Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-jwnkl Total loading time: 0 Render date: 2024-07-10T07:17:55.519Z Has data issue: false hasContentIssue false

33 - Brain Imaging for Alzheimer’s Disease Clinical Trials

from Section 4 - Imaging and Biomarker Development in Alzheimer’s Disease Drug Discovery

Published online by Cambridge University Press:  03 March 2022

Jeffrey Cummings
Affiliation:
University of Nevada, Las Vegas
Jefferson Kinney
Affiliation:
University of Nevada, Las Vegas
Howard Fillit
Affiliation:
Alzheimer’s Drug Discovery Foundation
Get access

Summary

Imaging biomarkers are important in the diagnosis and evaluation of treatment effect in AD. The “A/T/N” (amyloid/tau/neurodegeneration) classification notably focused on disease characteristics measurable using imaging or CSF biomarkers. Information obtained with imaging biomarkers can address several challenges in AD trials, by confirming pathology for patient inclusion and target engagement, enabling stratification for analysis based on likely rate of clinical decline, and detecting treatment effect with fewer subjects; it also help to characterize treatment responders and to better understand the neurological basis for clinical response. This chapter discusses how imaging data are generated, the applicability of various imaging endpoints within the overall AD progression pathway, technical issues influencing the reliability and interpretability of the data, and practical steps to incorporate imaging into clinical trials. Applications of volumetric MRI, MRI used in safety assessment, amyloid PET, tau PET, and FDG PET measurement of glucose metabolism are described. Relevant regulatory guidance and the fit of imaging data with blood based or other biomarkers are discussed.

Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 375 - 394
Publisher: Cambridge University Press
Print publication year: 2022

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Khachaturian, ZS. Revised criteria for diagnosis of Alzheimer’s disease: National Institute on Aging–Alzheimer’s Association diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011; 7: 253–6.CrossRefGoogle ScholarPubMed
Jack, CR Jr., Bennett, DA, Blennow, K, et al. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 2016; 87: 539–47.Google Scholar
Magistretti, PJ, Pellerin, L. Cellular mechanisms of brain energy metabolism and their relevance to functional brain imaging. Philos Trans R Soc Lond B Biol Sci 1999; 354: 1155–63.CrossRefGoogle ScholarPubMed
Minter, MR, Taylor, JM, Crack, PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J Neurochem 2016; 136: 457–74.CrossRefGoogle ScholarPubMed
Janelidze, S, Mattsson, N, Palmqvist, S, et al. Plasma p-tau181 in Alzheimer’s disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat Med 2020; 26: 379–86.Google Scholar
Rogers, MB. Tau PET scans turn positive when amyloid does; symptoms follow. AlzForum series: Clinical Trials on Alzheimer’s Disease 2019, Part 8 of 9. January, 2020. Available at: www.alzforum.org/news/conference-coverage/tau-pet-scans-turn-positive-when-amyloid-does-symptoms-follow (accessed November 15, 2020).Google Scholar
Palmqvist, S, Schöll, M, Strandberg, O, et al. Earliest accumulation of β-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat Comm 2017; 8: 1214.CrossRefGoogle ScholarPubMed
Ossenkoppele, R, Schonhaut, DR, Schöll, M, et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain 2016; 139: 1551–67.Google Scholar
Koychev, I, Gunn, RN, Firouzian, A, et al. PET tau and amyloid-β burden in mild Alzheimer’s disease: divergent relationship with age, cognition, and cerebrospinal fluid biomarkers. J Alzheimers Dis 2017; 60: 283–93.CrossRefGoogle ScholarPubMed
Guerrero-Muñoz, MJ, Gerson, J, Castillo-Carranza, DL. Tau oligomers: the toxic player at synapses in Alzheimer’s disease. Front Cell Neurosci 2015; 9: 464.Google Scholar
Vargas, LM, Cerpa, W, Muñoz, FJ, Zanlungo, S, Alvarez, AR. Amyloid-β oligomers synaptotoxicity: the emerging role of EphA4/c-Abl signaling in Alzheimer’s disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864A: 1148–59.Google Scholar
Insel, PS, Ossenkoppele, R, Gessert, D, et al. Time to amyloid positivity and preclinical changes in brain metabolism, atrophy, and cognition: evidence for emerging amyloid pathology in Alzheimer’s disease. Front Neurosci 2017; 11: 281.Google Scholar
La Joie, R, Visani, AV, Baker, SL, et al. Prospective longitudinal atrophy in Alzheimer’s disease correlates with the intensity and topography of baseline tau-PET. Sci Transl Med 2020; 12: eaau5732.Google Scholar
Timmers, T, Ossenkoppele, R, Wolters, EE, et al. Associations between quantitative [18F]flortaucipir tau PET and atrophy across the Alzheimer’s disease spectrum. Alzheimers Res Ther 2019; 11: 60.Google Scholar
Salloway, S, Sperling, R, Fox, NC, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 2014; 370: 322–33.Google Scholar
Weintraub, S, Carrillo, MC, Farias, ST, et al. Measuring cognition and function in the preclinical stage of Alzheimer’s disease. Alzheimers Dement (N Y) 2018; 4: 6475.Google Scholar
Fazekas, F, Chawluk, JB, Alavi, A, et al. MR signal abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. AJR Am J Roentgenol 1987; 149: 351–6.CrossRefGoogle ScholarPubMed
Mirza, SS, Saeed, U, Knight, J, et al. APOE ε4, white matter hyperintensities, and cognition in Alzheimer and Lewy body dementia. Neurology 2019; 93: e1807–19.CrossRefGoogle ScholarPubMed
Orgogozo, JM, Gilman, S, Dartigues, JF. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003; 61: 4654.CrossRefGoogle Scholar
Sperling, RA, Jack, CR Jr., Black, SE, et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement 2011; 7: 367–85.CrossRefGoogle ScholarPubMed
Greenberg, SM, Vernooij, MW, Cordonnier, C, et al. Cerebral microbleeds: a guide to detection and interpretation. Lancet Neurol 2009; 8: 165–74.CrossRefGoogle ScholarPubMed
Tolar, M, Abushakra, S, Hey, JA, Porsteinsson, A, Sabbagh, M. Aducanumab, gantenerumab, BAN2401, and ALZ-801-the first wave of amyloid-targeting drugs for Alzheimer’s disease with potential for near term approval. Alzheimers Res Ther 2020; 12: 95.CrossRefGoogle ScholarPubMed
Wu, J, Dong, Q, Gui, J, et al. Predicting brain amyloid using multivariate morphometry statistics, sparse coding, and correntropy: validation in 1,125 individuals from the ADNI and OASIS database. bioRxiv 2020;DOI: http://doi.org/10.1101/2020.10.16.343137.CrossRefGoogle Scholar
Lukic, AS, Andrews, RD, Bourakova, V, et al. MRI, FDG, and early frame amyloid image classifiers to characterize and differentiate Alzheimer’s disease variants and non-AD dementias. Alzheimers Dement 2018; 14: P1429–30.CrossRefGoogle Scholar
Davatzikos, C, Resnick, SM, Wu, X, Parmpi, P, Clark, CM. Individual patient diagnosis of AD and FTD via high-dimensional pattern classification of MRI. Neuroimage 2008; 41: 1220–7.Google Scholar
Giorgio, J, Jagust, WJ, Baker, S, et al. Predicting future regional tau accumulation in asymptomatic and early Alzheimer’s disease. bioRxiv 2020;DOI: http://doi.org/10.1101/2020.08.15.252601.Google Scholar
Yu, P, Sun, J, Wolz, R, et al. Operationalizing hippocampal volume as an enrichment biomarker for amnestic mild cognitive impairment trials: effect of algorithm, test-retest variability, and cut point on trial cost, duration, and sample size. Neurobiol Aging 2014; 35: 808–18.Google Scholar
Rabin, JS, Neal, TE, Nierle, HE, et al. Multiple markers contribute to risk of progression from normal to mild cognitive impairment. Neuroimage Clin 2020; 28: 102400.Google Scholar
Woodard, JL, Bellaali, Y, Dricot, L, et al. Multivariate prediction of rate of decline in memory functioning over six years using imaging biomarkers. Alzheimers Dement 2020; 16: e045645.Google Scholar
Leung, KK, Barnes, J, Ridgway, GR, et al. Alzheimer’s Disease Neuroimaging Initiative. Automated cross-sectional and longitudinal hippocampal volume measurement in mild cognitive impairment and Alzheimer’s disease. Neuroimage 2010; 51: 1345–59.Google Scholar
Hashimoto, M, Kazui, H, Matsumoto, K, et al. Does donepezil treatment slow the progression of hippocampal atrophy in patients with Alzheimer’s disease? Am J Psychiatry 2005; 162: 676–82.Google Scholar
Turner, RS, Hebron, ML, Lawler, A, et al. Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer’s disease. Ann Neurol 2020; 88: 183–94.Google Scholar
Gauthier, S, Aisen, PS, Ferris, SH. Effect of tramiprosate in patients with mild-to-moderate Alzheimer’s disease: exploratory analyses of the MRI sub-group of the Alphase study. J Nutr Health Aging 2009; 13: 550–7.CrossRefGoogle ScholarPubMed
Wessels, AM, Tariot, PN, Zimmer, JA, et al. Efficacy and safety of lanabecestat for treatment of early and mild Alzheimer disease: the AMARANTH and DAYBREAK-ALZ randomized clinical trials. JAMA Neurol 2020; 77: 199209.Google Scholar
Novak, G, Fox, N, Clegg, S, et al. Changes in brain volume with bapineuzumab in mild to moderate Alzheimer’s disease. J Alzheimers Dis 2016; 49: 1123–34.Google Scholar
Fox, NC, Black, RS, Gilman, S, et al. Effects of Abeta immunization (AN1792) on MRI measures of cerebral volume in Alzheimer disease. Neurology 2005; 64: 1563–72.Google Scholar
Cheriyan, J, Kim, S, Wolansky, LJ, Cook, SD, Cadavid, D. Impact of inflammation on brain volume in multiple sclerosis. Arch Neurol 2012; 69: 82–8.Google Scholar
Reuter, M, Schmansky, NJ, Rosas, HD, Fischl, B. Within-subject template estimation for unbiased longitudinal image analysis. Neuroimage 2012; 61: 1402–18.Google Scholar
Iglesias, JE, Van Leemput, K, Augustinack, J, et al. Bayesian longitudinal segmentation of hippocampal substructures in brain MRI using subject-specific atlases. Neuroimage 2016; 141: 542–55.Google Scholar
Teipel, SJ, Kuper-Smith, JO, Bartels, C, et al. Multicenter tract-based analysis of microstructural lesions within the Alzheimer’s disease spectrum: association with amyloid pathology and diagnostic usefulness. J Alzheimers Dis 2019; 72: 455–65.Google Scholar
Elahi, FM, Marx, G, Cobigo, Y, et al. Longitudinal white matter change in frontotemporal dementia subtypes and sporadic late onset Alzheimer’s disease. Neuroimage Clin 2017; 16: 595603.Google Scholar
Pasternak, O, Sochen, N, Gur, Y, Intrator, N, Assaf, Y. Free water elimination and mapping from diffusion MRI. Magn Reson Med 2009; 62: 717–30.CrossRefGoogle ScholarPubMed
Hoy, AR, Ly, M, Carlsson, CM, et al. Microstructural white matter alterations in preclinical Alzheimer’s disease detected using free water elimination diffusion tensor imaging. PloS One 2017; 12: e0173982.Google Scholar
Binnewijzend, MA, Kuijer, JP, van der Flier, WM, et al. Distinct perfusion patterns in Alzheimer’s disease, frontotemporal dementia and dementia with Lewy bodies. Eur Radiol 2014; 24: 2326–33.CrossRefGoogle ScholarPubMed
Dukart, J, Holiga, Š, Chatham, C, et al. Cerebral blood flow predicts differential neurotransmitter activity. Sci Rep 2018; 8: 4074.Google Scholar
Guo, H, Grajauskas, L, Habash, B, D’Arc, RC, Song, X. Functional MRI technologies in the study of medication treatment effect on Alzheimer’s disease. Aging Med (Milton) 2018; 1: 7595.Google Scholar
Zhang, N, Gordon, ML, Goldberg, TE. Cerebral blood flow measured by arterial spin labeling MRI at resting state in normal aging and Alzheimer’s disease. Neurosci Biobehav Rev 2017; 72: 168–75.CrossRefGoogle Scholar
Smith, LA, Melbourne, A, Owen, D, et al. Cortical cerebral blood flow in ageing: effects of haematocrit, sex, ethnicity and diabetes. Eur Radiol 2019; 29: 5549–58.CrossRefGoogle ScholarPubMed
Greicius, MD, Srivastava, G, Reiss, AL, Menon, V. Default-mode network activity distinguishes Alzheimer’s disease from healthy aging: evidence from functional MRI. Proc Natl Acad Sci USA 2004; 101: 4637–42.Google Scholar
Churchill, NW, Spring, R, Afshin-Pour, B, Dong, F, Strother, SC. An automated, adaptive framework for optimizing preprocessing pipelines in task-based functional MRI. PLoS One 2015; 10: e0131520.Google Scholar
Matthews, DC, Mao, X, Dowd, K, et al. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer’s disease: a pilot multimodal neuroimaging study. Brain 2021; Jun 18: awab222.Google Scholar
Maul, S, Giegling, I, Rujescu, D. Proton magnetic resonance spectroscopy in common dementias: current status and perspectives. Front Psychiatry 2020; 11: 769.Google Scholar
van Berckel, BN, Ossenkoppele, R, Tolboom, N, et al. Longitudinal amyloid imaging using 11C-PiB: methodologic considerations. J Nucl Med 2013; 54: 1570–6.Google Scholar
Sevigny, J, Chiao, P, Bussière, T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016; 537: 50–6.Google Scholar
Ostrowitzki, S, Lasser, RA, Dorflinger, E, et al. A Phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease. Alzheimers Res Ther 2017; 9: 95.Google Scholar
Schmidt, ME, Matthews, DC, Andrews, RD, Mosconi, L. Positron emission tomography in Alzheimer disease: diagnosis and use as biomarker endpoints. In Translational Neuroimaging, McArthur RA (ed.). New York: Academic Press; 2013: 131–74.Google Scholar
Klunk, WE, Koeppe, RA, Price, JC. The Centiloid project: standardizing quantitative amyloid plaque estimation by PET. Alzheimers Dement 2015; 11: 1–15.e154.CrossRefGoogle ScholarPubMed
Chen, K, Roontiva, A, Thiyyagura, P, et al. Alzheimer’s Disease Neuroimaging Initiative. Improved power for characterizing longitudinal amyloid-β PET changes and evaluating amyloid-modifying treatments with a cerebral white matter reference region. J Nucl Med 2015; 56: 560–6.Google Scholar
Schmidt, ME, Chiao, P, Klein, G, et al. The influence of biological and technical factors on quantitative analysis of amyloid PET: points to consider and recommendations for controlling variability in longitudinal data. Alzheimers Dement 2015; 11: 1050–68.Google Scholar
Rostomian, AH, Madison, C, Rabinovici, GD, Jagust, WJ. Early 11C-PIB frames and 18F-FDG PET measures are comparable: a study validated in a cohort of AD and FTLD patients. J Nucl Med 2011; 52: 173–9.Google Scholar
Brendel, M, Barthel, H, van Eimeren, T, et al. Assessment of 18F-PI-2620 as a biomarker in progressive supranuclear palsy. JAMA Neurol 2020; 77: 1408–19.Google Scholar
Tagai, K, Ono, M, Kubota, M, et al. High-contrast in vivo imaging of tau pathologies in Alzheimer’s and non-Alzheimer’s disease tauopathies. Neuron 2020; 109: 42–58;DOI: http://doi.org/10.1016/j.neuron.2020.09.042.Google Scholar
Betthauser, TJ, Cody, KA, Zammit, MD, et al. In vivo characterization and quantification of neurofibrillary tau PET radioligand 18F-MK-6240 in humans from Alzheimer disease dementia to young controls. J Nucl Med 2019; 60: 93–9.Google Scholar
Maass, A, Landau, S, Baker, SL, Alzheimer’s Disease Neuroimaging Initiative. Comparison of multiple tau-PET measures as biomarkers in aging and Alzheimer’s disease.Neuroimage 2017; 157: 448–63.Google Scholar
Baker, SL, Maass, A, Jagust, WJ. Considerations and code for partial volume correcting [18F]-AV-1451 tau PET data. Data Brief 2017; 15: 648–57;DOI: http://doi.org/10.1016/j.dib.2017.10.024.Google Scholar
Southekal, S, Devous, MD Sr., Kennedy, I, et al. Flortaucipir F 18 quantitation using parametric estimation of reference signal intensity. J Nucl Med 2018; 59: 944–51.Google Scholar
Beyer, L, Nitschmann, A, Barthel, H, et al. Early-phase [18F]PI-2620 tau-PET imaging as a surrogate marker of neuronal injury. Eur J Nucl Med Mol Imaging 2020; 47: 2911–22.Google Scholar
Foster, NL, Heidebrink, JL, Clark, CM, et al. FDG-PET improves accuracy in distinguishing frontotemporal dementia and Alzheimer’s disease. Brain 2007; 130: 2616–35.Google Scholar
Xia, Y, Lu, S, Wen, L, et al. Automated identification of dementia using FDG-PET imaging. Biomed Res Int 2014; 2014: 421743.CrossRefGoogle ScholarPubMed
Matthews, DC, Ritter, A, Thomas, RG, et al. Rasagiline effects on glucose metabolism, cognition, and tau in Alzheimer’s dementia. Alzheimers Dement (N Y) 2021; 7: e12106.Google Scholar
Beckett, L, Harvey, D, Donohue, M, et al. Biostatistics Core ADNI 2 summary and ADNI 3 plans. Available at https://slideplayer.com/slide/12666696/ (accessed November 20, 2020).Google Scholar
Chen, K, Langbaum, JB, Fleisher, AS, et al. Alzheimer’s Disease Neuroimaging Initiative. Twelve-month metabolic declines in probable Alzheimer’s disease and amnestic mild cognitive impairment assessed using an empirically pre-defined statistical region-of-interest: findings from the Alzheimer’s Disease Neuroimaging Initiative. Neuroimage 2010; 51: 654–64.Google Scholar
Kadir, A, Andreasen, N, Almkvist, O, et al. Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer’s disease. Ann Neurol 2008; 63: 621–31.Google Scholar
Kreisl, WC, Kim, MJ, Coughlin, JM, et al. PET imaging of neuroinflammation in neurological disorders. Lancet Neurol 2020; 19: 940–50.CrossRefGoogle ScholarPubMed
Mecca, AP, Chen, MK, O’Dell, RS, et al. In vivo measurement of widespread synaptic loss in Alzheimer’s disease with SV2A PET. Alzheimers Dement 2020; 16: 974–82.Google Scholar
Josephs, KA, Dickson, DW, Tosakulwong, N, et al. Rates of hippocampal atrophy and presence of post-mortem TDP-43 in patients with Alzheimer’s disease: a longitudinal retrospective study. Lancet Neurol 2017; 16: 917–24.Google Scholar
US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), and Center for Biologics Evaluation and Research (CBER). Early Alzheimer’s disease: developing drugs for treatment. Guidelines for industry. Available at: www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM596728.pdf (accessed November 15, 2020).Google Scholar
European Medicines Agency. Clinical investigation of medicines for the treatment of Alzheimer’s disease/ CPMP/EWP/553/1995. Available at: www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-investigation-medicines-treatment-alzheimers-disease-revision-2_en.pdf (accessed November 20, 2020).Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×