning of uncertain/boderline scans, the issue of incidental amyloidosis, the technical pittfalls leading to false negative/positive results, the position of the tool in the diagnostic flow-chart in the national reality, are the main topics that have been discussed. Also, a card to justify the examination to be filled by the dementia specialist and a card for the nuclear medicine physician to report the exam in detail have been approved and are available in the web, which should facilitate the creation of a national register, as previewed by the 2015 intersocietal recommendation on the use of amyloid PET in Italy. The content of this discussion could stimulate both public institutions and companies to support further research on these topics.
Collapse
Affiliation(s)
- Flavio Nobili
- Department of Neuroscience (DINOGMI), University of Genoa and Neurology Clinic, San Martino Polyclinic Hospital, Genoa, Italy -
| | - Annachiara Cagnin
- Department of Neurosciences (DNS), University of Padua, Padua, Italy.,San Camillo IRCCS Hospital, Venice, Italy
| | - Maria L Calcagni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Chincarini
- National Institute for Nuclear Physics (INFN), Genoa Section, Genoa, Italy
| | - Ugo P Guerra
- Unit of Nuclear Medicine, Poliambulanza Fundation, Brescia, Italy
| | - Silvia Morbelli
- Unit of Nuclear Medicine, Department of Health Sciences (DISSAL), Polyclinic San Martino Hospital, University of Genoa, Genoa, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Neurology Clinic, Spedali Civili, Brescia, Italy
| | - Barbara Paghera
- Unit of Nuclear Medicine, ASST-Spedali Civili, University of Brescia, Brescia, Italy
| | - Sabina Pappatà
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Lucilla Parnetti
- Center for Memory Disorders, Laboratory of Clinical Neurochemistry, Neurology Clinic, University of Perugia, Perugia, Italy
| | - Stelvio Sestini
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, N.O.P. - S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.,IRCCS Neuromed, Rome, Italy
| |
Collapse
|
133
|
Niemantsverdriet E, Ottoy J, Somers C, De Roeck E, Struyfs H, Soetewey F, Verhaeghe J, Van den Bossche T, Van Mossevelde S, Goeman J, De Deyn PP, Mariën P, Versijpt J, Sleegers K, Van Broeckhoven C, Wyffels L, Albert A, Ceyssens S, Stroobants S, Staelens S, Bjerke M, Engelborghs S. The Cerebrospinal Fluid Aβ1-42/Aβ1-40 Ratio Improves Concordance with Amyloid-PET for Diagnosing Alzheimer's Disease in a Clinical Setting. J Alzheimers Dis 2018; 60:561-576. [PMID: 28869470 PMCID: PMC5611891 DOI: 10.3233/jad-170327] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Evidence suggests that the concordance between amyloid-PET and cerebrospinal fluid (CSF) amyloid-β (Aβ) increases when the CSF Aβ1–42/Aβ1–40 ratio is used as compared to CSF Aβ1–42 levels alone. Objective: In order to test this hypothesis, we set up a prospective longitudinal study comparing the concordance between different amyloid biomarkers for Alzheimer’s disease (AD) in a clinical setting. Methods: Seventy-eight subjects (AD dementia (n = 17), mild cognitive impairment (MCI, n = 48), and cognitively healthy controls (n = 13)) underwent a [18F]Florbetapir ([18F]AV45) PET scan, [18F]FDG PET scan, MRI scan, and an extensive neuropsychological examination. In a large subset (n = 67), a lumbar puncture was performed and AD biomarkers were analyzed (Aβ1–42, Aβ1–40, T-tau, P-tau181). Results: We detected an increased concordance in the visual and quantitative (standardized uptake value ratio (SUVR) and total volume of distribution (VT)) [18F]AV45 PET measures when the CSF Aβ1–42/Aβ1–40 was applied compared to Aβ1–42 alone. CSF biomarkers were stronger associated to [18F]AV45 PET for SUVR values when considering the total brain white matter as reference region instead of cerebellar grey matter Conclusions: The concordance between CSF Aβ and [18F]AV45 PET increases when the CSF Aβ1–42/Aβ1–40 ratio is applied. This finding is of most importance for the biomarker-based diagnosis of AD as well as for selection of subjects for clinical trials with potential disease-modifying therapies for AD.
Collapse
Affiliation(s)
- Ellis Niemantsverdriet
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Julie Ottoy
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Charisse Somers
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Ellen De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Faculty of Psychology and Educational Sciences, Vrije Universiteit Brussels, Brussels, Belgium
| | - Hanne Struyfs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Femke Soetewey
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Tobi Van den Bossche
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Sara Van Mossevelde
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Johan Goeman
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Peter Paul De Deyn
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Peter Mariën
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.,Clinical and Experimental Neurolinguistics, CLIN, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jan Versijpt
- Vrije Universiteit Brussel(VUB), University Hospital Brussels (UZ Brussel), Department of Neurology, Brussels, Belgium
| | - Kristel Sleegers
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium.,Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Adrien Albert
- Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sarah Ceyssens
- Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium.,Departmentof Nuclear Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), UAntwerp, Antwerp, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| |
Collapse
|
134
|
Minhas DS, Price JC, Laymon CM, Becker CR, Klunk WE, Tudorascu DL, Abrahamson EE, Hamilton RL, Kofler JK, Mathis CA, Lopez OL, Ikonomovic MD. Impact of partial volume correction on the regional correspondence between in vivo [C-11]PiB PET and postmortem measures of Aβ load. NEUROIMAGE-CLINICAL 2018; 19:182-189. [PMID: 30023168 PMCID: PMC6050460 DOI: 10.1016/j.nicl.2018.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/02/2018] [Accepted: 04/01/2018] [Indexed: 11/19/2022]
Abstract
The positron emission tomography (PET) radiotracer Pittsburgh Compound B ([C-11]PiB) demonstrates a high affinity for fibrillary amyloid-beta (Aβ) aggregates. However, [C-11]PiB's in vivo sensitivity and specificity is an ongoing area of investigation in correlation studies with postmortem measures of Aβ pathology. One potential confound in PET-to-postmortem correlation studies is the limited spatial resolution of PET and resulting partial volume effects (PVEs). In this work, we evaluated the impact of three partial volume correction (PVC) techniques – the Meltzer, the modified Müller-Gärtner, and the Region-Based Voxel-Wise – on correlations between region-matched in vivo [C-11]PiB standardized uptake value ratios (SUVRs) and postmortem measures of Aβ pathology in a unique cohort of nine subjects. Postmortem Aβ pathology was assessed histologically as percent area coverage of 6-CN-PiB positive and Aβ immunoreactive (4G8 antibody) deposits. The application of all three PVC techniques resulted in minimally reduced PET-to-postmortem correlations relative to no PVC. However, correlations to both 6-CN-PiB and 4G8 percent area across all PVC techniques and no PVC were statistically significant at p < 0.01, suggesting that PVC is of minimal importance in understanding the relationship between Aβ PET and neuropathologically assessed Aβ. Thus, the utility of PVC in Aβ PET imaging should continue to be examined on an application-specific basis. Three partial volume correction techniques are applied to PiB PET SUVR data. Corrected and uncorrected data are correlated to postmortem measures of Aβ load. All 3 correction methods minimally impact PET-to-postmortem correlations.
Collapse
Affiliation(s)
- Davneet S Minhas
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Julie C Price
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carl R Becker
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neuropathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric E Abrahamson
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Ronald L Hamilton
- Department of Neuropathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julia K Kofler
- Department of Neuropathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
135
|
Sharma RA, Varga AW, Bubu OM, Pirraglia E, Kam K, Parekh A, Wohlleber M, Miller MD, Andrade A, Lewis C, Tweardy S, Buj M, Yau PL, Sadda R, Mosconi L, Li Y, Butler T, Glodzik L, Fieremans E, Babb JS, Blennow K, Zetterberg H, Lu SE, Badia SG, Romero S, Rosenzweig I, Gosselin N, Jean-Louis G, Rapoport DM, de Leon MJ, Ayappa I, Osorio RS. Obstructive Sleep Apnea Severity Affects Amyloid Burden in Cognitively Normal Elderly. A Longitudinal Study. Am J Respir Crit Care Med 2018; 197:933-943. [PMID: 29125327 PMCID: PMC6020410 DOI: 10.1164/rccm.201704-0704oc] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Recent evidence suggests that obstructive sleep apnea (OSA) may be a risk factor for developing mild cognitive impairment and Alzheimer's disease. However, how sleep apnea affects longitudinal risk for Alzheimer's disease is less well understood. OBJECTIVES To test the hypothesis that there is an association between severity of OSA and longitudinal increase in amyloid burden in cognitively normal elderly. METHODS Data were derived from a 2-year prospective longitudinal study that sampled community-dwelling healthy cognitively normal elderly. Subjects were healthy volunteers between the ages of 55 and 90, were nondepressed, and had a consensus clinical diagnosis of cognitively normal. Cerebrospinal fluid amyloid β was measured using ELISA. Subjects received Pittsburgh compound B positron emission tomography scans following standardized procedures. Monitoring of OSA was completed using a home sleep recording device. MEASUREMENTS AND MAIN RESULTS We found that severity of OSA indices (AHIall [F1,88 = 4.26; P < 0.05] and AHI4% [F1,87 = 4.36; P < 0.05]) were associated with annual rate of change of cerebrospinal fluid amyloid β42 using linear regression after adjusting for age, sex, body mass index, and apolipoprotein E4 status. AHIall and AHI4% were not associated with increases in ADPiB-mask (Alzheimer's disease vulnerable regions of interest Pittsburg compound B positron emission tomography mask) most likely because of the small sample size, although there was a trend for AHIall (F1,28 = 2.96, P = 0.09; and F1,28 = 2.32, not significant, respectively). CONCLUSIONS In a sample of cognitively normal elderly, OSA was associated with markers of increased amyloid burden over the 2-year follow-up. Sleep fragmentation and/or intermittent hypoxia from OSA are likely candidate mechanisms. If confirmed, clinical interventions for OSA may be useful in preventing amyloid build-up in cognitively normal elderly.
Collapse
Affiliation(s)
- Ram A. Sharma
- Center for Brain Health, Department of Psychiatry, and
| | - Andrew W. Varga
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Omonigho M. Bubu
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, Florida
| | | | - Korey Kam
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ankit Parekh
- College of Engineering, The University of Iowa, Iowa City, Iowa
| | | | | | | | - Clifton Lewis
- Center for Brain Health, Department of Psychiatry, and
| | | | - Maja Buj
- Center for Brain Health, Department of Psychiatry, and
| | - Po L. Yau
- Center for Brain Health, Department of Psychiatry, and
| | - Reem Sadda
- Harlem Hospital–Columbia University Medical Center, New York, New York
| | - Lisa Mosconi
- Center for Brain Health, Department of Psychiatry, and
| | - Yi Li
- Center for Brain Health, Department of Psychiatry, and
| | - Tracy Butler
- Center for Brain Health, Department of Psychiatry, and
| | - Lidia Glodzik
- Center for Brain Health, Department of Psychiatry, and
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York
| | - James S. Babb
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York
| | - Kaj Blennow
- Institute of Neuroscience and Psychiatry, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Psychiatry, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Shou E. Lu
- Department of Biostatistics, Rutgers School of Public Health, Piscataway, New Jersey
| | - Sandra G. Badia
- Sleep Unit, Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institute for Biomedical Research Sant Pau, CIBERSAM, Barcelona, Spain
- Department of Clinical Psychology and Psychobiology, University of Barcelona, Barcelona, Spain
| | - Sergio Romero
- Biomedical Engineering Research Centre, Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St. Thomas’ Hospital, GSTT NHS Trust, London, United Kingdom
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hospital du Sacre-Coeur de Montreal, Montreal, Quebec, Canada; and
- Department of Psychology, Universite de Montreal, Montreal, Quebec, Canada
| | - Girardin Jean-Louis
- Center for Healthful Behavior Change, Division of Health and Behavior, Department of Population Health, New York University Langone Medical Center, New York, New York
| | - David M. Rapoport
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Indu Ayappa
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
136
|
Klunk WE. Molecular imaging: What is right and what is an illusion? ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2018; 10:217-220. [PMID: 29780866 PMCID: PMC5956935 DOI: 10.1016/j.dadm.2018.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Over the past 40 years, brain molecular imaging has evolved from measuring cerebral metabolism with fluorodeoxyglucose, to neuroreceptor imaging, to imaging pathological protein deposits. In the early going, the characteristics of successful molecular imaging radiotracers were defined, and a detailed "Process" was developed for the collection of basic pharmacodynamic and pharmacokinetic data. These data are essential for the interpretation of in vivo imaging data and for defining the strengths, weaknesses, and limitations of new tracers. This perspective discusses the use of this "Process" in the development of the amyloid β positron emission tomography radiotracer, Pittsburgh Compound-B, and discusses some of the current controversies and difficulties in the field of tau positron emission tomography in the context of human data that preceded completion of this radiotracer characterization process-which still remains to be completed. As a field, we must decide which data are valid and which are artifacts and determine that when the artifacts are so overwhelming, the data are merely an illusion.
Collapse
|
137
|
Higashi T, Nishii R, Kagawa S, Kishibe Y, Takahashi M, Okina T, Suzuki N, Hasegawa H, Nagahama Y, Ishizu K, Oishi N, Kimura H, Watanabe H, Ono M, Saji H, Yamauchi H. 18F-FPYBF-2, a new F-18-labelled amyloid imaging PET tracer: first experience in 61 volunteers and 55 patients with dementia. Ann Nucl Med 2018; 32:206-216. [PMID: 29388083 PMCID: PMC5852179 DOI: 10.1007/s12149-018-1236-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 11/28/2022]
Abstract
Objective Recently, we developed a benzofuran derivative for the imaging of β-amyloid plaques, 5-(5-(2-(2-(2-18F-fluoroethoxy)ethoxy)ethoxy)benzofuran-2-yl)-N-methylpyridin-2-amine (18F-FPYBF-2) (Ono et al., J Med Chem 54:2971–9, 2011). The aim of this study was to assess the feasibility of 18F-FPYBF-2 as an amyloid imaging PET tracer in a first clinical study with healthy volunteers and patients with various dementia and in comparative dual tracer study using 11C-Pittsburgh Compound B (11C-PiB). Methods 61 healthy volunteers (age: 53.7 ± 13.1 years old; 19 male and 42 female; age range 24–79) and 55 patients with suspected dementia [Alzheimer’s Disease (AD); early AD: n = 19 and moderate stage AD: n = 8, other dementia: n = 9, mild cognitive impairment (MCI): n = 16, cognitively normal: n = 3] for first clinical study underwent static head PET/CT scan using 18F−FPYBF-2 at 50–70 min after injection. 13 volunteers and 14 patients also underwent dynamic PET scan at 0–50 min at the same instant. 16 subjects (volunteers: n = 5, patients with dementia: n = 11) (age: 66.3 ± 14.2 years old; 10 males and 6 females) were evaluated for comparative study (50–70 min after injection) using 18F-FPYBF-2 and 11C-PiB on separate days, respectively. Quantitative analysis of mean cortical uptake was calculated using Mean Cortical Index of SUVR (standardized uptake value ratio) based on the established method for 11C-PiB analysis using cerebellar cortex as control. Results Studies with healthy volunteers showed that 18F-FPYBF-2 uptake was mainly observed in cerebral white matter and that average Mean Cortical Index at 50–70 min was low and stable (1.066 ± 0.069) basically independent from age or gender. In patients with AD, 18F-FPYBF-2 uptake was observed both in cerebral white and gray matter, and Mean Cortical Index was significantly higher (early AD: 1.288 ± 0.134, moderate AD: 1.342 ± 0.191) than those of volunteers and other dementia (1.018 ± 0.057). In comparative study, the results of 18F-FPYBF-2 PET/CT were comparable with those of 11C-PiB, and the Mean Cortical Index (18F-FPYBF-2: 1.173 ± 0.215; 11C-PiB: 1.435 ± 0.474) showed direct proportional relationship with each other (p < 0.0001). Conclusions Our first clinical study suggest that 18F-FPYBF-2 is a useful PET tracer for the evaluation of β-amyloid deposition and that quantitative analysis of Mean Cortical Index of SUVR is a reliable diagnostic tool for the diagnosis of AD. Electronic supplementary material The online version of this article (10.1007/s12149-018-1236-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatsuya Higashi
- Shiga Medical Center Research Institute, Moriyama, Japan. .,Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan.
| | - Ryuichi Nishii
- Shiga Medical Center Research Institute, Moriyama, Japan.,Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Shinya Kagawa
- Shiga Medical Center Research Institute, Moriyama, Japan
| | | | | | - Tomoko Okina
- Department of Geriatric Medicine, Shiga General Hospital, Moriyama, Japan
| | - Norio Suzuki
- Department of Geriatric Medicine, Shiga General Hospital, Moriyama, Japan
| | - Hiroshi Hasegawa
- Department of Geriatric Medicine, Shiga General Hospital, Moriyama, Japan
| | | | - Koichi Ishizu
- Shiga Medical Center Research Institute, Moriyama, Japan.,Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- Research and Educational Unit of Leaders for Integrated Medical System, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hideo Saji
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
138
|
Coskuner-Weber O, Uversky VN. Insights into the Molecular Mechanisms of Alzheimer's and Parkinson's Diseases with Molecular Simulations: Understanding the Roles of Artificial and Pathological Missense Mutations in Intrinsically Disordered Proteins Related to Pathology. Int J Mol Sci 2018; 19:E336. [PMID: 29364151 PMCID: PMC5855558 DOI: 10.3390/ijms19020336] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/18/2022] Open
Abstract
Amyloid-β and α-synuclein are intrinsically disordered proteins (IDPs), which are at the center of Alzheimer's and Parkinson's disease pathologies, respectively. These IDPs are extremely flexible and do not adopt stable structures. Furthermore, both amyloid-β and α-synuclein can form toxic oligomers, amyloid fibrils and other type of aggregates in Alzheimer's and Parkinson's diseases. Experimentalists face challenges in investigating the structures and thermodynamic properties of these IDPs in their monomeric and oligomeric forms due to the rapid conformational changes, fast aggregation processes and strong solvent effects. Classical molecular dynamics simulations complement experiments and provide structural information at the atomic level with dynamics without facing the same experimental limitations. Artificial missense mutations are employed experimentally and computationally for providing insights into the structure-function relationships of amyloid-β and α-synuclein in relation to the pathologies of Alzheimer's and Parkinson's diseases. Furthermore, there are several natural genetic variations that play a role in the pathogenesis of familial cases of Alzheimer's and Parkinson's diseases, which are related to specific genetic defects inherited in dominant or recessive patterns. The present review summarizes the current understanding of monomeric and oligomeric forms of amyloid-β and α-synuclein, as well as the impacts of artificial and pathological missense mutations on the structural ensembles of these IDPs using molecular dynamics simulations. We also emphasize the recent investigations on residual secondary structure formation in dynamic conformational ensembles of amyloid-β and α-synuclein, such as β-structure linked to the oligomerization and fibrillation mechanisms related to the pathologies of Alzheimer's and Parkinson's diseases. This information represents an important foundation for the successful and efficient drug design studies.
Collapse
Affiliation(s)
- Orkid Coskuner-Weber
- Türkisch-Deutsche Universität, Theoretical and Computational Biophysics Group, Molecular Biotechnology, Sahinkaya Caddesi, No. 86, Beykoz, Istanbul 34820, Turkey.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| |
Collapse
|
139
|
Shokouhi S, Campbell D, Brill AB, Gwirtsman HE. Longitudinal Positron Emission Tomography in Preventive Alzheimer's Disease Drug Trials, Critical Barriers from Imaging Science Perspective. Brain Pathol 2018; 26:664-71. [PMID: 27327527 PMCID: PMC5958602 DOI: 10.1111/bpa.12399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 06/16/2016] [Indexed: 12/30/2022] Open
Abstract
Recent Alzheimer's trials have recruited cognitively normal people at risk for Alzheimer's dementia. Due to the lack of clinical symptoms in normal population, conventional clinical outcome measures are not suitable for these early trials. While several groups are developing new composite cognitive tests that could serve as potential outcome measures by detecting subtle cognitive changes in normal people, there is a need for longitudinal brain imaging techniques that can correlate with temporal changes in these new tests and provide additional objective measures of neuropathological changes in brain. Positron emission tomography (PET) is a nuclear medicine imaging procedure based on the measurement of annihilation photons after positron emission from radiolabeled molecules that allow tracking of biological processes in body, including the brain. PET is a well-established in vivo imaging modality in Alzheimer's disease diagnosis and research due to its capability of detecting abnormalities in three major hallmarks of this disease. These include (1) amyloid beta plaques; (2) neurofibrillary tau tangles and (3) decrease in neuronal activity due to loss of nerve cell connection and death. While semiquantitative PET imaging techniques are commonly used to set discrete cut-points to stratify abnormal levels of amyloid accumulation and neurodegeneration, they are suboptimal for detecting subtle longitudinal changes. In this study, we have identified and discussed four critical barriers in conventional longitudinal PET imaging that may be particularly relevant for early Alzheimer's disease studies. These include within and across subject heterogeneity of AD-affected brain regions, PET intensity normalization, neuronal compensations in early disease stages and cerebrovascular amyloid deposition.
Collapse
Affiliation(s)
- Sepideh Shokouhi
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center
| | - Desmond Campbell
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center
| | - Aaron B Brill
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center
| | | | | |
Collapse
|
140
|
Lao PJ, Handen BL, Betthauser TJ, Mihaila I, Hartley SL, Cohen AD, Tudorascu DL, Bulova PD, Lopresti BJ, Tumuluru RV, Murali D, Mathis CA, Barnhart TE, Stone CK, Price JC, Devenny DA, Johnson SC, Klunk WE, Christian BT. Alzheimer-Like Pattern of Hypometabolism Emerges with Elevated Amyloid-β Burden in Down Syndrome. J Alzheimers Dis 2018; 61:631-644. [PMID: 29254096 PMCID: PMC5994924 DOI: 10.3233/jad-170720] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The Down syndrome (DS) population is genetically predisposed to amyloid-β protein precursor overproduction and Alzheimer's disease (AD). OBJECTIVE The temporal ordering and spatial association between amyloid-β, glucose metabolism, and gray matter (GM) volume in the DS population can provide insight into those associations in the more common sporadic AD. METHODS Twenty-four adults (13 male, 11 female; 39±7 years) with DS underwent [11C]PiB, [18F]FDG, and volumetric MRI scans. Voxel-wise associations between PiB SUVR, FDG SUVR, and GM volume were investigated, with and without individual adjustments for variables of interest. RESULTS Positive associations of PiB and age were widespread throughout the neocortex and striatum. Negative associations of FDG and age (frontal, parietal, and temporal cortex) and of GM volume and age (frontal and insular cortex) were observed. PiB and FDG were negatively associated in parietal cortex, after adjustment for GM volume. CONCLUSIONS In adults with DS, early amyloid-β accumulation in the striatum is divergent from sporadic AD; however, despite the early striatal amyloid-β, glucose hypometabolism was confined to the typical AD-associated regions, which occurs similarly in autosomal dominant AD. Importantly, the glucose hypometabolism was not explained solely by increased partial volume effect due to GM volume reductions.
Collapse
Affiliation(s)
- Patrick J. Lao
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
| | - Ben L. Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Instruction and Learning, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tobey J. Betthauser
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
| | - Iulia Mihaila
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
- Department of Human Development and Family Studies, University of Wisconsin-Madison, Madison, WI, USA
| | - Sigan L. Hartley
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
- Department of Human Development and Family Studies, University of Wisconsin-Madison, Madison, WI, USA
| | - Annie D. Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter D. Bulova
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J. Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Dhanabalan Murali
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
| | - Chester A. Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Charles K. Stone
- Department of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Julie C. Price
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Darlynne A. Devenny
- New York State Institute for Research in Developmental Disabilities, Staten Island, NY, USA
| | - Sterling C. Johnson
- Department of Medicine-Geriatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley T. Christian
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin-Madison, Waisman Center, Madison, WI, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
141
|
Buckley RF, Hanseeuw B, Schultz AP, Vannini P, Aghjayan SL, Properzi MJ, Jackson JD, Mormino EC, Rentz DM, Sperling RA, Johnson KA, Amariglio RE. Region-Specific Association of Subjective Cognitive Decline With Tauopathy Independent of Global β-Amyloid Burden. JAMA Neurol 2017; 74:1455-1463. [PMID: 28973551 DOI: 10.1001/jamaneurol.2017.2216] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Importance The ability to explore associations between reports of subjective cognitive decline (SCD) and biomarkers of early Alzheimer disease (AD) pathophysiologic processes (accumulation of neocortical β-amyloid [Aβ] and tau) provides an important opportunity to understand the basis of SCD and AD risk. Objective To examine associations between SCD and global Aβ and tau burdens in regions of interest in clinically healthy older adults. Design, Setting, and Participants This imaging substudy of the Harvard Aging Brain Study included 133 clinically healthy older participants (Clinical Dementia Rating Scale global scores of 0) participating in the Harvard Aging Brain Study who underwent cross-sectional flortaucipir F 18 (previously known as AV 1451, T807) positron emission tomography (FTP-PET) imaging for tau and Pittsburgh compound B carbon 11-labeled PET (PiB-PET) imaging for Aβ. The following 2 regions for tau burden were identified: the entorhinal cortex, which exhibits early signs of tauopathy, and the inferior temporal region, which is more closely associated with AD-related pathologic mechanisms. Data were collected from June 11, 2012, through April 7, 2016. Main Outcomes and Measures Subjective cognitive decline was measured using a previously published method of z-transforming subscales from the Memory Functioning Questionnaire, the Everyday Cognition battery, and a 7-item questionnaire. The Aβ level was measured according to a summary distribution volume ratio of frontal, lateral temporal and parietal, and retrosplenial PiB-PET tracer uptake. The FTP-PET measures were computed as standardized uptake value ratios. Linear regression models focused on main and interactive effects of Aβ, entorhinal cortical, and inferior temporal tau on SCD, controlling for age, sex, educational attainment, and Geriatric Depression Scale score. Results Of the 133 participants, 75 (56.3%) were women and 58 (43.6%) were men; mean (SD) age was 76 (6.9) years (range, 55-90 years). Thirty-nine participants (29.3%) exhibited a high Aβ burden. Greater SCD was associated with increasing entorhinal cortical tau burden (β = 0.35; 95% CI, 0.19-.52; P < .001) and Aβ burden (β = 0.24; 95% CI, 0.08-.40; P = .005), but not inferior temporal tau burden (β = 0.10; 95% CI, -0.08 to 0.28; P = .27). This association between entorhinal cortical tau burden and SCD was largely unchanged after accounting for Aβ burden (β = 0.36; 95% CI, 0.15-.58; P = .001), and no interaction influenced SCD (β = -0.36; 95% CI, -0.34 to 0.09; P = .25). An exploratory post hoc whole-brain analysis also indicated that SCD was predominantly associated with greater tau burden in the entorhinal cortex. Conclusions and Relevance Subjective cognitive decline is indicative of accumulation of early tauopathy in the medial temporal lobe, specifically in the entorhinal cortex, and to a lesser extent, elevated global levels of Aβ. Our findings suggest multiple underlying pathways that motivate SCD that do not necessarily interact to influence SCD endorsement. As such, multiple biological factors must be considered when assessing SCD in clinically healthy older adults.
Collapse
Affiliation(s)
- Rachel F Buckley
- Florey Institutes of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.,Melbourne School of Psychological Science, University of Melbourne, Australia.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Bernard Hanseeuw
- Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Boston.,now affiliated with Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Aaron P Schultz
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Massachusetts General Hospital, Boston.,Department of Psychiatry, Massachusetts General Hospital, Boston
| | - Patrizia Vannini
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Boston
| | - Sarah L Aghjayan
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael J Properzi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown
| | - Jonathan D Jackson
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Elizabeth C Mormino
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Dorene M Rentz
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Reisa A Sperling
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Keith A Johnson
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Department of Radiology, Massachusetts General Hospital, Boston.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts.,Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston
| | - Rebecca E Amariglio
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Neurology, Massachusetts General Hospital, Charlestown.,Department of Radiology, Harvard Medical School, Boston, Massachusetts.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
142
|
Grecchi E, Veronese M, Bodini B, García-Lorenzo D, Battaglini M, Stankoff B, Turkheimer FE. Multimodal partial volume correction: Application to [ 11C]PIB PET/MRI myelin imaging in multiple sclerosis. J Cereb Blood Flow Metab 2017; 37:3803-3817. [PMID: 28569617 PMCID: PMC5718330 DOI: 10.1177/0271678x17712183] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022]
Abstract
The [11C]PIB PET tracer, originally developed for amyloid imaging, has been recently repurposed to quantify demyelination and remyelination in multiple sclerosis (MS). Myelin PET imaging, however, is limited by its low resolution that deteriorates the quantification accuracy of white matter (WM) lesions. Here, we introduce a novel partial volume correction (PVC) method called Multiresolution-Multimodal Resolution-Recovery (MM-RR), which uses the wavelet transform and a synergistic statistical model to exploit MRI structural images to improve the resolution of [11C]PIB PET myelin imaging. MM-RR performance was tested on a phantom acquisition and in a dataset comprising [11C]PIB PET and MR T1- and T2-weighted images of 8 healthy controls and 20 MS patients. For the control group, the MM-RR PET images showed an average increase of 5.7% in WM uptake while the grey-matter (GM) uptake remained constant, resulting in +31% WM/GM contrast. Furthermore, MM-RR PET binding maps correlated significantly with the mRNA expressions of the most represented proteins in the myelin sheath (R2 = 0.57 ± 0.09). In the patient group, MM-RR PET images showed sharper lesion contours and significant improvement in normal-appearing tissue/WM-lesion contrast compared to standard PET (contrast improvement > +40%). These results were consistent with MM-RR performances in phantom experiments.
Collapse
Affiliation(s)
- Elisabetta Grecchi
- Centre for Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Benedetta Bodini
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, UPMC Paris, France
- Service Hospitalier Fréderic Joliot, SHFJ, Orsay, France
| | - Daniel García-Lorenzo
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, UPMC Paris, France
| | - Marco Battaglini
- Department of Neurological and Behavioural Sciences, University of Siena, Siena, Italy
| | - Bruno Stankoff
- Institut du Cerveau et de la Moelle épinière (ICM), CNRS UMR 7225, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, UPMC Paris, France
- Service Hospitalier Fréderic Joliot, SHFJ, Orsay, France
- Department of Neurological and Behavioural Sciences, University of Siena, Siena, Italy
| | - Federico E Turkheimer
- Centre for Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
143
|
Southekal S, Devous MD, Kennedy I, Navitsky M, Lu M, Joshi AD, Pontecorvo MJ, Mintun MA. Flortaucipir F 18 Quantitation Using Parametric Estimation of Reference Signal Intensity. J Nucl Med 2017; 59:944-951. [PMID: 29191858 DOI: 10.2967/jnumed.117.200006] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/03/2017] [Indexed: 11/16/2022] Open
Abstract
PET imaging of tau pathology in Alzheimer disease may benefit from the use of white matter reference regions. These regions have shown reduced variability compared with conventional cerebellar regions in amyloid imaging. However, they are susceptible to contamination from partial-volume blurring of tracer uptake in the cortex. We present a new technique, PERSI (Parametric Estimation of Reference Signal Intensity), for flortaucipir F 18 count normalization that leverages the advantages of white matter reference regions while mitigating potential partial-volume effects. Methods: Subjects with a clinical diagnosis of Alzheimer disease, mild cognitive impairment, or normal cognition underwent T1-weighted MRI and florbetapir imaging (to determine amyloid [Aβ] status) at screening and flortaucipir F 18 imaging at single or multiple time points. Flortaucipir F 18 images, acquired as 4 × 5 min frames 80 min after a 370-MBq injection, were motion-corrected, averaged, and transformed to Montreal Neurological Institute (MNI) space. The PERSI reference region was calculated for each scan by fitting a bimodal gaussian distribution to the voxel-intensity histogram within an atlas-based white matter region and using the center and width of the lower-intensity peak to identify the voxel intensities to be included. Four conventional reference regions were also evaluated: whole cerebellum, cerebellar gray matter, atlas-based white matter, and subject-specific white matter. SUVr (standardized uptake value ratio) was calculated for a statistically defined neocortical volume of interest. Performance was evaluated with respect to test-retest variability in a phase 2 study of 21 subjects (5-34 d between scans). Baseline variability in controls (SD of SUVr and ΔSUVr) and effect sizes for group differences (Cohen d; Aβ-positive impaired vs. Aβ-negative normal) were evaluated in another phase 2 study with cross-sectional data (n = 215) and longitudinal data (n = 142/215; 18 ± 2 mo between scans). Results: PERSI showed superior test-retest reproducibility (1.84%) and group separation ability (cross-sectional Cohen d = 9.45; longitudinal Cohen d = 2.34) compared with other reference regions. Baseline SUVr variability and ΔSUVr were minimal in Aβ control subjects with no specific flortaucipir F 18 uptake (SUVr, 1.0 ± 0.04; ΔSUVr, 0.0 ± 0.02). Conclusion: PERSI reduced variability while enhancing discrimination between diagnostic cohorts. Such improvements could lead to more accurate disease staging and robust measurements of changes in tau burden over time for the evaluation of putative therapies.
Collapse
Affiliation(s)
- Sudeepti Southekal
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Michael D Devous
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Ian Kennedy
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Michael Navitsky
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Ming Lu
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Abhinay D Joshi
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Michael J Pontecorvo
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| | - Mark A Mintun
- Avid Radiopharmaceuticals, Inc. (a wholly owned subsidiary of Eli Lilly and Company), Philadelphia Pennsylvania
| |
Collapse
|
144
|
Entorhinal Tau Pathology, Episodic Memory Decline, and Neurodegeneration in Aging. J Neurosci 2017; 38:530-543. [PMID: 29192126 DOI: 10.1523/jneurosci.2028-17.2017] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/14/2017] [Accepted: 10/07/2017] [Indexed: 12/17/2022] Open
Abstract
The medial temporal lobe (MTL) is an early site of tau accumulation and MTL dysfunction may underlie episodic-memory decline in aging and dementia. Postmortem data indicate that tau pathology in the transentorhinal cortex is common by age 60, whereas spread to neocortical regions and worsening of cognition is associated with β-amyloid (Aβ). We used [18F]AV-1451 and [11C]PiB positron emission tomography, structural MRI, and neuropsychological assessment to investigate how in vivo tau accumulation in temporal lobe regions, Aβ, and MTL atrophy contribute to episodic memory in cognitively normal older adults (n = 83; age, 77 ± 6 years; 58% female). Stepwise regressions identified tau in MTL regions known to be affected in old age as the best predictor of episodic-memory performance independent of Aβ status. There was no interactive effect of MTL tau with Aβ on memory. Higher MTL tau was related to higher age in the subjects without evidence of Aβ. Among temporal lobe subregions, episodic memory was most strongly related to tau-tracer uptake in the parahippocampal gyrus, particularly the posterior entorhinal cortex, which in our parcellation includes the transentorhinal cortex. In subjects with longitudinal MRI and cognitive data (n = 57), entorhinal atrophy mirrored patterns of tau pathology and their relationship with memory decline. Our data are consistent with neuropathological studies and further suggest that entorhinal tau pathology underlies memory decline in old age even without Aβ.SIGNIFICANCE STATEMENT Tau tangles and β-amyloid (Aβ) plaques are key lesions in Alzheimer's disease (AD) but both pathologies also occur in cognitively normal older people. Neuropathological data indicate that tau tangles in the medial temporal lobe (MTL) underlie episodic-memory impairments in AD dementia. However, it remains unclear whether MTL tau pathology also accounts for memory impairments often seen in elderly people and how Aβ affects this relationship. Using tau-specific and Aβ-specific positron emission tomography tracers, we show that in vivo MTL tau pathology is associated with episodic-memory performance and MTL atrophy in cognitively normal adults, independent of Aβ. Our data point to MTL tau pathology, particularly in the entorhinal cortex, as a substrate of age-related episodic-memory loss.
Collapse
|
145
|
Current Role for Biomarkers in Clinical Diagnosis of Alzheimer Disease and Frontotemporal Dementia. Curr Treat Options Neurol 2017; 19:46. [PMID: 29134465 DOI: 10.1007/s11940-017-0484-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose of review Alzheimer's disease (AD) and frontotemporal dementia can often be diagnosed accurately with careful clinical history, cognitive testing, neurological examination, and structural brain MRI. However, there are certain circumstances wherein detection of specific biomarkers of neurodegeneration or underlying AD pathology will impact the clinical diagnosis or treatment plan. We will review the currently available biomarkers for AD and frontotemporal dementia (FTD) and discuss their clinical importance. Recent findings With the advent of 18F-labeled tracers that bind amyloid plaques, amyloid PET is now clinically available for the detection of amyloid pathology and to aid in a biomarker-supported diagnosis of AD or mild cognitive impairment (MCI) due to AD. It is not yet possible to test for the specific FTD pathologies (tau or TDP-43); however, a diagnosis of FTD may be "imaging supported" based upon specific MRI or FDG-PET findings. Cerebrospinal fluid measures of amyloid-beta, total-tau, and phospho-tau are clinically available and allow detection of both of the cardinal pathologies of AD: amyloid and tau pathology. Summary It is appropriate to pursue biomarker testing in cases of MCI and dementia when there remains diagnostic uncertainty and the result will impact diagnosis or treatment. Practically speaking, due to the rising prevalence of amyloid positivity with advancing age, measurement of biomarkers in cases of MCI and dementia is most helpful in early-onset patients, patients with atypical clinical presentations, or when considering referral for AD clinical trials.
Collapse
|
146
|
Chen YJ, Nasrallah IM. Brain amyloid PET interpretation approaches: from visual assessment in the clinic to quantitative pharmacokinetic modeling. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0257-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
147
|
d'Oleire Uquillas F, Jacobs HIL, Hanseeuw B, Marshall GA, Properzi M, Schultz AP, LaPoint MR, Johnson KA, Sperling RA, Vannini P. Interactive versus additive relationships between regional cortical thinning and amyloid burden in predicting clinical decline in mild AD and MCI individuals. NEUROIMAGE-CLINICAL 2017; 17:388-396. [PMID: 29159051 PMCID: PMC5683806 DOI: 10.1016/j.nicl.2017.10.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/09/2017] [Accepted: 10/28/2017] [Indexed: 11/13/2022]
Abstract
The biological mechanisms that link Beta-amyloid (Aβ) plaque deposition, neurodegeneration, and clinical decline in Alzheimer's disease (AD) dementia, have not been completely elucidated. Here we studied whether amyloid accumulation and neurodegeneration, independently or interactively, predict clinical decline over time in a group of memory impaired older individuals [diagnosed with either amnestic mild cognitive impairment (MCI), or mild AD dementia]. We found that baseline Aβ-associated cortical thinning across clusters encompassing lateral and medial temporal and parietal cortices was related to higher baseline Clinical Dementia Rating Sum-of-Boxes (CDR-SB). Baseline Aβ-associated cortical thinning also predicted CDR-SB over time. Notably, the association between CDR-SB change and cortical thickness values from the right lateral temporo-parietal cortex and right precuneus was driven by individuals with high Aβ burden. In contrast, the association between cortical thickness in the medial temporal lobe (MTL) and clinical decline was similar for individuals with high or low Aβ burden. Furthermore, amyloid pathology was a stronger predictor for clinical decline than MTL thickness. While this study validates previous findings relating AD biomarkers of neurodegeneration to clinical impairment, here we show that regions outside the MTL may be more vulnerable and specific to AD dementia. Additionally, excluding mild AD individuals revealed that these relationships remained, suggesting that lower cortical thickness values in specific regions, vulnerable to amyloid pathology, predict clinical decline already at the prodromal stage. Aβ burden is associated with cortical thinning in a pattern consistent with AD. Interaction between Aβ and neocortical thinning predicts clinical decline. MTL thickness predicts clinical decline regardless of Aβ burden. Amyloid pathology is a stronger predictor for clinical decline than MTL thickness.
Collapse
Affiliation(s)
| | - Heidi I L Jacobs
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Bernard Hanseeuw
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Department of Neurology, Saint-Luc University Hospital, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Gad A Marshall
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Molly R LaPoint
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrizia Vannini
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
148
|
Whole-body biodistribution and the influence of body activity on brain kinetic analysis of the 11C-PiB PET scan. Radiol Phys Technol 2017; 10:464-474. [PMID: 28895034 DOI: 10.1007/s12194-017-0419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/27/2017] [Accepted: 09/02/2017] [Indexed: 10/18/2022]
Abstract
Dynamic 11C-PiB PET imaging with kinetic analysis has been performed for accurate quantification of amyloid binding in patients with Alzheimer's disease (AD). In this study, we measured the whole-body biodistribution of 11C-PiB in nine subjects. We then evaluated the effect of body activity on quantitative accuracy of brain 11C-PiB three-dimensional (3D) dynamic PET. Based on clinical biodistribution data, we conducted phantom experiments to estimate the effect of body activity on quantification of the brain 3D dynamic 11C-PiB PET data and the error introduced by body activity using six different PET camera models. One of the PET cameras was used to acquire 11C-PiB brain 3D dynamic PET data on a patient with AD. We calculated the distribution volume ratio (DVR) in two kinetic methods using both the original human time-activity-curve (TAC) data and the TAC corrected for the error caused by body activity. In the early phase, both healthy subjects and patients with AD showed a biodistribution of 11C-PiB that reflected regional blood flow. In the simulated early phase of the phantom experiments, activity outside the field of view led to a maximum 6.0% overestimation of brain activity in the vertex region. Conversely, the effect of body activity on the DVR estimate was small (≤1.2%), probably because the tested kinetic methods did not rely heavily on early phase data. These results indicate that the effect of body activity on brain 11C-PiB PET quantification is generally small and that it depends on the method of kinetic analysis, the region of interest, and the PET camera model used.
Collapse
|
149
|
Maass A, Landau S, Baker SL, Horng A, Lockhart SN, La Joie R, Rabinovici GD, Jagust WJ. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer's disease. Neuroimage 2017; 157:448-463. [PMID: 28587897 PMCID: PMC5814575 DOI: 10.1016/j.neuroimage.2017.05.058] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/20/2017] [Accepted: 05/27/2017] [Indexed: 12/16/2022] Open
Abstract
The recent development of tau-specific positron emission tomography (PET) tracers enables in vivo quantification of regional tau pathology, one of the key lesions in Alzheimer's disease (AD). Tau PET imaging may become a useful biomarker for clinical diagnosis and tracking of disease progression but there is no consensus yet on how tau PET signal is best quantified. The goal of the current study was to evaluate multiple whole-brain and region-specific approaches to detect clinically relevant tau PET signal. Two independent cohorts of cognitively normal adults and amyloid-positive (Aβ+) patients with mild cognitive impairment (MCI) or AD-dementia underwent [18F]AV-1451 PET. Methods for tau tracer quantification included: (i) in vivo Braak staging, (ii) regional uptake in Braak composite regions, (iii) several whole-brain measures of tracer uptake, (iv) regional uptake in AD-vulnerable voxels, and (v) uptake in a priori defined regions. Receiver operating curves characterized accuracy in distinguishing Aβ- controls from AD/MCI patients and yielded tau positivity cutoffs. Clinical relevance of tau PET measures was assessed by regressions against cognition and MR imaging measures. Key tracer uptake patterns were identified by a factor analysis and voxel-wise contrasts. Braak staging, global and region-specific tau measures yielded similar diagnostic accuracies, which differed between cohorts. While all tau measures were related to amyloid and global cognition, memory and hippocampal/entorhinal volume/thickness were associated with regional tracer retention in the medial temporal lobe. Key regions of tau accumulation included medial temporal and inferior/middle temporal regions, retrosplenial cortex, and banks of the superior temporal sulcus. Our data indicate that whole-brain tau PET measures might be adequate biomarkers to detect AD-related tau pathology. However, regional measures covering AD-vulnerable regions may increase sensitivity to early tau PET signal, atrophy and memory decline.
Collapse
Affiliation(s)
- Anne Maass
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States; German Center for Neurodegenerative Diseases, Magdeburg, Germany.
| | - Susan Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States.
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Lab, Berkeley, CA, United States.
| | - Andy Horng
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States.
| | - Samuel N Lockhart
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States.
| | - Renaud La Joie
- Memory and Aging Center, University of California San Francisco, San Francisco, CA, United States.
| | - Gil D Rabinovici
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Lab, Berkeley, CA, United States; Memory and Aging Center, University of California San Francisco, San Francisco, CA, United States.
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Lab, Berkeley, CA, United States; Memory and Aging Center, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
150
|
Lowe VJ, Lundt E, Knopman D, Senjem ML, Gunter JL, Schwarz CG, Kemp BJ, Jack CR, Petersen RC. Comparison of [ 18F]Flutemetamol and [ 11C]Pittsburgh Compound-B in cognitively normal young, cognitively normal elderly, and Alzheimer's disease dementia individuals. NEUROIMAGE-CLINICAL 2017; 16:295-302. [PMID: 28856092 PMCID: PMC5565786 DOI: 10.1016/j.nicl.2017.08.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/12/2017] [Accepted: 08/12/2017] [Indexed: 11/26/2022]
Abstract
Background Understanding the variation in uptake between different amyloid PET tracers is important to appropriately interpret data using different amyloid tracers. Therefore, we compared the uptake differences in [18F]Flutemetamol (FMT) and [11C]PiB (PiB) PET in the same people. Methods Structural MRI, FMT PET and PiB PET were each performed in 30 young cognitively normal (yCN), 31 elderly cognitively normal (eCN) and 21 Alzheimer's disease dementia (AD) participants. PiB and FMT images for each participant were compared quantitatively using voxel- and region-based analyses. Region of interest (ROI) analyses included comparisons of grey matter (GM) regions as well as white matter (WM) regions. Regional comparisons of each tracer between different groups and comparisons of the two modalities within the different groups were performed. To compare mean SUVr between modalities, and between diagnostic groups, we used paired t-tests and Student's t-test, respectively. We also compared the ability of the two tracers to discriminate between diagnostic groups using AUROC estimates. The effect of using different normalization regions on SUVr values was also evaluated. Results Both FMT and PiB showed greater uptake throughout GM structures in AD vs. eCN or yCN. In all dual-modality group comparisons (FMT vs. PiB in yCN, eCN, and AD), greater WM uptake was seen with FMT vs. PiB. In yCN and eCN greater diffuse GM uptake was seen with FMT vs. PiB. When comparing yCN to eCN within each tracer, greater WM uptake was seen in eCN vs yCN. Conclusions Flutemetamol and PiB show similar topographical GM uptake in AD and CN participants and the tracers show comparable group discrimination. Greater WM accumulation with FMT suggests that quantitative differences vs. PiB will be apparent when using WM or GM as a reference region. Both imaging tracers demonstrate increased WM uptake in older people. These findings suggest that using different amyloid tracers or different methods of analyses in serial brain imaging in an individual may result in artifactual amyloid change measurements. Clinical use of several amyloid tracers in the same patient will have challenges that need to be carefully considered. White matter uptake is quantitatively elevated in elderly vs. younger control subjects in both FMT and PiB PET scans. Quantitatively greater white matter uptake is seen in FMT vs. PiB in Alzheimer’s disease dementia and control subjects. Variations between FMT and PiB may create the false appearance of changing brain amyloid levels on serial imaging. FMT and PiB have comparable ability to categorize Alzheimer’s disease dementia and control subjects cross-sectionally. Methods to cross-calibrate different amyloid PET tracers will need to be validated in serial imaging paradigms.
Collapse
Affiliation(s)
- Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Emily Lundt
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - David Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, United States.,Department of Information Technology, Mayo Clinic, Rochester, MN, United States
| | - Jeffrey L Gunter
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | | | - Bradley J Kemp
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | | |
Collapse
|