Relationships between flortaucipir PET tau binding and amyloid burden, clinical diagnosis, age and cognition

Michael J Pontecorvo, Michael D Devous Sr, Michael Navitsky, Ming Lu, Stephen Salloway, Frederick W Schaerf, Danna Jennings, Anupa K Arora, Anne McGeehan, Nathaniel C Lim, Hui Xiong, Abhinay D Joshi, Andrew Siderowf, Mark A Mintun, 18F-AV-1451-A05 investigators, Michael J Pontecorvo, Michael D Devous Sr, Michael Navitsky, Ming Lu, Stephen Salloway, Frederick W Schaerf, Danna Jennings, Anupa K Arora, Anne McGeehan, Nathaniel C Lim, Hui Xiong, Abhinay D Joshi, Andrew Siderowf, Mark A Mintun, 18F-AV-1451-A05 investigators

Abstract

The advent of tau-targeted positron emission tomography tracers such as flortaucipir (18F-AV-1451, also known as 18F-T807) have made it possible to investigate the sequence of development of tau and amyloid-β in relationship to age, and to the development of cognitive impairment due to Alzheimer's disease. In this study, flortaucipir tau and florbetapir amyloid positron emission tomography were obtained for 217 subjects including 16 young and 58 older cognitively normal subjects, 95 subjects with mild cognitive impairment (Mini-Mental State Examination 24-30) and 48 subjects with clinically-defined possible or probable Alzheimer's disease (Mini-Mental State Examination >10). Images were evaluated visually and quantitatively by regional and voxel-based cortical to cerebellar standard uptake value ratios. For amyloid positron emission tomography positive (Aβ+) subjects, flortaucipir neocortical standard uptake value ratio was significantly higher with more advanced clinical stage (Alzheimer's disease > mild cognitive impairment > older cognitively normal) and was significantly elevated for Aβ+ mild cognitive impairment and Alzheimer's disease subjects relative to the respective Aβ- subjects. In contrast, florbetapir Aβ- older cognitively normal subjects showed an increase in flortaucipir standard uptake value ratios in mesial temporal lobe regions (amygdala, hippocampus/choroid plexus region of interest) compared to younger cognitively normal subjects, but no increased standard uptake value ratios in neocortical regions. Analysis of covariance with planned contrasts showed no differences in regional or composite posterior neocortical flortaucipir standard uptake value ratio as a function of diagnostic group among Aβ- older cognitively normal or clinically diagnosed Alzheimer's disease or mild cognitive impairment subjects. The pattern of flortaucipir distribution among Aβ+ subjects was reminiscent of the cross-sectional distribution of tau reported in post-mortem pathology studies, in that the most commonly affected regions were the inferior and lateral temporal lobes, the same regions where the first signs of increased retention appeared in Aβ+ cognitively normal subjects. However, there was large variability in extent/density of flortaucipir tau binding among Aβ+ subjects. Although high neocortical flortaucipir retention was consistently associated with an Aβ+ florbetapir positron emission tomography scan, not all Aβ+ subjects had elevated flortaucipir standard uptake value ratios. Finally, within the Aβ+ group, increasing levels of flortaucipir tau binding were associated with increased cognitive impairment, as assessed by Mini-Mental State Examination and Alzheimer's Disease Assessment Scale. These results suggest development of tau beyond the mesial temporal lobe is associated with, and may be dependent on, amyloid accumulation. Further, the results are consistent with the hypothesis that cortical tau is associated with cognitive impairment.

Keywords: 18F-AV-1451; PET; florbetapir; flortaucipir; tau.

© The Author (2017). Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

Figure 1
Figure 1
Mean flortaucipir voxel-wise SUVr images for each age/diagnostic cohort by florbetapir PET amyloid status (Aβ+ or Aβ−). Images are scaled from 1–2.5 SUVr units and overlaid on an average of MRI from all study patients in radiologic orientation such that the right side of each image represents left hemisphere of the brain. (A) Aβ−; (B) Aβ+. AD = Alzheimer’s disease; ROI = region of interest.
Figure 2
Figure 2
Flortaucipir voxel-wise SUVr images for representative individual Aβ− control (YCN/OCN) and Aβ+ OCN, MCI, and Alzheimer’s disease subjects. (A) Aβ− control (YNC/OCN), (B–D) Aβ+ OCN, MCI and Alzheimer’s disease subjects, respectively. PET images are scaled from 1–2.5 SUVr units and overlaid on each subject’s respective MRI. Note in addition to differences in overall extent of retention, there are also subjects with relatively limbic sparing (D, first row) and limbic predominant (D, third row) patterns (arrows) consistent with the pathology literature (Murray et al., 2011). AD = Alzheimer’s disease; y/o = years old.
Figure 3
Figure 3
Posterior neocortical composite SUVr for individual subjects by diagnostic category.X-axis: (diagnostic group, amyloid status), YCN are shown in red. Y-axis: flortaucipir (AV1451) combination SUVr. Horizontal reference line shows upper 99% confidence limit for YCN. AD = Alzheimer’s disease; CN = cognitively normal.
Figure 4
Figure 4
Aβ+ subjects with low cortical average SUVR values. Subjects with low flortaucipir SUVr values may still have abnormal tracer retention in limited cortical regions. AD = Alzheimer’s disease; CN = cognitively normal; ROI = region of interest.
Figure 5
Figure 5
Relationship between florbetapir PET SUVr, flortaucipir SUVr, diagnosis, age and ADAS score. (A) Scatterplot of relationship between florbetapir and flortaucipir SUVr for individual subjects by age, diagnosis and amyloid status. (B) Scatterplot of relationship between age and flortaucipir SUVr for individual subjects by age, diagnosis and amyloid status (r and P-values refer to Aβ+ subjects only). (C and D) Scatterplot of relationship between flortaucipir SUVr and cognition for individual subjects by diagnosis and amyloid status and age (<75 years, C; >75 years D). AD = Alzheimer’s disease; CN = cognitively normal.
Figure 6
Figure 6
Mean flortaucipir voxel-wise SUVr images for Aβ+ MCI and Alzheimer’s disease subjects <75 years and ≥75 years old. Note the similarity between the pattern of demented (Alzheimer’s disease) subjects ≥75 years to MCI subjects <75 years of age. AD = Alzheimer’s disease.

References

    1. Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer's disease: past, present and future. Neuropharmacology 2014; 76 (Part A): 27–50.
    1. Bolmont T, Clavaguera F, Meyer-Luehmann M, Herzig MC, Radde R, Staufenbiel M. et al. Induction of tau pathology by intracerebral infusion of amyloid-beta-containing brain extract and by amyloid-beta deposition in APP x tau transgenic mice. Am J Pathol 2007; 171: 2012–20.
    1. Bouras C, Hof PR, Giannakopoulos Pl, Michel JP, Morrison JH. Regional distribution of neurofibrillary tangles and senile plaques in the cerebral cortex of elderly patients: a quantitative evaluation of a one-year autopsy population from a geriatric hospital. Cerebral Cortex 1994; 4: 138–50.
    1. Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol 2006; 112: 389–404.
    1. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991; 82: 239–59.
    1. Brier MR, Gordon B, Friedrichsen K, McCarthy J, Stern A, Christensen J. et al. Tau and Aβ imaging, CSR measures, and cognition in Alzheimer’s disease. Sci Transl Med 2016; 8: 338ra66.
    1. Chhatwal JP, Schultz AP, Marshall GA, Boot B, Gomez-Isla T, Dumurgier J. et al. Temporal T807 binding correlates with CSF tau and phospho-tau in normal elderly. Neurology 2016; 87: 920–6.
    1. Chien DT, Szardenings AK, Bahri S, Walsh JC, Mu F, Xia C. et al. Early clinical PET imaging results with the novel PHF-Tau radioligand [F18]-T808. J Alzheimers Dis 2014; 38: 171–84.
    1. Cho H, Choi JY, Hwang MS, Kim YJ, Lee HM, Lee HS. et al. In vivo cortical spreading patter of tau and amyloid in the Alzheimer’s disease spectrum. Ann Neurol. 2016a; 80: 247–58.
    1. Cho H, Choi JY, Hwang MS, Lee JH, Kim YJ, Lyoo CH. et al. Tau PET in Alzheimer disease and mild cognitive impairment. Neurology 2016b; 87: 375–83.
    1. Clark CM, Pontecorvo MJ, Beach TG, Bedell BJ, Coleman RE, Doraiswamy PM. et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-beta plaques: a prospective cohort study. Lancet Neurol 2012;11: 669–78.
    1. Crary JF, Trojanowski JQ, Schneider JA, Abisambra JF, Abner EL, Alafuzoff I. et al. Primary age-related tauopathy (PART): a common pathology associated with human aging. Acta Neuropathol 2014; 128: 755–66.
    1. Curtis C, Gamez JE, Singh U, Sadowsky CH, Villena T, Sabbagh MN. et al. Phase 3 trial of flutemetamol labeled with radioactive fluorine 18 imaging and neuritic plaque density. JAMA Neurology 2015; 72: 287–94.
    1. Del Ser T, Steinwachs KC, Gertz HJ, Andres MV, Gomez-Carrillo B, Medina M. et al. Treatment of Alzheimer's disease with the GSK-3 inhibitor tideglusib: a pilot study. J Alzheimers Dis 2013; 33: 205–15.
    1. Friston KJ, Ashburner JT, Kiebel SJ, Nichols TE, Penny WD, editors. Statistical parametric mapping: the analysis of functional brain images. London: Academic Press; 2007.
    1. Fonov VS, Evans AC, McKinstry RC, Almli CR, Collins DL. Unbiased nonlinear average age-appropriate brain templates from birth to adulthood. NeuroImage 2009; 47: S102.
    1. Giacobini E, Gold G. Alzheimer disease therapy[mdash]moving from amyloid-[beta] to tau. Nat Rev Neurol 2013; 9: 677–86.
    1. Goedert M, Spillantini MG, Cairns NJ, Crowther RA. Tau proteins of alzheimer paired helical filaments: abnormal phosphorylation of all six brain isoforms. Neuron 1992; 8: 159–68.
    1. Gordon BA, Friedrichsen K, Brier M, Blazey T, Su Y, Christensen J. et al. The relationship between cerebrospinal fluid markers of Alzheimer pathology and positron emission tomography tau imaging. Brain 2016; 139 (Pt 8): 2249–60.
    1. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002; 297: 353–56.
    1. Holtzman DM, Carrillo MC, Hendrix JA, Bain LJ, Catafau AM, Gault LM. et al. Tau: from research to clinical development. Alzheimers Dement. 2016; 12: 1033–39.
    1. Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC. et al. National institute on aging Alzheimer's association guidelines for the neuropathologic assessment of Alzheimer's disease. Alzheimers Dement 2012; 8: 1–13.
    1. Ikonomovic MD, Abrahamson EE, Price JC, Mathis CA, Klunk WE. [F-18]AV-1451 PET retention in choroid plexus: more than “off target” binding. Ann Neurol. 2016; 80: 307–8.
    1. Jack CR, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW. et al. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol 2010; 9: 119–28.
    1. Jack CR, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, Aisen PS. et al. Tracking pathophysiological processes in Alzheimer's disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol 2013;12: 207–16.
    1. Jack CR, Therneau TM, Wiste HJ, Weigand SD, Knopman DS, Lowe VJ. et al. Transition rates between amyloid and neurodegeneration biomarker states and to dementia: a population-based longitudinal cohort study. Lancet Neurol 2016; 15: 56–64.
    1. Johnson KA, Schultz A, Betensky RA, Becker JA, Sepulcre J, Rentz D. et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol 2016; 79: 110–19.
    1. Joshi AD, Pontecorvo MJ, Lu M, Grundman M, Skovronsky DM, Mintun MA. et al. A semi-automated method for quantification of florbetapir F 18 PET images. J Nucl Med, 2015; 56: 1736–41.
    1. Kidd M. Paired helical filaments in electron microscopy of Alzheimer's disease. Nature 1963; 197: 192–3.
    1. Lewis J, Dickerson DW, Lin WL, Chisholm L, Corral A, Jones G. et al. Enhanced neurofibrillary degenereration in transgenic mice expressing mutant tau and APP. Science 2001; 293: 1487–91.
    1. Lockhart SN, Baker SL, Okamura N, Furukawa K, Ishiki A, Furumoto S. et al. Dynamic PET measures of tau accumulation in cognitively normal older adults and Alzheimer’s disease patients measured using [18F] THK-5351. PLoS One 2016; 11: e0158460.
    1. Lowe VJ, Curran G, Fang P, Liesinger AM, Josephs KA, Parisi JE. et al. An autoradiographic evaluation of AV-1451 Tau PET in dementia. Acta Neuropathologica Communications 2016; 4: 58.
    1. Marquié M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG. et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol 2015; 78: 787–800.
    1. Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J. et al. Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron 2013; 79: 1094–108.
    1. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and down syndrome. Proc. Natl Acad Sci 1985; 82: 4245–9.
    1. Middleton LE, Grinberg LT, Miller B, Kawas C, Yaffe K. Neuropathologic features associated with Alzheimer disease diagnosis: age matters. Neurology 2011;77: 1737–44.
    1. Murray ME, Graff-Radford NR, Ross OA, Petersen RC, Duara R, Dickson DW. Neuropathologically defined subtypes of Alzheimer’s disease with distinct clinical characteristics: a retrospective study. Lancet Neurol 2011; 10: 785–96.
    1. Nelson PT, Jicha GA, Schmitt FA, Liu H, Davis DG, Mendiondo MS. et al. Clinicopathologic correlations in a large Alzheimer disease center autopsy cohort: neuritic plaques and neurofibrillary tangles “do count” when staging disease severity. J Neuropathol Exp Neurol 2007; 66: 1136–46.
    1. Okamura N, Furumoto S, Harada R, Tago T, Yoshikawa T, Fodero-Tavoletti M. et al. Novel 18F-labeled arylquinoline derivatives for noninvasive imaging of tau pathology in Alzheimers disease. J Nucl Med 2013; 54: 1420–27.
    1. Okamura N, Furumoto S, Fodero-Tavoletti MT, Mulligan RS, Harada R, Yates P. et al. Non-invasive assessment of Alzheimer’s disease neurofibrillary pathology using 18F-THK5105 PET. Brain 2014; 137(Pt 6): 1762–71.
    1. Ossenkoppele R, Schonhaut DR, Scholl M, Lockhart SN, Ayakta N, Baker SL. et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain 2016; 139(Pt 5): 1551–67.
    1. Pooler AM, Noble W, Hanger DP. A role for tau at the synapse in Alzheimer's isease pathogenisis. Neuropharmacology 2014; 76: 1–8.
    1. Prohovnik I, Perl DP, Davis KL, Libow L, Lesser G, Haroutunian V. Dissociation of neuropathology from severity of dementia in late-onset Alzheimer disease. Neurology 2006;66: 49–55.
    1. Querfurth HW, LaFerla FM. Alzheimer's disease. NEJM 2010; 362: 329–44.
    1. Riley KP, Snowdon DA, Markesbery WR. Alzheimer's neurofibrillary pathology and the spectrum of cognitive function: findings from the Nun Study. Ann Neurol 2002; 51: 567–77.
    1. Sabri O, Sabbagh MN, Seibyl J, Barthel H, Akatsu H, Ouchi Y. et al. Florbetaben PET imaging to detect amyloid beta plaques in Alzheimer's disease: phase 3 study. Alzheimers Dement 2015; 11: 964–74.
    1. Sander K, Lashley T, Gami P, Gendron T, Lythgoe MF, Rohrer JD. et al. Characterization of tau positron emission tomography tracer [18F] AV-1451 binding to postmortem tissue in Alzheimer’s disease, primary tauopathies, and other dementias. Alzheimers Dement 2016; 12: 1116–124.
    1. Shcherbinin S, Schwarz AJ, Joshi A, Navitsky M, Flitter M, Shankle WR. et al. Kinetics of the tau PET tracer [18F]-AV-1451 (T807) in subjects with normal cognitive function, mild cognitive impairment and Alzheimer’s disease. J Nucl Med 2016; 57: 1535–42. jnumed.115.170027.
    1. Schöll M, Lockhart SN, Schonhaut DR, O'Neil JP, Janabi M, Ossenkoppele R. et al. PET imaging of tau deposition in the aging human brain. Neuron 2016; 89: 971–82.
    1. Schwarz AJ, Yu P, Miller BB, Shcherbinin S, Dickson J, Navitsky M. et al. Regional profiles of [18F]-AV-1451 tau PET images recapitulate key features of Braak histopathological stages. Brain 2016; 139 (Pt 5): 1539–50.
    1. Selkoe DJ. The molecular pathology of Alzheimer’s disease. Neuron 1991; 6: 487–98.
    1. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM. et al. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 2011; 7: 280–92.
    1. Stancu IC, Vasconcelos B, Terwel D, Dewachter I. Models of β-amyloid induced tau-pathology: the long and folded road to understand the mechanism. Mol Neuroidegen 2014; 9: 51–64.
    1. Tomlinson BE, Blessed G, Roth M. Observations on the brains of demented old people. J Neurol Sci 1970; 11: 205–42.
    1. Vasconcelos B, Stancu IC, Buist A, Bird M, Wang P, Vanoosthuyse A. et al. Heterotypic seeding of tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo. Acta Neuropathol. 2016; 131: 549–569.
    1. Xia CF, Arteaga J, Chen G, Gangadharmath U, Gomez LF, Kasi D. et al. [18F]T807, a novel tau positron emission tomography imaging agent for Alzheimer's disease. Alzheimers Dement 2013; 9: 666–76.
    1. Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE. et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 2013; 80: 402–14.

Source: PubMed

3
Abonneren