Early detection of amyloid load using 18F-florbetaben PET

Santiago Bullich, Núria Roé-Vellvé, Marta Marquié, Susan M Landau, Henryk Barthel, Victor L Villemagne, Ángela Sanabria, Juan Pablo Tartari, Oscar Sotolongo-Grau, Vincent Doré, Norman Koglin, Andre Müller, Audrey Perrotin, Aleksandar Jovalekic, Susan De Santi, Lluís Tárraga, Andrew W Stephens, Christopher C Rowe, Osama Sabri, John P Seibyl, Mercè Boada, Santiago Bullich, Núria Roé-Vellvé, Marta Marquié, Susan M Landau, Henryk Barthel, Victor L Villemagne, Ángela Sanabria, Juan Pablo Tartari, Oscar Sotolongo-Grau, Vincent Doré, Norman Koglin, Andre Müller, Audrey Perrotin, Aleksandar Jovalekic, Susan De Santi, Lluís Tárraga, Andrew W Stephens, Christopher C Rowe, Osama Sabri, John P Seibyl, Mercè Boada

Abstract

Background: A low amount and extent of Aβ deposition at early stages of Alzheimer's disease (AD) may limit the use of previously developed pathology-proven composite SUVR cutoffs. This study aims to characterize the population with earliest abnormal Aβ accumulation using 18F-florbetaben PET. Quantitative thresholds for the early (SUVRearly) and established (SUVRestab) Aβ deposition were developed, and the topography of early Aβ deposition was assessed. Subsequently, Aβ accumulation over time, progression from mild cognitive impairment (MCI) to AD dementia, and tau deposition were assessed in subjects with early and established Aβ deposition.

Methods: The study population consisted of 686 subjects (n = 287 (cognitively normal healthy controls), n = 166 (subjects with subjective cognitive decline (SCD)), n = 129 (subjects with MCI), and n = 101 (subjects with AD dementia)). Three categories in the Aβ-deposition continuum were defined based on the developed SUVR cutoffs: Aβ-negative subjects, subjects with early Aβ deposition ("gray zone"), and subjects with established Aβ pathology.

Results: SUVR using the whole cerebellum as the reference region and centiloid (CL) cutoffs for early and established amyloid pathology were 1.10 (13.5 CL) and 1.24 (35.7 CL), respectively. Cingulate cortices and precuneus, frontal, and inferior lateral temporal cortices were the regions showing the initial pathological tracer retention. Subjects in the "gray zone" or with established Aβ pathology accumulated more amyloid over time than Aβ-negative subjects. After a 4-year clinical follow-up, none of the Aβ-negative or the gray zone subjects progressed to AD dementia while 91% of the MCI subjects with established Aβ pathology progressed. Tau deposition was infrequent in those subjects without established Aβ pathology.

Conclusions: This study supports the utility of using two cutoffs for amyloid PET abnormality defining a "gray zone": a lower cutoff of 13.5 CL indicating emerging Aβ pathology and a higher cutoff of 35.7 CL where amyloid burden levels correspond to established neuropathology findings. These cutoffs define a subset of subjects characterized by pre-AD dementia levels of amyloid burden that precede other biomarkers such as tau deposition or clinical symptoms and accelerated amyloid accumulation. The determination of different amyloid loads, particularly low amyloid levels, is useful in determining who will eventually progress to dementia. Quantitation of amyloid provides a sensitive measure in these low-load cases and may help to identify a group of subjects most likely to benefit from intervention.

Trial registration: Data used in this manuscript belong to clinical trials registered in ClinicalTrials.gov ( NCT00928304 , NCT00750282 , NCT01138111 , NCT02854033 ) and EudraCT (2014-000798-38).

Keywords: Alzheimer’s disease; Amyloid-beta; Florbetaben; Mild cognitive impairment; PET; Subjective memory complainers.

Conflict of interest statement

SB, NRV, NK, AM, AP, AJ, and AS are employees of Life Molecular Imaging GmbH (formerly Piramal Imaging GmbH). SDS is an employee of Eisai Inc. and a former employee of Life Molecular Imaging Inc. (formerly Piramal Pharma Inc). HB and OS received research support, consultant honoraria, and travel expenses from Piramal Imaging GmbH. Victor L. Villemagne has received speaker’s honoraria from Piramal Imaging, GE Healthcare, Avid Pharmaceuticals, AstraZeneca, and Hoffmann-La Roche and consulting fees for Novartis, Lundbeck, Abbvie, Shanghai Green Valley Pharmaceutical Co. LTD, and Hoffmann-La Roche. Christopher C. Rowe has received research grants from Bayer Schering Pharma, Piramal Imaging, Avid Radiopharmaceuticals, Navidea, GE Healthcare, AstraZeneca, and Biogen. John Seibyl holds equity in Invicro and consulting fees from LMI, Roche, Biogen, AbVie, and Invicro. M. Boada has received research funds from the following private donors: Grifols SA, Caixabank S.A., Piramal Imaging, Araclon Biotech, Laboratorios Echevarne, Fundació Castell de Peralada, and Fundació La Pedrera and has participated in advisory boards of Araclon Biotech, Biogen, Bioibérica, Eisai, Grifols, Lilly, Merck, Nutricia, Roche, Schwabe Farma, Servier, and Kyowa Kirin.

No other potential conflict of interest relevant to this article was reported.

Figures

Fig. 1
Fig. 1
Histograms of standardized uptake value ratios (SUVRs) and cortex centiloids (CLs) in young healthy controls (n = 65, dataset #1), fitted Gaussian distribution (red), and SUVR cutoff derived for the detection of early Aβ pathology (red dashed line)
Fig. 2
Fig. 2
Receiver operating characteristic curves obtained using MRI-derived regions of interest (ROIs, left) and centiloid (right) used to derive standardized uptake value ratio cutoffs for the established Alzheimer’s disease pathology from a group of elderly healthy controls (n = 66) and subjects with AD dementia (n = 73, dataset #2)
Fig. 3
Fig. 3
Heat maps of standardized uptake value ratios (SUVRs, left) and ΔSUVRs (=SUVR − SUVR(t = T50)) (right) of all the participants in the analysis (n = 686, datasets #1, #2, #3, #4, and #5). Each column of the heat map represents one subject of the sample. The subjects were sorted according to their composite SUVR (increasing from left to right)
Fig. 4
Fig. 4
Histograms of composite standardized uptake value ratios (SUVRs) and centiloids (CLs) for the sample of subjective cognitive decline (SCD) (n = 168, dataset #3) subjects at baseline (first column) and at follow-up (central column). Red and blue lines represent the SUVR abnormality cutoffs for early Aβ detection and established Aβ pathology, respectively. The rate of Aβ accumulation in SCD (and 95% confidence interval in red) in three categories of the composite SUVR continuum (Aβ-negative, gray zone, and established Aβ deposition) is shown on the right column. ROI region of interest
Fig. 5
Fig. 5
Histograms of composite standardized uptake value ratios (SUVRs) and centiloids (CLs) for the sample of mild cognitive impairment (MCI) (n = 44, dataset #4) subjects are shown on the top row. Subjects that progressed to Alzheimer’s disease (AD) dementia after a 4-year clinical follow-up are shown in gray. Red and blue lines represent the SUVR abnormality cutoffs for early Aβ detection and established Aβ pathology, respectively. The rate of Aβ accumulation in MCI subjects (and 95% confidence interval in red) in three categories of the composite SUVR continuum: Aβ-negative, gray zone, and with established Aβ deposition, is shown on the bottom row. ROI region of interest
Fig. 6
Fig. 6
Scatter plots of Flortaucipir (FTP) standardized uptake value ratios (SUVRs) versus 18F-florbetaben composite SUVRs using MRI-based regions of interest (ROIs, top row) and FTP SUVRs versus centiloids (CLs, bottom row) (n = 270, dataset #5). Red and blue lines represent the composite SUVR abnormality cutoffs for early Aβ detection and established Aβ pathology, respectively
Fig. 7
Fig. 7
Sensitivities, specificities, and agreement rates between visual assessment and quantitative assessment when using several cutoffs to dichotomize the sample (top row) and composite standardized uptake value ratio (SUVR) versus subject identifier (bottom row) (n = 416) (datasets #1, #2, #3, and #4). Red and blue lines represent the composite SUVR abnormality cutoffs for early Aβ detection and established Aβ pathology, respectively. ROI region of interest, CL centiloid

References

    1. Braak H, Braak E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging. 1997;18(4):351–357. doi: 10.1016/S0197-4580(97)00056-0.
    1. Villemagne VL, Burnham S, Bourgeat P, Brown B, Ellis KA, Salvado O, et al. Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: a prospective cohort study. Lancet Neurol. 2013;12(4):357–367. doi: 10.1016/S1474-4422(13)70044-9.
    1. Blennow K, Zetterberg H, Rinne JO, Salloway S, Wei J, Black R, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol. 2012;69(8):1002–1010. doi: 10.1001/archneurol.2012.90.
    1. Uenaka K, Nakano M, Willis BA, Friedrich S, Ferguson-Sells L, Dean RA, et al. Comparison of pharmacokinetics, pharmacodynamics, safety, and tolerability of the amyloid beta monoclonal antibody solanezumab in Japanese and white patients with mild to moderate Alzheimer disease. Clin Neuropharmacol. 2012;35(1):25–29. doi: 10.1097/WNF.0b013e31823a13d3.
    1. Weninger S, Carrillo MC, Dunn B, Aisen PS, Bateman RJ, Kotz JD, et al. Collaboration for Alzheimer’s prevention: principles to guide data and sample sharing in preclinical Alzheimer’s disease trials. Alzheimers Dement. 2016;12(5):631–632. doi: 10.1016/j.jalz.2016.04.001.
    1. McDade E, Bateman RJ. Stop Alzheimer’s before it starts. Nature. 2017;547(7662):153–155. doi: 10.1038/547153a.
    1. Ritchie CW, Molinuevo JL, Truyen L, Satlin A, Van der Geyten S, Lovestone S. Development of interventions for the secondary prevention of Alzheimer’s dementia: the European Prevention of Alzheimer’s Dementia (EPAD) project. Lancet Psychiatry. 2016;3(2):179–186. doi: 10.1016/S2215-0366(15)00454-X.
    1. Sperling RA, Jack CR, Jr, Aisen PS. Testing the right target and right drug at the right stage. Sci Transl Med. 2011;3(111):111cm33. doi: 10.1126/scitranslmed.3002609.
    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(8):964–974. doi: 10.1016/j.jalz.2015.02.004.
    1. Seibyl J, Catafau AM, Barthel H, Ishii K, Rowe CC, Leverenz JB, et al. Impact of training method on the robustness of the visual assessment of 18F-florbetaben PET scans: results from a phase-3 study. J Nucl Med. 2016;57(6):900–906. doi: 10.2967/jnumed.115.161927.
    1. Bullich S, Villemagne VL, Catafau AM, Jovalekic A, Koglin N, Rowe CC, et al. Optimal reference region to measure longitudinal amyloid-beta change with 18F-florbetaben PET. J Nucl Med. 2017;58(8):1300–1306. doi: 10.2967/jnumed.116.187351.
    1. Bullich S, Seibyl J, Catafau AM, Jovalekic A, Koglin N, Barthel H, et al. Optimized classification of 18F-florbetaben PET scans as positive and negative using an SUVR quantitative approach and comparison to visual assessment. Neuroimage Clin. 2017;15:325–332. doi: 10.1016/j.nicl.2017.04.025.
    1. Barthel H, Gertz HJ, Dresel S, Peters O, Bartenstein P, Buerger K, et al. Cerebral amyloid-beta PET with florbetaben (18F) in patients with Alzheimer’s disease and healthy controls: a multicentre phase 2 diagnostic study. Lancet Neurol. 2011;10(5):424–435. doi: 10.1016/S1474-4422(11)70077-1.
    1. Jennings D, Seibyl J, Sabbagh M, Lai F, Hopkins W, Bullich S, et al. Age dependence of brain beta-amyloid deposition in Down syndrome: an [18F] florbetaben PET study. Neurology. 2015;84(5):500–507. doi: 10.1212/WNL.0000000000001212.
    1. Tuszynski T, Rullmann M, Luthardt J, Butzke D, Tiepolt S, Gertz HJ, et al. Evaluation of software tools for automated identification of neuroanatomical structures in quantitative beta-amyloid PET imaging to diagnose Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2016;43(6):1077–1087. doi: 10.1007/s00259-015-3300-6.
    1. Villemagne VL, Ong K, Mulligan RS, Holl G, Pejoska S, Jones G, et al. Amyloid imaging with (18)F-florbetaben in Alzheimer disease and other dementias. J Nucl Med. 2011;52(8):1210–1217. doi: 10.2967/jnumed.111.089730.
    1. Doré V, Bullich S, Rowe CC, Bourgeat P, Konate S, Sabri O, et al. Comparison of 18F-florbetaben quantification results using the standard centiloid, MR-based, and MR-less CapAIBL((R)) approaches: validation against histopathology. Alzheimers Dement. 2019;15(6):807–816. doi: 10.1016/j.jalz.2019.02.005.
    1. Ong K, Villemagne VL, Bahar-Fuchs A, Lamb F, Chételat G, Raniga P, et al. (18)F-florbetaben Abeta imaging in mild cognitive impairment. Alzheimers Res Ther. 2013;5(1):4. doi: 10.1186/alzrt158.
    1. Bischof GN, Jacobs HIL. Subthreshold amyloid and its biological and clinical meaning: long way ahead. Neurology. 2019;93(2):72–79. doi: 10.1212/WNL.0000000000007747.
    1. Fantoni E, Collij L, Lopes Alves I, Buckley C, Farrar G. AMYPAD consortium. The spatial-temporal ordering of amyloid pathology and opportunities for PET imaging. J Nucl Med. 2020;61(2):166–171. doi: 10.2967/jnumed.119.235879.
    1. Grothe MJ, Barthel H, Sepulcre J, Dyrba M, Sabri O, Teipel SJ. In vivo staging of regional amyloid deposition. Neurology. 2017;89(20):2031–2038. doi: 10.1212/WNL.0000000000004643.
    1. Mattsson N, Palmqvist S, Stomrud E, Vogel J, Hansson O. Staging beta-amyloid pathology with amyloid positron emission tomography. JAMA Neurol. 2019;76(11):1319–29.
    1. Collij LE, Heeman F, Salvadó G, Ingala S, Altomare D, de Wilde A, et al. Multitracer model for staging cortical amyloid deposition using PET imaging. Neurology. 2020;95(11):e1538–e1553. doi: 10.1212/WNL.0000000000010256.
    1. Pascoal TA, Therriault J, Mathotaarachchi S, Kang MS, Shin M, Benedet AL, et al. Topographical distribution of Abeta predicts progression to dementia in Abeta positive mild cognitive impairment. Alzheimers Dement (Amst) 2020;12(1):e12037.
    1. Hanseeuw BJ, Betensky RA, Mormino EC, Schultz AP, Sepulcre J, Becker JA, et al. PET staging of amyloidosis using striatum. Alzheimers Dement. 2018;14(10):1281–1292. doi: 10.1016/j.jalz.2018.04.011.
    1. Rodriguez-Gomez O, Sanabria A, Perez-Cordon A, Sanchez-Ruiz D, Abdelnour C, Valero S, et al. FACEHBI: a prospective study of risk factors, biomarkers and cognition in a cohort of individuals with subjective cognitive decline. Study rationale and research protocols. J Prev Alzheimers Dis. 2017;4(2):100–108.
    1. Joshi A, Koeppe RA, Fessler JA. Reducing between scanner differences in multi-center PET studies. Neuroimage. 2009;46(1):154–159. doi: 10.1016/j.neuroimage.2009.01.057.
    1. Tzourio-Mazoyer N, Landeau B, Papathanassiou D, Crivello F, Etard O, Delcroix N, et al. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. Neuroimage. 2002;15(1):273–289. doi: 10.1006/nimg.2001.0978.
    1. Rowe CC, Ackerman U, Browne W, Mulligan R, Pike KL, O'Keefe G, et al. Imaging of amyloid beta in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: proof of mechanism. Lancet Neurol. 2008;7(2):129–135. doi: 10.1016/S1474-4422(08)70001-2.
    1. Klunk WE, Koeppe RA, Price JC, Benzinger TL, Devous MD, Sr, Jagust WJ, et al. The Centiloid Project: standardizing quantitative amyloid plaque estimation by PET. Alzheimers Dement. 2015;11(1):1–15 e1–4. doi: 10.1016/j.jalz.2014.07.003.
    1. Rowe CC, Doré V, Jones G, Baxendale D, Mulligan RS, Bullich S, et al. 18F-Florbetaben PET beta-amyloid binding expressed in centiloids. Eur J Nucl Med Mol Imaging. 2017;44(12):2053–2059. doi: 10.1007/s00259-017-3749-6.
    1. Whittington A, Sharp DJ, Gunn RN. Spatiotemporal distribution of β-amyloid in Alzheimer disease is the result of heterogeneous regional carrying capacities. J Nucl Med. 2018;59(5):822–827. doi: 10.2967/jnumed.117.194720.
    1. Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, et al. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991;41:479–486. doi: 10.1212/WNL.41.4.479.
    1. Bullich S. Converging evidence for a “gray-zone” of amyloid burden and its relevance. in AAIC. 2020. Virtual.
    1. Salvadó G, Molinuevo JL, Brugulat-Serrat A, Falcon C, Grau-Rivera O, Suárez-Calvet M, et al. Centiloid cut-off values for optimal agreement between PET and CSF core AD biomarkers. Alzheimers Res Ther. 2019;11(1):27. doi: 10.1186/s13195-019-0478-z.
    1. Mormino EC, Brandel MG, Madison CM, Rabinovici GD, Marks S, Baker SL, et al. Not quite PIB-positive, not quite PIB-negative: slight PIB elevations in elderly normal control subjects are biologically relevant. Neuroimage. 2012;59(2):1152–1160. doi: 10.1016/j.neuroimage.2011.07.098.
    1. La Joie R, Ayakta N, Seeley WW, Borys E, Boxer AL, DeCarli C, et al. Multisite study of the relationships between antemortem [11C]PIB-PET Centiloid values and postmortem measures of Alzheimer’s disease neuropathology. Alzheimers Dement. 2019;15(2):205–216. doi: 10.1016/j.jalz.2018.09.001.
    1. Villeneuve S, Rabinovici GD, Cohn-Sheehy BI, Madison C, Ayakta N, Ghosh PM, et al. Existing Pittsburgh Compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain. 2015;138(Pt 7):2020–2033. doi: 10.1093/brain/awv112.
    1. Cho H, Choi JY, Hwang MS, Kim YJ, Lee HM, Lee HS, et al. In vivo cortical spreading pattern of tau and amyloid in the Alzheimer disease spectrum. Ann Neurol. 2016;80(2):247–258. doi: 10.1002/ana.24711.
    1. Palmqvist S, Schöll M, Strandberg O, Mattsson N, Stomrud E, Zetterberg H, et al. Earliest accumulation of β-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat Commun. 2017;8(1):1214. doi: 10.1038/s41467-017-01150-x.
    1. Guo T, Landau SM, Jagust WJ. Detecting earlier stages of amyloid deposition using PET in cognitively normal elderly adults. Neurology. 2020;94(14):e1512–e1524. doi: 10.1212/WNL.0000000000009216.
    1. Guo T, Dukart J, Brendel M, Rominger A, Grimmer T, Yakushev I. Rate of beta-amyloid accumulation varies with baseline amyloid burden: implications for anti-amyloid drug trials. Alzheimers Dement. 2018;14(11):1387–1396. doi: 10.1016/j.jalz.2018.05.013.
    1. Insel PS, Mormino EC, Aisen PS, Thompson WK, Donohue MC. Neuroanatomical spread of amyloid beta and tau in Alzheimer’s disease: implications for primary prevention. Brain Commun. 2020;2(1):fcaa007. doi: 10.1093/braincomms/fcaa007.
    1. Lopes Alves I, Collij LE, Altomare D, Frisoni GB, Saint-Aubert L, Payoux P, et al. Quantitative amyloid PET in Alzheimer’s disease: the AMYPAD prognostic and natural history study. Alzheimers Dement. 2020;16(5):750–758. doi: 10.1002/alz.12069.
    1. Bullich S, Barthel H, Koglin N, Becker GA, De Santi S, Jovalekic A, et al. Validation of noninvasive tracer kinetic analysis of (18)F-Florbetaben PET using a dual-time-window acquisition protocol. J Nucl Med. 2018;59(7):1104–1110. doi: 10.2967/jnumed.117.200964.

Source: PubMed

3
Sottoscrivi