Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-25T06:03:54.875Z Has data issue: false hasContentIssue false

Section 3 - Alzheimer’s Disease Clinical Trials

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

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 135 - 360
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

References

Zhou, Y. Choice of designs and doses for early phase trials. Fundam Clin Pharmacol 2004; 18: 373–8.CrossRefGoogle ScholarPubMed
Lindstrom-Gommers, L, Mullin, T. International Conference on Harmonization: recent reforms as a driver of global regulatory harmonization and innovation in medical products. Clin Pharmacol Ther 2019; 105: 926–31.CrossRefGoogle ScholarPubMed
Nair, AB, Jacob, S. A simple practice guide for dose conversion between animals and humans. J Basic Clin Pharm 2016; 7: 2731.CrossRefGoogle Scholar
Muller, PY, Milton, M, Lloyd, P, Sims, J, Brennan, FR. The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal antibodies. Curr Opin Biotechnol 2009; 20: 722–9.Google Scholar
Agoram, BM. Use of pharmacokinetic/pharmacodynamic modelling for starting dose selection in first-in-human trials of high-risk biologics. Br J Clin Pharmacol 2009; 67: 153–60.CrossRefGoogle ScholarPubMed
Wheeler, GM, Mander, AP, Bedding, A, et al. How to design a dose-finding study using the continual reassessment method. BMC Med Res Methodol 2019; 19: 18.CrossRefGoogle ScholarPubMed
Dekker, M, Bouvy, JC, O’Rourke, D, et al. Alignment of European regulatory and health technology assessments: a review of licensed products for Alzheimer’s disease. Front Med (Lausanne) 2019; 6: 73.Google Scholar
Jaki, T, Clive, S, Weir, CJ. Principles of dose finding studies in cancer: a comparison of trial designs. Cancer Chemother Pharmacol 2013; 71: 1107–14.CrossRefGoogle Scholar
Algorta, J, Pena, MA, Maraschiello, C, et al. Phase I clinical trial with desoxypeganine, a new cholinesterase and selective MAO-A inhibitor: tolerance and pharmacokinetics study of escalating single oral doses. Methods Find Exp Clin Pharmacol 2008; 30: 141–7.Google Scholar
Schneider, LS, Geffen, Y, Rabinowitz, J, et al. Low-dose ladostigil for mild cognitive impairment: a Phase 2 placebo-controlled clinical trial. Neurology 2019; 93: e1474–84.Google Scholar
Ivanova, A, Murphy, M. An adaptive first in man dose-escalation study of NGX267: statistical, clinical, and operational considerations. J Biopharm Stat 2009; 19: 247–55.CrossRefGoogle ScholarPubMed
Kutzsche, J, Jurgens, D, Willuweit, A, et al. Safety and pharmacokinetics of the orally available antiprionic compound PRI-002: a single and multiple ascending dose phase I study. Alzheimers Dement (N Y) 2020; 6: e12001.Google Scholar
Grundman, M, Morgan, R, Lickliter, JD, et al. A Phase 1 clinical trial of the sigma-2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer’s disease. Alzheimers Dement (N Y) 2019; 5: 20–6.Google ScholarPubMed
Ahn, JE, Carrieri, C, Dela Cruz, F, et al. Pharmacokinetic and pharmacodynamic effects of a gamma-secretase modulator, PF-06648671, on CSF amyloid-beta peptides in randomized Phase I studies. Clin Pharmacol Ther 2020; 107: 211–20.CrossRefGoogle ScholarPubMed
Ye, N, Monk, SA, Daga, P, et al. Clinical bioavailability of the novel BACE1 inhibitor lanabecestat (AZD3293): assessment of tablet formulations versus an oral solution and the impact of gastric pH on pharmacokinetics. Clin Pharmacol Drug Dev 2018; 7: 233–43.Google Scholar
Ferrero, J, Williams, L, Stella, H, et al. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimers Dement (N Y) 2016; 2: 169–76.Google Scholar
Lues, I, Weber, F, Meyer, A, et al. A Phase 1 study to evaluate the safety and pharmacokinetics of PQ912, a glutaminyl cyclase inhibitor, in healthy subjects. Alzheimers Dement (N Y) 2015; 1: 182–95.Google ScholarPubMed
Kerbrat, A, Ferre, JC, Fillatre, P, et al. Acute neurologic disorder from an inhibitor of fatty acid amide hydrolase. N Engl J Med 2016; 375: 1717–25.Google Scholar
Cutler, NR, Jhee, SS, Cyrus, P, et al. Safety and tolerability of metrifonate in patients with Alzheimer’s disease: results of a maximum tolerated dose study. Life Sci 1998; 62: 1433–41.Google Scholar
Lopez-Arrieta, JM, Schneider, L. Metrifonate for Alzheimer’s disease. Cochrane Database Syst Rev 2006; 2: CD003155.Google Scholar
Sramek, JJ, Block, GA, Reines, SA, et al. A multiple-dose safety trial of eptastigmine in Alzheimer’s disease, with pharmacodynamic observations of red blood cell cholinesterase. Life Sci 1995; 56: 319–26.Google ScholarPubMed
Moss, DE, Fariello, RG, Sahlmann, J, et al. A randomized Phase I study of methanesulfonyl fluoride, an irreversible cholinesterase inhibitor, for the treatment of Alzheimer’s disease. Br J Clin Pharmacol 2013; 75: 1231–9.Google Scholar
Jia, JY, Zhao, QH, Liu, Y, et al. Phase I study on the pharmacokinetics and tolerance of ZT-1, a prodrug of huperzine A, for the treatment of Alzheimer’s disease. Acta Pharmacol Sin 2013; 34: 976–82.Google Scholar
Patat, A, Parks, V, Raje, S, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of ascending single and multiple doses of lecozotan in healthy young and elderly subjects. Br J Clin Pharmacol 2009; 67: 299308.Google Scholar
Nirogi, R, Mudigonda, K, Bhyrapuneni, G, et al. Safety, tolerability and pharmacokinetics of the serotonin 5-HT6 receptor antagonist, SUVN-502, in healthy young adults and elderly subjects. Clin Drug Investig 2018; 38: 401–15.CrossRefGoogle ScholarPubMed
Othman, AA, Haig, G, Florian, H, et al. Safety, tolerability and pharmacokinetics of the histamine H3 receptor antagonist, ABT-288, in healthy young adults and elderly volunteers. Br J Clin Pharmacol 2013; 75: 1299–311.Google Scholar
Hey, JA, Yu, JY, Versavel, M, et al. Clinical pharmacokinetics and safety of ALZ-801, a novel prodrug of tramiprosate in development for the treatment of Alzheimer’s disease. Clin Pharmacokinet 2018; 57: 315–33.CrossRefGoogle ScholarPubMed
Cebers, G, Alexander, RC, Haeberlein, SB, et al. AZD3293: pharmacokinetic and pharmacodynamic effects in healthy subjects and patients with Alzheimer’s disease. J Alzheimers Dis 2017; 55: 1039–53.Google Scholar
Brazier, D, Perry, R, Keane, J, Barrett, K, Elmaleh, DR. Pharmacokinetics of cromolyn and ibuprofen in healthy elderly volunteers. Clin Drug Investig 2017; 37: 1025–34.Google Scholar
Sevigny, J, Chiao, P, Bussiere, T, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature 2016; 537: 50–6.Google Scholar
Novak, P, Schmidt, R, Kontsekova, E, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer’s disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol 2017; 16: 123–34.CrossRefGoogle ScholarPubMed
Novak, P, Schmidt, R, Kontsekova, E, et al. FUNDAMANT: an interventional 72-week phase 1 follow-up study of AADvac1, an active immunotherapy against tau protein pathology in Alzheimer’s disease. Alzheimers Res Ther 2018; 10: 108.CrossRefGoogle ScholarPubMed
Karch, FE, Lasagna, L. Toward the operational identification of adverse drug reactions. Clin Pharmacol Ther 1977; 21: 247–54.Google Scholar
Timmers, M, Sinha, V, Darpo, B, et al. Evaluating potential QT effects of JNJ-54861911, a BACE inhibitor in single- and multiple-ascending dose studies, and a thorough QT trial with additional retrospective confirmation, using concentration-QTc analysis. J Clin Pharmacol 2018; 58: 952–64.Google Scholar
Laczo, J, Markova, H, Lobellova, V, et al. Scopolamine disrupts place navigation in rats and humans: a translational validation of the Hidden Goal Task in the Morris water maze and a real maze for humans. Psychopharmacology (Ber) 2017; 234: 535–47.Google Scholar
Jobert, M, Wilson, FJ, Ruigt, GS, et al. Guidelines for the recording and evaluation of pharmaco-EEG data in man: the International Pharmaco-EEG Society (IPEG). Neuropsychobiology 2012; 66: 201–20.CrossRefGoogle ScholarPubMed
Tsolaki, A, Kazis, D, Kompatsiaris, I, Kosmidou, V, Tsolaki, M. Electroencephalogram and Alzheimer’s disease: clinical and research approaches. Int J Alzheimers Dis 2014; 2014: 349249.Google Scholar
Liem-Moolenaar, M, de Boer, P, Timmers, M, et al. Pharmacokinetic–pharmacodynamic relationships of central nervous system effects of scopolamine in healthy subjects. Br J Clin Pharmacol 2011; 71: 886–98.Google Scholar
Ebert, U, Kirch, W. Scopolamine model of dementia: electroencephalogram findings and cognitive performance. Eur J Clin Invest 1998; 28: 944–9.CrossRefGoogle ScholarPubMed
Adler, G, Brassen, S. Short-term rivastigmine treatment reduces EEG slow-wave power in Alzheimer patients. Neuropsychobiology 2001; 43: 273–6.CrossRefGoogle ScholarPubMed
Adler, G, Brassen, S, Chwalek, K, Dieter, B, Teufel, M. Prediction of treatment response to rivastigmine in Alzheimer’s dementia. J Neurol Neurosurg Psychiatry 2004; 75: 292–4.Google ScholarPubMed
Balsters, JH, O’Connell, RG, Martin, MP, et al. Donepezil impairs memory in healthy older subjects: behavioural, EEG and simultaneous EEG/fMRI biomarkers. PLoS One 2011; 6: e24126.Google Scholar
Uslaner, JM, Kuduk, SD, Wittmann, M, et al. Preclinical to human translational pharmacology of the novel M1 positive allosteric modulator MK-7622. J Pharmacol Exp Ther 2018; 365: 556–66.Google Scholar
Smailovic, U, Koenig, T, Kareholt, I, et al. Quantitative EEG power and synchronization correlate with Alzheimer’s disease CSF biomarkers. Neurobiol Aging 2018; 63: 8895.Google Scholar
Simpraga, S, Alvarez-Jimenez, R, Mansvelder, HD, et al. EEG machine learning for accurate detection of cholinergic intervention and Alzheimer’s disease. Sci Rep 2017; 7: 5775.Google Scholar
Van Broeck, B, Timmers, M, Ramael, S, et al. Impact of frequent cerebrospinal fluid sampling on Abeta levels: systematic approach to elucidate influencing factors. Alzheimers Res Ther 2016; 8: 21.Google Scholar

References

Blass, BE. Basic Principles of Drug Discovery and Development. Boston, MA: Academic Press; 2015.Google Scholar
Yuan, J, Pang, H, Tong, T, et al. Seamless Phase IIa/IIb and enhanced dose-finding adaptive design. J Biopharm Stat 2016; 26: 912–23.Google Scholar
Cummings, J, Ritter, A, Zhong, K. Clinical trials for disease-modifying therapies in Alzheimer’s disease: a primer, lessons learned, and a blueprint for the future. J Alzheimers Dis 2018; 64: S322.CrossRefGoogle Scholar
Jack, CR Jr., Bennett, DA, Blennow, K, et al. NIA–AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14: 535–62.CrossRefGoogle Scholar
Cummings, J, Feldman, HH, Scheltens, P. The “rights” of precision drug development for Alzheimer’s disease. Alzheimers Res Ther 2019; 11: 76.Google Scholar
Bauer, P, Kieser, M. Combining different phases in the development of medical treatments within a single trial. Stat Med 1999; 18: 1833–48.Google Scholar
Scheltens, P, De Strooper, B, Kivipelto, M, et al. Alzheimer’s disease. Lancet 2021; 397: 1577–90.CrossRefGoogle ScholarPubMed
Molinuevo, JL, Ayton, S, Batrla, R, et al. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol 2018; 136: 821–53.CrossRefGoogle ScholarPubMed
National Institute on Aging, Alzheimer’s Association. International Alzheimer’s and Related Dementias Research Portfolio: Common Alzheimer’s and Related Dementias Research Ontology (CADRO). Bethesda, MD: National Institutes of Health; 2020.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (N Y) 2020; 6: e12050.Google Scholar
de Wilde, A, Reimand, J, Teunissen, CE, et al. Discordant amyloid-β PET and CSF biomarkers and its clinical consequences. Alzheimers Res Ther 2019; 11: 7884.Google Scholar
Reimand, J, Boon, BDC, Collij, LE, et al. Amyloid-β PET and CSF in an autopsy-confirmed cohort. Ann Clin Transl Neurol 2020; 7: 2150–60.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
Golla, SS, Wolters, EE, Timmers, T, et al. Parametric methods for 18[F]flortaucipir PET. J Cereb Blood Flow Metab 2020; 40: 365–73.Google Scholar
Wolters, EE, Ossenkoppele, R, Verfaillie, SCJ, et al. Regional 18[F]flortaucipir PET is more closely associated with disease severity than CSF p-tau in Alzheimer’s disease. Eur J Nucl Med Mol Imaging 2020; 47: 2866–78.Google Scholar
Scheltens, NME, Briels, CT, Yaqub, M, et al. Exploring effects of souvenaid on cerebral glucose metabolism in Alzheimer’s disease. Alzheimers Dement (N Y) 2019; 5: 492500.Google Scholar
Soininen, H, Solomon, A, Visser, PJ, et al. 36-month LipiDiDiet multinutrient clinical trial in prodromal Alzheimer’s disease. Alzheimers Dement 2021; 17: 2940.CrossRefGoogle ScholarPubMed
Bennet, CF, Krainer, AR, Cleveland, DW. Antisense oligonucleotide therapies for neurodegenerative diseases. Annu Rev Neurosci 2019; 42: 385406.Google Scholar
Babiloni, C, Blinowska, K, Bonanni, L, et al. What electrophysiology tells us about Alzheimer’s disease: a window into the synchronization and connectivity of brain neurons. Neurobiol Aging 2020; 85: 5873.Google Scholar
Busche, MA, Chen, X, Henning, AH, et al. Critical role of soluble amyloid-β for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA 2012; 109: 8740–5.Google Scholar
Palop, JJ, Mucke, L. Network abnormalities and interneuron dysfunction in Alzheimer disease. Nat Rev Neurosci 2016; 17: 777–92.CrossRefGoogle ScholarPubMed
Engels, M, Hillebrand, A, van der Flier, WM, et al. Slowing of hippocampal activity correlates with cognitive decline in early onset Alzheimer’s disease. An MEG study with virtual electrodes. Front Hum Neurosci 2016; 10: 238.Google Scholar
van Straaten, EC, Scheltens, P, Gouw, AA, Stam, CJ. Eyes-closed task-free electroencephalography in clinical trials for Alzheimer’s disease: an emerging method based upon brain dynamics. Alzheimers Res Ther 2014; 6: 8692.Google Scholar
Gouw, AA, Alsema, AM, Tijms, BM, et al. EEG spectral analysis as a putative early prognostic biomarker in nondemented, amyloid positive subjects. Neurobiol Aging 2017; 57: 133–42.Google Scholar
Rossini, PM, Di Iorio, R, Vecchio, F, et al. Early diagnosis of Alzheimer’s disease: the role of biomarkers including advanced EEG signal analysis. Report from the IFCN-sponsored panel of experts. Clin Neurophysiol 2020; 131: 1287–310.Google Scholar
Scheltens, P, Hallikainen, M, Grimmer, T, et al. Safety, tolerability and efficacy of the glutaminyl cyclase inhibitor PQ912 in Alzheimer’s disease: results of a randomized, double-blind, placebo-controlled Phase 2a study. Alzheimers Res Ther 2018; 10: 107–12.Google Scholar
Briels, CT, Stam, CJ, Scheltens, P, et al. In pursuit of a sensitive EEG functional connectivity outcome measure for clinical trials in Alzheimer’s disease. Clin Neurophysiol 2020; 131: 8895.Google Scholar
Food and Drug Administration. Early Alzheimer’s Disease: Developing Drugs for Treatment. Guidance for Industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER); 2018.Google Scholar
Prinsen, CA, Vohra, S, Rose, MR, et al. How to select outcome measurement instruments for outcomes included in a “core outcome set”: a practical guideline. Trials 2016; 17: 449.Google Scholar
Jutten, RJ, Sikkes, SAM, Amariglio, RE, et al. Identifying sensitive measures of cognitive decline at different clinical stages of Alzheimer’s disease. J Int Neuropsychol Soc 2020; 27: 113.Google Scholar
Folstein, MF, Folstein, SE, McHugh, PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975; 12: 189–98.Google Scholar
Rosen, WG, Mohs, RC, Davis, KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64.Google Scholar
Galasko, D, Bennett, D, Sano, M, et al. An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. Alzheimer Dis Assoc Disord 1997; 11: S33–9.Google Scholar
Guy, W. Clinical Global Impressions. ECDEU Assessment Manual for Psychopharmacology – Revised. Rockville, MD: US Department of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, National Institute of Mental Health, Psychopharmacology Research Branch, Division of Extramural Research Programs; 1976: 218–22.Google Scholar
Williams, MM, Storandt, M, Roe, CM, Morris, JC. Progression of Alzheimer’s disease as measured by Clinical Dementia Rating Sum of Boxes scores. Alzheimers Dement 2013; 9: S3944.Google Scholar
Evans, S, McRae-McKee, K, Wong, MM, et al. The importance of endpoint selection: How effective does a drug need to be for success in a clinical trial of a possible Alzheimer’s disease treatment? Eur J Epidemiol 2018; 33: 635–44.Google Scholar
Donohue, MC, Sperling, RA, Salmon, DP, et al. The Preclinical Alzheimer Cognitive Composite: measuring amyloid-related decline. JAMA Neurol 2014; 71: 961–70.CrossRefGoogle ScholarPubMed
Langbaum, JB, Hendrix, SB, Ayutyanont, N, et al. An empirically derived composite cognitive test score with improved power to track and evaluate treatments for preclinical Alzheimer’s disease. Alzheimers Dement 2014; 10: 666–74.Google Scholar
Wang, J, Logovinsky, V, Hendrix, SB, et al. ADCOMS: a composite clinical outcome for prodromal Alzheimer’s disease trials. J Neurol Neurosurg Psychiatry 2016; 87: 993–9.Google Scholar
Jutten, RJ, Harrison, JE, Brunner, AJ, et al. The cognitive-functional composite is sensitive to clinical progression in early dementia: longitudinal findings from the Catch-Cog study cohort. Alzheimers Dement (N Y) 2020; 6: e12020.Google Scholar
Reimers, S, Stewart, N. Presentation and response timing accuracy in Adobe Flash and HTML5/JavaScript web experiments. Behav Res Methods 2015; 47: 309–27.Google Scholar
Bilder, RM, Reise, SP. Neuropsychological tests of the future: how do we get there from here? Clin Neuropsychologist 2019; 33: 220–45.Google Scholar
Sikkes, SA, Knol, DL, Pijnenburg, YA, et al. Validation of the Amsterdam IADL Questionnaire©, a new tool to measure instrumental activities of daily living in dementia. Neuroepidemiology 2013; 41: 3541.Google Scholar
Gold, M, Amatniek, J, Carrillo, MC, et al. Digital technologies as biomarkers, clinical outcomes assessment, and recruitment tools in Alzheimer’s disease clinical trials. Alzheimers Dement (N Y) 2018; 4: 234–42.Google Scholar
Buckley, RF, Sparks, KP, Papp, KV, et al. Computerized cognitive testing for use in clinical trials: a comparison of the NIH toolbox and Cogstate C3 batteries. J Prev Alzheimers Dis 2017; 4: 311.Google Scholar
Rentz, DM, Dekhtyar, M, Sherman, J, et al. The feasibility of at-home iPad cognitive testing for use in clinical trials. J Prev Alzheimers Dis 2016; 3: 812.Google Scholar
Koo, BM, Vizer, LM. Mobile technology for cognitive assessment of older adults: a scoping review. Innov Aging 2019; 3: igy038.Google Scholar
Sliwinski, MJ, Mogle, JA, Hyun, J, et al. Reliability and validity of ambulatory cognitive assessments. Assessment 2018; 25: 1430.CrossRefGoogle ScholarPubMed
Gray, JA, Fleet, D, Winblad, B. The need for thorough Phase II studies in medicines development for Alzheimer’s disease. Alzheimers Res Ther 2015; 7: 67.Google Scholar
Cummings, J, Morstorf, T, Lee, G. Alzheimer’s drug-development pipeline: 2016. Alzheimers Dement (N Y) 2016; 2: 222–32.Google Scholar
Cole, MA, Seabrook, GR. On the horizon: the value and promise of the global pipeline of Alzheimer’s disease therapeutics. Alzheimers Dement (N Y) 2020; 6: 19.Google Scholar
Sabbagh, M, Hendrix, S, Harrison, JE. FDA position statement “early Alzheimer disease: developing drugs for treatment, guidance for industry”. Alzheimers Dement (N Y) 2019; 5: 1319.Google Scholar

References

Siemers, E. Drug development in AD: point of view from the industry. J Prev Alzheimers Dis 2015; 2: 216–18.Google Scholar
Cummings, J, Ritter, A, Zhong, K. Clinical trials for disease-modifying therapies in Alzheimer’s disease: a primer, lessons learned, and a blueprint for the future. J Alzheimers Dis 2018; 64: S322.CrossRefGoogle Scholar
Rinaldi, A. Setbacks and promises for drugs against Alzheimer’s disease: as pharmaceutical companies are retreating from drug development for Alzheimer’s, new approaches are being tested in academia and biotech companies. EMBO Rep 2018; 19: e46714.CrossRefGoogle ScholarPubMed
Biogen. Biogen plans regulatory filing for aducanumab in Alzheimer’s disease based on new analysis of larger dataset from Phase 3 studies. Investor Relations 2019; Oct. 22. Available at: https://investors.biogen.com/news-releases/news-release-details/biogen-plans-regulatory-filing-aducanumab-alzheimers-disease (accessed January 2021).Google Scholar
Food and Drug Administration. FDA’s decision to approve new treatment for Alzheimer’s disease. Available at: www.fda.gov/drugs/news-events-human-drugs/fdas-decision-approve-new-treatment-alzheimers-disease (accessed June 2021).Google Scholar
Food and Drug Administration. Early Alzheimer’s Disease: Developing Drugs for Treatment Guidance for Industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER); 2018.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 January 2021).Google Scholar
Pharmaceuticals and Medical Devices Agency (PMDA). Project to promote the development of innovative pharmaceuticals, medical devices, and regenerative medical products (Ministry of Health, Labour, and Welfare) regulatory science research for the establishment of criteria for clinical evaluation of drugs for Alzheimer’s disease. Available at: www.pmda.go.jp/files/000221585.pdf (accessed January 2021).Google Scholar
Morant, AV, Vestergaard, HT, Lassen, AB, Navikas, V. US, EU, and Japanese regulatory guidelines for development of drugs for treatment of Alzheimer’s disease: implications for global drug development. Clin Transl Sci 2020; 13: 652–64.Google Scholar
Food and Drug Administration. Expedited programs for serious conditions: drugs and biologics, May 2014. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/expedited-programs-serious-conditions-drugs-and-biologics (accessed June 2021).Google Scholar
Food and Drug Administration. Summary memorandum. Available at: www.accessdata.fda.gov/drugsatfda_docs/nda/2021/Aducanumab_BLA761178_Dunn_2021_06_07.pdf (accessed June 2021).Google Scholar
Centers for Medicare and Medicaid Services. Decision memo for beta amyloid positron emission tomography in dementia and neurodegenerative disease (CAG-00431N). Available at: www.cms.gov/medicare-coverage-database/details/nca-decision-memo.aspx?NCAId=265 (accessed January 2021).Google Scholar

References

Oxford, AE, Stewart, ES, Rohn, TT. Clinical trials in Alzheimer’s disease: a hurdle in the path of remedy. Int J Alzheimers Dis 2020; 2020: 5380346.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug-development pipeline: 2020. Alzheimer Dement (N Y) 2020; 6: e12050.Google Scholar
Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Morris, J. Clinical Dementia Rating: a reliable and valid diagnostic and staging measure for dementia of the Alzheimer type. Int Psychogeriatr 1997; 9: 173–6.Google Scholar
Schneider, LS, Olin, JT, Doody, RS, et al. Validity and reliability of the Alzheimer’s Disease Cooperative Study–Clinical Global Impression of Change. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: S2232.Google Scholar
Reisberg, B, Ferris, SH, de Leon, MJ, Crook, T. The global deterioration scale for assessment of primary degenerative dementia. Am J Psychiatry 1982; 139: 1135–9.Google Scholar
Rosen, WG, Mohs, RC, Davis, KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64.Google Scholar
Monsch, AU, Bondi, MW, Salmon, DP, et al. Clinical validity of the Mattis Dementia Rating Scale in detecting dementia of the Alzheimer type. A double cross-validation and application to a community-dwelling sample. Arch Neurol 1995; 52: 899904.Google Scholar
Panisset, M, Roudier, M, Saxton, J, Boller, F. Severe Impairment Battery. A neuropsychological test for severely demented patients. Arch Neurol 1994; 51: 41–5.Google Scholar

References

Summers, WK, Majovski, LV, Marsh, GM, Tachiki, K, Kling, A. Oral tetrahydroaminoacridine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med 1986; 315: 1241–5.Google Scholar
Davis, KL, Thal, LJ, Gamzu, ER, et al. A double-blind, placebo-controlled multicenter study of tacrine for Alzheimer’s disease. The Tacrine Collaborative Study Group. N Engl J Med 1992; 327: 1253–9.Google Scholar
Rosen, WG, Mohs, RC, Davis, KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64.Google Scholar
Thal, LJ. The Alzheimer’s Disease Cooperative Study in 2004. Alzheimer Dis Assoc Disord 2004; 18: 183–5.Google Scholar
Sano, M, Ernesto, C, Thomas, RG, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. N Engl J Med 1997; 336: 1216–22.Google Scholar
Aisen, PS, Schneider, LS, Sano, M, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA 2008; 300: 1774–83.Google Scholar
Galasko, DR, Peskind, E, Clark, CM, et al. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol 2012; 69: 836–41.Google Scholar
Petersen, RC, Thomas, RG, Grundman, M, et al. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med 2005; 352: 2379–88.Google Scholar
Aisen, PS, Davis, KL, Berg, JD, et al. A randomized controlled trial of prednisone in Alzheimer’s disease. Alzheimer’s Disease Cooperative Study. Neurology 2000; 54: 588–93.Google Scholar
Aisen, PS, Schafer, KA, Grundman, M, et al. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA 2003; 289: 2819–26.Google Scholar
Mulnard, RA, Cotman, CW, Kawas, C, et al. Estrogen replacement therapy for treatment of mild to moderate Alzheimer disease: a randomized controlled trial. Alzheimer’s Disease Cooperative Study. JAMA 2000; 283: 1007–15.Google Scholar
Sano, M, Bell, KL, Galasko, D, et al. A randomized, double-blind, placebo-controlled trial of simvastatin to treat Alzheimer disease. Neurology 2011; 77: 556–63.CrossRefGoogle ScholarPubMed
Teri, L, Logsdon, RG, Peskind, E, et al. Treatment of agitation in AD: a randomized, placebo-controlled clinical trial. Neurology 2000; 55: 1271–8.Google Scholar
Grundman, M, Farlow, M, Peavy, G, et al. A phase I study of AIT-082 in healthy elderly volunteers. J Mol Neurosci 2002; 18: 283–93.Google Scholar
Petersen, RC, Aisen, PS, Beckett, LA, et al. Alzheimer’s Disease Neuroimaging Initiative (ADNI): clinical characterization. Neurology 2010; 74: 201–9.Google Scholar
Weninger, S, Carrillo, MC, Dunn, B, et al. Collaboration for Alzheimer’s Prevention: principles to guide data and sample sharing in preclinical Alzheimer’s disease trials. Alzheimers Dement 2016; 12: 631–2.Google Scholar
Vellas, B, Carrillo, MC, Sampaio, C, et al. Designing drug trials for Alzheimer’s disease: what we have learned from the release of the Phase III antibody trials: a report from the EU/US/CTAD Task Force. Alzheimers Dement 2013; 9: 438–44.Google Scholar
Sperling, RA, Donohue, MC, Raman, R, et al. Association of factors with elevated amyloid burden in clinically normal older individuals. JAMA Neurol 2020; 77: 735–45.Google Scholar
Sperling, RA, Rentz, DM, Johnson, KA, et al. The A4 study: stopping AD before symptoms begin? Sci Transl Med 2014; 6: 228fs13.CrossRefGoogle ScholarPubMed
Donohue, MC, Sperling, RA, Petersen, R, et al. Association between elevated brain amyloid and subsequent cognitive decline among cognitively normal persons. JAMA 2017; 317: 2305–16.Google Scholar
Donohue, MC, Sperling, RA, Salmon, DP, et al. The Preclinical Alzheimer Cognitive Composite: measuring amyloid-related decline. JAMA Neurol 2014; 71: 961–70.Google Scholar
Siemers, ER, Sundell, KL, Carlson, C, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer’s disease patients. Alzheimers Dement 2016; 12: 110–20.Google Scholar
Doody, RS, Thomas, RG, Farlow, M, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 2014; 370: 311–21.Google Scholar
Weiner, MW, Harvey, D, Hayes, J, et al. Effects of traumatic brain injury and posttraumatic stress disorder on development of Alzheimer’s disease in Vietnam Veterans using the Alzheimer’s Disease Neuroimaging Initiative: preliminary report. Alzheimers Dement (N Y) 2017; 3: 177–88.Google Scholar
Craft, S, Raman, R, Chow, TW, et al. Safety, efficacy, and feasibility of intranasal insulin for the treatment of mild cognitive impairment and alzheimer disease dementia: a randomized clinical trial. JAMA Neurol 2020; 77: 1099–109.Google Scholar
Kaufman, AC, Salazar, SV, Haas, LT, et al. Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol 2015; 77: 953–71.Google Scholar
van Dyck, CH, Nygaard, HB, Chen, K, et al. Effect of AZD0530 on cerebral metabolic decline in Alzheimer disease: a randomized clinical trial. JAMA Neurol 2019; 76: 1219–29.Google Scholar
Newhouse, P, Kellar, K, Aisen, P, et al. Nicotine treatment of mild cognitive impairment: a 6-month double-blind pilot clinical trial. Neurology 2012; 78: 91101.Google Scholar
Henley, D, Raghavan, N, Sperling, R, et al. Preliminary results of a trial of atabecestat in preclinical Alzheimer’s disease. N Engl J Med 2019; 380: 1483–5.Google Scholar
Egan, MF, Kost, J, Voss, T, et al. Randomized trial of verubecestat for prodromal Alzheimer’s disease. N Engl J Med 2019; 380: 1408–20.Google Scholar
Aisen, PS, Sperling, RA, Cummings, J, et al. The Trial-Ready Cohort for Preclinical/Prodromal Alzheimer’s Disease (TRC-PAD) project: an overview. J Prev Alzheimers Dis 2020; 7: 208–12.Google Scholar
Galasko, D, Bennett, DA, Sano, M, et al. ADCS Prevention Instrument Project: assessment of instrumental activities of daily living for community-dwelling elderly individuals in dementia prevention clinical trials. Alzheimer Dis Assoc Disord 2006; 20: S152–69.Google Scholar
Cummings, JL, Raman, R, Ernstrom, K, Salmon, D, Ferris, SH, Alzheimer’s Disease Cooperative Study Group. ADCS Prevention Instrument Project: behavioral measures in primary prevention trials. Alzheimer Dis Assoc Disord 2006; 20: S147–51.Google Scholar
Schneider, LS, Clark, CM, Doody, R, et al. ADCS Prevention Instrument Project: ADCS-clinicians’ global impression of change scales (ADCS–CGIC), self-rated and study partner-rated versions. Alzheimer Dis Assoc Disord 2006; 20: S124–38.Google Scholar
Ferris, SH, Aisen, PS, Cummings, J, et al. ADCS Prevention Instrument Project: overview and initial results. Alzheimer Dis Assoc Disord 2006; 20: S109–23.Google Scholar
Logsdon, RG, Teri, L, Weiner, MF, et al. Assessment of agitation in Alzheimer’s disease: the agitated behavior in dementia scale. Alzheimer’s Disease Cooperative Study. J Am Geriatr Soc 1999; 47: 1354–8.Google Scholar
Morris, JC, Ernesto, C, Schafer, K, et al. Clinical dementia rating training and reliability in multicenter studies: the Alzheimer’s Disease Cooperative Study experience. Neurology 1997; 48: 1508–10.Google Scholar
Schmitt, FA, Ashford, W, Ernesto, C, et al. The severe impairment battery: concurrent validity and the assessment of longitudinal change in Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: S51–6.Google Scholar
Galasko, D, Bennett, D, Sano, M, et al. An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: S33–9.Google Scholar
Schneider, LS, Olin, JT, Doody, RS, et al. Validity and reliability of the Alzheimer’s Disease Cooperative Study–Clinical Global Impression of Change. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: S2232.CrossRefGoogle ScholarPubMed
Ferris, SH, Mackell, JA, Mohs, R, et al. A multicenter evaluation of new treatment efficacy instruments for Alzheimer’s disease clinical trials: overview and general results. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: S112.Google Scholar
Walsh, SP, Raman, R, Jones, KB, Aisen, PS, Alzheimer’s Disease Cooperative Study Group. ADCS Prevention Instrument Project: the Mail-In Cognitive Function Screening Instrument (MCFSI). Alzheimer Dis Assoc Disord 2006; 20: S170–8.Google Scholar
Amariglio, RE, Donohue, MC, Marshall, GA, et al. Tracking early decline in cognitive function in older individuals at risk for Alzheimer disease dementia: the Alzheimer’s Disease Cooperative Study Cognitive Function Instrument. JAMA Neurol 2015; 72: 446–54.Google Scholar
Aisen, PS, Andrieu, S, Sampaio, C, et al. Report of the task force on designing clinical trials in early (predementia) AD. Neurology 2011; 76: 280–6.Google Scholar
Coley, N, Raman, R, Donohue, MC, et al. Defining the optimal target population for trials of polyunsaturated fatty acid supplementation using the erythrocyte omega-3 index: a step towards personalized prevention of cognitive decline? J Nutr Health Aging 2018; 22: 982–98.Google Scholar
Donohue, MC, Sun, CK, Raman, R, et al. Cross-validation of optimized composites for preclinical Alzheimer’s disease. Alzheimers Dement (N Y) 2017; 3: 123–9.Google Scholar
Langford, O, Raman, R, Sperling, RA, et al. Predicting amyloid burden to accelerate recruitment of secondary prevention clinical trials. J Prev Alzheimers Dis 2020; 7: 213–18.Google Scholar
Papp, KV, Rentz, DM, Maruff, P, et al. The Computerized Cognitive Composite (C3) in an Alzheimer’s disease secondary prevention trial. J Prev Alzheimers Dis 2021; 8: 5967.Google Scholar
Sano, M, Raman, R, Emond, J, et al. Adding delayed recall to the Alzheimer Disease Assessment Scale is useful in studies of mild cognitive impairment but not Alzheimer disease. Alzheimer Dis Assoc Disord 2011; 25: 122–7.Google Scholar
Li, D, Iddi, S, Thompson, WK, et al. Bayesian latent time joint mixed-effects model of progression in the Alzheimer’s Disease Neuroimaging Initiative. Alzheimers Dement (Amst) 2018; 10: 657–68.Google Scholar
Iddi, S, Li, D, Aisen, PS, et al. Estimating the evolution of disease in the Parkinson’s progression markers initiative. Neurodegener Dis 2018; 18: 173–90.Google Scholar
Li, D, Donohue, MC. Disease progression models for dominantly-inherited Alzheimer’s disease. Brain 2018; 141: 1244–6.Google Scholar
Donohue, MC, Jacqmin-Gadda, H, Le Goff, M, et al. Estimating long-term multivariate progression from short-term data. Alzheimers Dement 2014; 10: S400–10.Google Scholar
Donohue, MC, Aisen, PS. Mixed model of repeated measures versus slope models in Alzheimer’s disease clinical trials. J Nutr Health Aging 2012; 16: 360–4.Google Scholar
Donohue, MC, Gamst, AC, Thomas, RG, et al. The relative efficiency of time-to-threshold and rate of change in longitudinal data. Contemp Clin Trials 2011; 32: 685–93.Google Scholar
Li, D, Iddi, S, Aisen, PS, Thompson, WK, Donohue, MC. The relative efficiency of time-to-progression and continuous measures of cognition in presymptomatic Alzheimer’s disease. Alzheimers Dement (N Y) 2019; 5: 308–18.Google Scholar
Nicoll, JA, Wilkinson, D, Holmes, C. Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med 2003; 9: 448–52.Google Scholar
Satlin, A, Wang, J, Logovinsky, V, et al. Design of a Bayesian adaptive phase 2 proof-of-concept trial for BAN2401, a putative disease-modifying monoclonal antibody for the treatment of Alzheimer’s disease. Alzheimers Dement (N Y) 2016; 2: 112.Google Scholar
Rafii, MS, Zaman, S, Handen, BL. Integrating biomarker outcomes into clinical trials for Alzheimer’s disease in Down syndrome. J Prev Alzheimers Dis 2021; 8: 4851.Google Scholar
Handen, BL, Lott, IT, Christian, BT, et al. The Alzheimer’s Biomarker Consortium – Down syndrome: rationale and methodology. Alzheimers Dement (Amst) 2020; 12: e12065.Google Scholar
Ovod, V, Ramsey, KN, Mawuenyega, KG, et al. Amyloid beta concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement 2017; 13: 841–9.Google Scholar
Nakamura, A, Kaneko, N, Villemagne, VL, et al. High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature 2018; 554: 249–54.Google Scholar
Janelidze, S, Berron, D, Smith, R, et al. Associations of plasma phospho-tau217 levels with tau positron emission tomography in early Alzheimer Disease. JAMA Neurol 2021; 78: 149–56.Google Scholar

References

Ritchie, CW, Molinuevo, JL, Satlin, A, et al. The European Prevention of Alzheimer’s Dementia (EPAD) Consortium: a platform to enable the secondary prevention of Alzheimer’s dementia through improved proof of concept trials. Lancet Psychiatry 2016; 3: 179–86.Google Scholar
Gregory, S, Wells, K, Forsyth, K, et al. Research participants as collaborators: background, experience and policies from the PREVENT Dementia and EPAD programmes. Dementia 2018; 17: 1045–54.Google Scholar
Ritchie, K, Ritchie, CW. The PREVENT study: a prospective cohort study to identify mid-life biomarkers of late-onset Alzheimer’s disease. BMJ Open 2012; 2: e001893.Google Scholar
Vermunt, L, Veal, CD, Ter Meulen, L, et al. European Prevention of Alzheimer’s Dementia Registry: recruitment and prescreening approach for a longitudinal cohort and prevention trials. Alzheimers Dement 2018; 14: 837–42.Google Scholar
Solomon, A, Kivipelto, M, Molinuevoet, JL, al. European Prevention of Alzheimer’s Dementia Longitudinal Cohort Study (EPAD LCS): study protocol. BMJ Open 2018; 8: e021017.Google Scholar
ten Kate, M, Ingala, S, Schwarz, A, et al. Secondary prevention of Alzheimer’s dementia: neuroimaging contributions. Alzheimers Res Ther 2018; 10: 112.Google Scholar
Ritchie, K, Ropacki, M, Albala, B, et al. Recommended cognitive outcomes in preclinical Alzheimer’s disease: consensus statement from the European Prevention of Alzheimer’s Dementia project. Alzheimers Dement 2017; 13: 186–95.Google Scholar
Mortamais, M, Ash, J, Harrison, J, et al. Detecting cognitive changes in preclinical Alzheimer’s disease: a review of its feasibility. Alzheimers Dement 2017; 13: 468–92.Google Scholar
Ritchie, K, Carriere, I, Howett, D, et al. Allocentric and egocentric spatial processing in middle-aged adults at high risk of late-onset Alzheimer’s disease: the PREVENT Dementia Study. J Alzheimers Dis 2018; 65: 885–96.Google Scholar
Ritchie, CW, Muniz-Terrera, G, Kivipelto, M, et al. The European Prevention of Alzheimer’s Dementia (EPAD) longitudinal cohort study: baseline data release V500.0. J Prev Alzheimers Dis 2019; 7: 813.Google Scholar
Ingala, S, De Boer, C, Masselink, LA, the EPAD consortium. Application of the ATN classification scheme in a population without dementia: findings from the EPAD cohort. Alzheimers Dement 2021;DOI: http://doi.org/10.1002/alz.12292.CrossRefGoogle Scholar
Angus, D, Alexander, B, Berry, S, et al. Adaptive platform trials: definition, design, conduct, and reporting considerations. Nat Rev Drug Discov 2019; 18: 797807.Google Scholar
Frisoni, G, Ritchie, C, Carrera, E, et al. Re-aligning scientific and lay narratives of Alzheimer’s disease. Lancet Neurol 2019; 18: 918–19.Google Scholar
Frisoni, GB, Molinuevo, JL, Altomare, D, et al. Precision prevention of Alzheimer’s and other dementias: anticipating future needs in the control of risk factors and implementation of disease-modifying therapies. Alzheimers Dement 2020; 16: 1457–68.Google Scholar
Ritchie, CW, Russ, TC, Banerjee, S, et al. The Edinburgh Consensus: preparing for the advent of disease-modifying therapies for Alzheimer’s disease. Alzheimers Res Ther 2017; 9: 85.Google Scholar
Jack, CR Jr., Bennett, DA, Blennow, K, et al. NIA–AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14: 535–62.Google Scholar

References

Vradenburg, G. Catalyzing the Landmark G8 Commitment. HuffPost 2013; December 23. Available at: www.huffpost.com/entry/catalyzing-the-landmark-g8-commitment_b_4489405 (accessed November 24, 2020).Google Scholar
Mathias, A, Lamberti, MJ, Getz, K. ‘START’ (Start-up Time And Readiness Tracking) study: Working Group final report. Tufts Center for the Study of Drug Development (CSDD). Released June 15, 2012.Google Scholar
Amjad, H, Roth, DL, Sheehan, OC, et al. Underdiagnosis of dementia: an observational study of patterns in diagnosis and awareness in US older adults. J Gen Intern Med 2018; 33: 1131–8.Google Scholar
Carr, SA, Davis, R, Spencer, D, et al. Comparison of recruitment efforts targeted at primary care physicians versus the community at large for participation in Alzheimer disease clinical trials. Alzheimer Dis Assoc Disord 2010; 24: 165–70.Google Scholar
Research!America. Public perception of clinical trials. July 2017. Available at: www.researchamerica.org/sites/default/files/July2017ClinicalResearchSurveyPressReleaseDeck_0.pdf (accessed November 24, 2020).Google Scholar
Murphy-Barron, C, Pyenson, B, Ferro, C, Emery, M. Comparing the demographics of enrollees in Medicare Advantage and fee-for-service medicare. Milliman report. October 2020. Available at: www.bettermedicarealliance.org/publication/comparing-the-demographics-of-enrollees-in-medicare-advantage-and-fee-for-service-medicare/ (accessed November 24, 2020).Google Scholar
Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Shin, J, Doraiswamy, PM. Underrepresentation of African-Americans in Alzheimer’s trials: a call for affirmative action. Front Aging Neurosci 2016; 8: 123.Google Scholar
National Institutes of Health. NIH RCDC inclusion statistics report. Available at: https://report.nih.gov/RISR/#/ (accessed November 24, 2020).Google Scholar
United States Census Bureau. Quick facts United States. 2020. Available at: www.census.gov/quickfacts/fact/table/US/PST045219 (accessed November 24, 2020).Google Scholar
US Food and Drug Administration. Qualification process for drug development tools guidance for industry and FDA staff draft guidance. November 2020. Available at: www.fda.gov/media/133511/download (accessed November 24, 2020).Google Scholar
Clark, CM, Pontecorvo, MJ, Beach, TB, et al. Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-β plaques: a prospective cohort study. Lancet Neurol 2012; 11: 669–78.Google Scholar
Schwarz, AJ, Yu, P, Miller, BB, et al. Regional profiles of the candidate tau PET ligand 18F-AV-1451 recapitulate key features of Braak histopathological stages. Brain 2016; 139: 1539–50.Google Scholar
Jack, CR, 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
Nakamura, A, Kaneko, N, Villemagne, VL, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 2018; 554: 249–54.Google Scholar
Palmqvist, S, Janelidze, S, Quiroz, YT, et al. Discriminative accuracy of plasma phosphor-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 2020; 324: 772–81.Google Scholar
Morgan, AR, Touchard, S, Leckey, C, et al. Inflammatory biomarkers in Alzheimer’s disease plasma. Alzheimers Dement 2019; 15: 776–87.Google Scholar
Welsh, KA, Butters, N, Mohs, RC, et al. The Consortium to Establish a Registry for Alzheimer’s Disease (CERAD). Part V. A normative study of the neuropsychological battery. Neurology 1994; 44: 609–14.Google Scholar
Kourtis, LC, Regele, OB, Wright, JM, Jones, GB. Digital biomarkers for Alzheimer’s disease: the mobile wearable devices opportunity. NPJ Digit Med 2019; 2: 19.Google Scholar
McKhann, GM, Knopman, DS, Chertkow, H, et al. The diagnosis of dementia due to 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: 263–9.Google Scholar
Sperling, RA, Donohue, MC, Raman, R, et al. Association of factors with elevated amyloid burden in clinically normal older individuals. JAMA Neurol 2020; 77: 735–45.Google Scholar
Ritchie, CW, Molinuevo, JL, Truyen, L, et al. Development of interventions for the secondary prevention of Alzheimer’s dementia: the European Prevention of Alzheimer’s Dementia (EPAD) project. Lancet Psychiatry 2016; 3: 179–86.Google Scholar

References

Onyike, CU, Diehl-Schmid, J. The epidemiology of frontotemporal dementia. Int Rev Psychiatry 2013; 25: 130–7.Google Scholar
Perry, DC, Brown, JA, Possin, KL, et al. Clinicopathological correlations in behavioural variant frontotemporal dementia. Brain 2017; 140: 3329–45.Google Scholar
Armstrong, MJ, Litvan, I, Lang, AE, et al. Criteria for the diagnosis of corticobasal degeneration. Neurology 2013; 80: 496503.Google Scholar
Höglinger, GU, Respondek, G, Stamelou, M, et al. Clinical diagnosis of progressive supranuclear palsy: the movement disorder society criteria. Mov Disord 2017; 32: 853–64.Google Scholar
Rascovsky, K, Hodges, JR, Knopman, D, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain 2011; 134: 2456–77.Google Scholar
Gorno-Tempini, M, Hillis, A, Weintraub, S, et al. Classification of primary progressive aphasia and its variants. Neurology 2011; 76: 1006–14.Google Scholar
Jadhav, S, Avila, J, Schöll, M, et al. A walk through tau therapeutic strategies. Acta Neuropathol Commun 2019; 7: 22.Google Scholar
Weingarten, MD, Lockwood, AH, Hwo, SY, Kirschner, MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci USA 1975; 72: 1858–62.Google Scholar
Mackenzie, IRA, Neumann, M, Baborie, A, et al. A harmonized classification system for FTLD-TDP pathology. Acta Neuropathol 2011; 122: 111–13.Google Scholar
Svetoni, F, Frisone, P, Paronetto, MP. Role of FET proteins in neurodegenerative disorders. RNA Biol 2016; 13: 1089–102.Google Scholar
Spinelli, EG, Mandelli, ML, Miller, ZA, et al. Typical and atypical pathology in primary progressive aphasia variants. Ann Neurol 2017; 81: 430–43.Google Scholar
Boxer, AL, Lang, AE, Grossman, M, et al. Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled Phase 2/3 trial. Lancet Neurol 2014; 13: 676–85.Google Scholar
Forrest, SL, Kril, JJ, Stevens, CH, et al. Retiring the term FTDP-17 as MAPT mutations are genetic forms of sporadic frontotemporal tauopathies. Brain 2018; 141: 521–34.Google Scholar
Lee, SE, Rabinovici, GD, Mayo, MC, et al. Clinicopathological correlations in corticobasal degeneration. Ann Neurol 2011; 70: 327–40.Google Scholar
Haynes, BA, Rhinn, H, Yeh, FL, et al. AL001 restores CSF PGRN levels and normalizes disease-associated biomarkers in individuals with frontotemporal dementia due to heterozygous mutations in the progranulin gene. Alzheimers Dement 2020; 16: e046114.Google Scholar
Boxer, AL, Moebius, HJ, Harris, B, et al. Phase 2a randomized, double-blind, placebo-controlled trial of the histone deacetylase inhibitor (HDACi), FRM-0334, in asymptomatic carriers of, or patients with frontotemporal lobar degeneration (FTLD) due to progranulin gene mutations. Alzheimer’s Association International Conference, Los Angeles, CA, 2019.Google Scholar
Sha, SJ, Miller, ZA, won, Min S, et al. An 8-week, open-label, dose-finding study of nimodipine for the treatment of progranulin insufficiency from GRN gene mutations. Alzheimers Dement (N Y) 2017; 3: 507–12.Google Scholar
The Angel Fund for ALS Research. Research at the day lab. Available at: https://theangelfund.org/research-at-day-lab/ (accessed December 28, 2020).Google Scholar
Boxer, A, Ljubenkov, P, VandeVrede, L, et al. A Phase 1b, randomized, double-blind, placebo-controlled, parallel cohort safety, tolerability, pharmacokinetics, pharmacodynamics and preliminary efficacy study of intravenously infused BIIB092 in patients with four different tauopathy syndromes. 13th Clinical Trials on Alzheimer’s Disease (CTAD) Congress, November 4–7, 2020.Google Scholar
Boxer, AL, Gold, M, Feldman, H, et al. New directions in clinical trials for frontotemporal lobar degeneration: methods and outcome measures. Alzheimers Dement 2020; 16: 131–43.Google Scholar
ALLFTD. History. Available at: www.allftd.org/history (accessed December 30, 2020).Google Scholar
Richardson, E, Burnell, J, Adams, HR, et al. Developing and implementing performance outcome assessments: evidentiary, methodologic, and operational considerations. Ther Innov Regul Sci 2019; 53: 146–53.Google Scholar
Gaasterland, CMW, Jansen-Van Der Weide, MC, Weinreich, SS, Van Der Lee, JH . A systematic review to investigate the measurement properties of goal attainment scaling, towards use in drug trials. BMC Med Res Methodol 2016; 16: 99.Google Scholar
Guinart, D, de Filippis, R, Rosson, S, et al. Development and validation of a computerized adaptive assessment tool for discrimination and measurement of psychotic symptoms. Schizophr Bull 2021; 47: 644–52.Google Scholar
Mioshi, E, Hsieh, S, Savage, S, Hornberger, M, Hodges, JR. Clinical staging and disease progression in frontotemporal dementia. Neurology 2010; 74: 1591–7.Google Scholar
Pfeffer, RI, Kurosaki, TT, Harrah, CH, Chance, JM, Filos, S. Measurement of functional activities in older adults in the community. J Gerontol 1982; 37: 323–9.Google Scholar
Miyagawa, T, Brushaber, D, Syrjanen, J, et al. Use of the CDR® plus NACC FTLD in mild FTLD: data from the ARTFL/LEFFTDS consortium. Alzheimers Dement 2020; 16: 7990.Google Scholar
Boeve, B, Rosen, H, Boxer, A, et al. The Multidomain Impairment Rating (MIR) Scale: initial reliability data on a multidimensional scale for FTLD (P5.1–010). Neurology 2019; 92;https://n.neurology.org/content/92/15_Supplement/P5.1-010.abstract.Google Scholar
Staffaroni, AM, Ljubenkov, PA, Kornak, J, et al. Longitudinal multimodal imaging and clinical endpoints for frontotemporal dementia clinical trials. Brain 2019; 142: 443–59.Google Scholar
Toller, G, Ranasinghe, K, Cobigo, Y, et al. Revised Self-Monitoring Scale: A potential endpoint for frontotemporal dementia clinical trials. Neurology 2020; 94: e238495.Google Scholar
Staffaroni, AM, Bajorek, L, Casaletto, KB, et al. Assessment of executive function declines in presymptomatic and mildly symptomatic familial frontotemporal dementia: NIH-EXAMINER as a potential clinical trial endpoint. Alzheimers Dement 2020; 16: 1121.Google Scholar
FDA. Table of surrogate endpoints that were the basis of drug approval or licensure. www.fda.gov/drugs/development-resources/table-surrogate-endpoints-were-basis-drug-approval-or-licensure (accessed December 30, 2020).Google Scholar
Swift, IJ, Sogorb-Esteve, A, Heller, C, et al. Fluid biomarkers in frontotemporal dementia: past, present and future. J Neurol Neurosurg Psychiatry 2021; 92: 204–15.Google Scholar
Palmqvist, S, Janelidze, S, Quiroz, YT, et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 2020; 324: 772–81.Google Scholar
Thijssen, EH, La Joie, R, Wolf, A, et al. Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration. Nat Med 2020; 26: 387–97.Google Scholar
Paterson, RW, Slattery, CF, Poole, T, et al. Cerebrospinal fluid in the differential diagnosis of Alzheimer’s disease: clinical utility of an extended panel of biomarkers in a specialist cognitive clinic. Alzheimers Res Ther 2018; 10: 32.Google Scholar
Meeter, LHH, Vijverberg, EG, Del Campo, M, et al. Clinical value of neurofilament and phospho-tau/tau ratio in the frontotemporal dementia spectrum. Neurology 2018; 90: e1231–9.Google Scholar
Rojas, JC, Karydas, A, Bang, J, et al. Plasma neurofilament light chain predicts progression in progressive supranuclear palsy. Ann Clin Transl Neurol 2016; 3: 216–25.Google Scholar
Ljubenkov, PA, Staffaroni, AM, Rojas, JC, et al. Cerebrospinal fluid biomarkers predict frontotemporal dementia trajectory. Ann Clin Transl Neurol 2018; 5: 1250–63.Google Scholar
Meeter, LH, Dopper, EG, Jiskoot, LC, et al. Neurofilament light chain: a biomarker for genetic frontotemporal dementia. Ann Clin Transl Neurol 2016; 3: 623–36.Google Scholar
Gass, J, Cannon, A, Mackenzie, IR, et al. Mutations in progranulin are a major cause of ubiquitin-positive frontotemporal lobar degeneration. Hum Mol Genet 2006; 15: 29883001.Google Scholar
Baker, M, Mackenzie, IR, Pickering-Brown, SM, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006; 442: 916–19.Google Scholar
Shankaran, SS, Capell, A, Hruscha, AT, et al. Missense mutations in the progranulin gene linked to frontotemporal lobar degeneration with ubiquitin-immunoreactive inclusions reduce progranulin production and secretion. J Biol Chem 2008; 283: 1744–53.Google Scholar
Kao, AW, McKay, A, Singh, PP, Brunet, A, Huang, EJ. Progranulin, lysosomal regulation and neurodegenerative disease. Nat Rev Neurosci 2017; 18: 325–33.Google Scholar
Meeter, LHH, Patzke, H, Loewen, G, et al. Progranulin levels in plasma and cerebrospinal fluid in granulin mutation carriers. Dement Geriatr Cogn Dis Extra 2016; 6: 330–40.Google Scholar
Finch, N, Baker, M, Crook, R, et al. Plasma progranulin levels predict progranulin mutation status in frontotemporal dementia patients and asymptomatic family members. Brain 2009; 132: 583–91.Google Scholar
Ash, PEA, Bieniek, KF, Gendron, TF, et al. Unconventional translation of C9orf72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 2013; 77: 639–46.Google Scholar
Mori, K, Arzberger, T, Grässer, FA, et al. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol 2013; 126: 881–93.Google Scholar
Gendron, TF, Chew, J, Stankowski, JN, et al. Poly(GP) proteins are a useful pharmacodynamic marker for C9orf72-associated amyotrophic lateral sclerosis. Sci Transl Med 2017; 9: eaai7866.Google Scholar
Cammack, AJ, Atassi, N, Hyman, T, et al. Prospective natural history study of C9orf72 ALS clinical characteristics and biomarkers. Neurology 2019; 93: E1605–17.Google Scholar
Lee, SE, Sias, AC, Kosik, EL, et al. Thalamo-cortical network hyperconnectivity in preclinical progranulin mutation carriers. Neuroimage Clin 2019; 22;DOI: https://doi.10.1016/j.nicl.2019.101751.Google Scholar
Jacova, C, Hsiung, GYR, Tawankanjanachot, I, et al. Anterior brain glucose hypometabolism predates dementia in progranulin mutation carriers. Neurology 2013; 81: 1322–31.Google Scholar
Binney, RJ, Pankov, A, Marx, G, et al. Data-driven regions of interest for longitudinal change in three variants of frontotemporal lobar degeneration. Brain Behav 2017; 7: e00675.Google Scholar
Staffaroni, AM, Cobigo, Y, Goh, S-YM, et al. Individualized atrophy scores predict dementia onset in familial frontotemporal lobar degeneration. Alzheimers Dement 2020; 16: 3748.Google Scholar
Cenik, B, Sephton, CF, Dewey, CM, et al. Suberoylanilide hydroxamic acid (vorinostat) up-regulates progranulin transcription: rational therapeutic approach to frontotemporal dementia. J Biol Chem 2011; 286: 16101–8.Google Scholar
Lee, WC, Almeida, S, Prudencio, M, et al. Targeted manipulation of the sortilin–progranulin axis rescues progranulin haploinsufficiency. Hum Mol Genet 2014; 23: 1467–78.Google Scholar
Paul, R. AL001 Phase 1b/2 update. Alzheimer’s Association International Conference, July 26–30, 2020.Google Scholar
Alector Inc. Alector showcases progress in immuno-neurology clinical programs and research portfolio at R&D day. Available at: www.globenewswire.com/news-release/2019/12/13/1960338/0/en/Alector-Showcases-Progress-in-Immuno-Neurology-Clinical-Programs-and-Research-Portfolio-at-R-D-Day.html (accessed February 17, 2020).Google Scholar
Passage Bio. Pipeline: frontotemporal dementia. Available at: www.passagebio.com/pipeline/frontotemporal-dementia/default.aspx. (accessed June 20, 2020).Google Scholar
Prevail Therapeutics. Prevail Therapeutics announces first patient dosed in Phase 1/2 PROCLAIM clinical trial evaluating PR006 for the treatment of frontotemporal dementia patients with GRN mutations. Available at: www.globenewswire.com/news-release/2020/12/11/2143673/0/en/Prevail-Therapeutics-Announces-First-Patient-Dosed-in-Phase-1-2-PROCLAIM-Clinical-Trial-Evaluating-PR006-for-the-Treatment-of-Frontotemporal-Dementia-Patients-with-GRN-Mutations.html (accessed December 27, 2020).Google Scholar
Denali. Our pipeline. Available at:https://denalitherapeutics.com/pipeline (accessed June 20, 2020).Google Scholar
Alzprotect. AZP2006: a mechanism of action with multiple effects, a unique solution for neurodegeneration. Available at: www.alzprotect.com/en/pipeline/azp2006?view=article&layout=alzprotect:hero-article (accessed December 30, 2020).Google Scholar
Taylor, JP, Brown, RH, Cleveland, DW. Decoding ALS: from genes to mechanism. Nature 2016; 539: 197206.Google Scholar
DeVos, SL, Miller, TM. Antisense oligonucleotides: treating neurodegeneration at the level of RNA. Neurotherapeutics 2013; 10: 486–97.Google Scholar
Lim, KRQ, Maruyama, R, Yokota, T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther 2017; 11: 533–45.Google Scholar
Hoy, SM. Nusinersen: first global approval. Drugs 2017; 77: 473–9.Google Scholar
Miller, T, Cudkowicz, M, Shaw, PJ, et al. Phase 1–2 trial of antisense oligonucleotide tofersen for SOD1 ALS. N Engl J Med 2020; 383: 109–19.Google Scholar
Jiang, J, Zhu, Q, Gendron, TF, et al. Gain of toxicity from ALS/FTD-linked repeat expansions in C9orf72 is alleviated by antisense oligonucleotides targeting GGGGCC-containing RNAs. Neuron 2016; 90: 535–50.Google Scholar
Sanders, DW, Kaufman, SK, DeVos, SL, et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 2014; 82: 1271–88.Google Scholar
Yanamandra, K, Kfoury, N, Jiang, H, 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.Google Scholar
Zhou, Y, Shi, J, Chu, D, et al. Relevance of phosphorylation and truncation of tau to the etiopathogenesis of Alzheimer’s disease. Front Aging Neurosci 2018; 10: 27.Google Scholar
CurePSP. UCB pauses the development of bepranemab for progressive supranuclear palsy. Available at: www.psp.org/ucb-shift-2020/ (accessed December 28, 2020).Google Scholar
Novak, P, Schmidt, R, Kontsekova, E, et al. FUNDAMANT: an interventional 72-week phase 1 follow-up study of AADvac1, an active immunotherapy against tau protein pathology in Alzheimer’s disease. Alzheimers Res Ther 2018; 10: 108.Google Scholar
Hung, S-Y, Fu, W-M. Drug candidates in clinical trials for Alzheimer’s disease. J Biomed Sci 2017; 24: 47.Google Scholar
Tsai, RM, Miller, Z, Koestler, M, et al. Reactions to multiple ascending doses of the microtubule stabilizer TPI-287 in patients with Alzheimer disease, progressive supranuclear palsy, and corticobasal syndrome: a randomized clinical trial. JAMA Neurol 2020; 77: 215–24.Google Scholar
Mignon, L, Kordasiewicz, H, Lane, R, et al. Design of the first-in-human study of IONIS-MAPTRx, a tau-lowering antisense oligonucleotide, in patients with Alzheimer disease (S2.006). Neurology 2018; 90.Google Scholar
Rodriguez-Martin, T, Anthony, K, Garcia-Blanco, MA, Mansfield, SG, Anderton, BH, Gallo, JM. Correction of tau mis-splicing caused by FTDP-17 MAPT mutations by spliceosome-mediated RNA trans-splicing. Hum Mol Genet 2009; 18: 3266–73.Google Scholar
Wischik, CM, Edwards, PC, Lai, RYK, Roth, M, Harrington, CR. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc Natl Acad Sci USA. 1996; 93: 11213–18.Google Scholar
Gauthier, S, Feldman, HH, Schneider, LS, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: a randomised, controlled, double-blind, parallel-arm, Phase 3 trial. Lancet 2016; 388: 2873–84.Google Scholar
Tolosa, E, Litvan, I, Höglinger, GU, et al. A Phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov Disord 2014; 29: 470–8.Google Scholar
Gentry, EG, Henderson, BW, Arrant, AE, et al. Rho kinase inhibition as a therapeutic for progressive supranuclear palsy and corticobasal degeneration. J Neurosci 2016; 36: 1316–23.Google Scholar
Woolsey Pharmaceuticals. PSP and CSP patients. Available at: www.woolseypharma.com/psp-cbs/ (accessed December 28, 2020).Google Scholar
Min, SW, Chen, X, Tracy, TE, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med 2015; 21: 1154–62.Google Scholar
VandeVrede, L, Dale, ML, Fields, S, et al. Open-label Phase 1 futility studies of salsalate and young plasma in progressive supranuclear palsy. Mov Disord Clin Pract 2020; 7: 440–7.Google Scholar
Hastings, NB, Wang, X, Song, L, et al. Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener 2017; 12: 116.Google Scholar
Asceneuron. R&D pipeline. Available at: www.asceneuron.com/pipeline (accessed December 28, 2020).Google Scholar
Giordano, R, Canesi, M, Isalberti, M, et al. Autologous mesenchymal stem cell therapy for progressive supranuclear palsy: translation into a Phase I controlled, randomized clinical study. J Transl Med 2014; 12: 14.Google Scholar
Katsimpardi, L, Litterman, NK, Schein, PA, et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 2014; 344: 630–4.Google Scholar
Villeda, SA, Plambeck, KE, Middeldorp, J, et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med 2014; 20: 659–63.Google Scholar
Desmarais, P, Rohrer, JD, Nguyen, QD, et al. Therapeutic trial design for frontotemporal dementia and related disorders. J Neurol Neurosurg Psychiatry 2019; 90: 412–23.Google Scholar
Tao, JJ, Schram, AM, Hyman, DM. Basket studies: redefining clinical trials in the era of genome-driven oncology. Annu Rev Med 2018; 69: 319–31.Google Scholar
Berry, SM, Connor, JT, Lewis, RJ. The platform trial: an efficient strategy for evaluating multiple treatments. JAMA 2015; 313: 1619–20.Google Scholar

References

Boada, M, López, OL, Olazarán, J, et al. A randomized, controlled clinical trial of plasma exchange with albumin replacement for Alzheimer’s disease: primary results of the AMBAR study. Alzheimers Dement 2020; 16: 1412–25. https://doi.org/10.1002/alz.12137.Google Scholar
Kivipelto, M, Mangialasche, F, Ngandu, T. Lifestyle interventions to prevent cognitive impairment, dementia and Alzheimer disease. Nat Rev Neurol 2018; 14: 653–66. https://doi.org/10.1038/s41582-018-0070-3.Google Scholar
Cummings, J, Ballard, C, Tariot, P, et al. Pimavanserin: potential treatment for dementia-related psychosis. J Prev Alzheimers Dis 2018; 5: 253–8. https://doi.org/10.14283/jpad.2018.29.Google Scholar
Huang, P, Goetz, CG, Woolson, RF, et al. Using global statistical tests in long-term Parkinson’s disease clinical trials. Mov Disord 2009; 24: 1732–9. https://doi.org/10.1002/mds.22645.Google Scholar
Dickson, SP. Comparison of aducanumab, solanezumab and BAN2401 using a global statistical test for assessing impact on overall strength of evidence. 13th Clinical Trials on Alzheimer’s Disease (CTAD) Congress, November 4–7, 2020.Google Scholar
Schneider, LS, Goldberg, TE. Composite cognitive and functional measures for early stage Alzheimer’s disease trials. Alzheimers Dement Diagnosis, Assess Dis Monit 2020; 12: 19. https://doi.org/10.1002/dad2.12017.Google Scholar
Hendrix, SB, Dickson, SP, Ellison, N. Composite endpoints – pro perspective: the key to successful clinical development in Alzheimer’s disease. Alzheimers Dement 2019; 15: P1274. https://doi.org/10.1016/j.jalz.2019.06.3660.Google Scholar
Hendrix, SB, Ellison, N. ADCOMS: a sensitive measure of disease progression in early AD supporting better development decisions. Alzheimers Dement 2019; 15: P520. https://doi.org/10.1016/j.jalz.2019.06.4426.Google Scholar
Wang, J, Logovinsky, V, Hendrix, SB, et al. ADCOMS: a composite clinical outcome for prodromal Alzheimer’s disease trials. J Neurol Neurosurg Psychiatry 2016; 87: 993–9. https://doi.org/10.1136/jnnp-2015-312383.Google Scholar
Wessels, AM, Siemers, ER, Yu, P, et al. A combined measure of cognition and function for clinical trials: the Integrated Alzheimer’s Disease Rating Scale (iADRS). J Prev Alzheimers Dis 2015; 2: 227–41. https://doi.org/10.14283/jpad.2015.82.Google Scholar
Langbaum, JB, Ellison, NN, Caputo, A, et al. The Alzheimer’s Prevention Initiative Composite Cognitive test: a practical measure for tracking cognitive decline in preclinical Alzheimer’s disease. Alzheimers Res Ther 2020; 12: 111. https://doi.org/10.1186/s13195-020-00633-2.Google Scholar
Donohue, MC, Sperling, RA, Salmon, DP, et al. The Preclinical Alzheimer Cognitive Composite: measuring amyloid-related decline. JAMA Neurol 2014; 71: 961–70. https://doi.org/10.1001/jamaneurol.2014.803.Google Scholar
Blennow, K, Zetterberg, H. Biomarkers for Alzheimer’s disease: current status and prospects for the future. J Intern Med 2018; 284: 643–63. https://doi.org/10.1111/joim.12816.Google Scholar
Ismail, R, Parbo, P, Madsen, LS, et al. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: a longitudinal PET study. J Neuroinflammation 2020; 17: 151. https://doi.org/10.1186/s12974-020-01820-6.Google Scholar
Preische, O, Schultz, SA, Apel, A, et al. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat Med 2019; 25: 277–83. https://doi.org/10.1038/s41591-018-0304-3.Google Scholar
Liu, W, Lin, H, He, X, et al. Neurogranin as a cognitive biomarker in cerebrospinal fluid and blood exosomes for Alzheimer’s disease and mild cognitive impairment. Transl Psychiatry 2020; 10: 125. https://doi.org/10.1038/s41398-020-0801-2.Google Scholar
O’Brien, PC, Fleming, TR. A multiple testing procedure for clinical trials. Biometrics 1979; 35: 549–56. https://doi.org/10.2307/2530245.Google Scholar
Lan, KKG, Demets, DL. Discrete sequential boundaries for clinical trials. Biometrika 1983; 70: 659–63. https://doi.org/10.1093/biomet/70.3.659.Google Scholar
Whitehead, J, Stratton, I. Group sequential clinical trials with triangular continuation regions. Biometrics 1983; 39: 227–36. https://doi.org/10.2307/2530822.Google Scholar
Hume, S, Aerts, J, Sarnikar, S, Huser, V. Current applications and future directions for the CDISC Operational Data Model standard: a methodological review. J Biomed Inform 2016; 60: 352–62. https://doi.org/10.1016/j.jbi.2016.02.016.Google Scholar
Hendrix, SB, Wilcock, GK. What we have learned from the Myriad trials. J Nutr Health Aging 2009; 13: 362–4. https://doi.org/10.1007/s12603-009-0044-7.Google Scholar
Hendrix, SB. Measuring clinical progression in MCI and pre-MCI populations: enrichment and optimizing clinical outcomes over time. Alzheimers Res Ther 2012; 4: 24. https://doi.org/10.1186/alzrt127.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. Guidance for industry. Available at: www.fda.gov/files/drugs/published/Alzheimer%E2%80%99s-Disease---Developing-Drugs-for-Treatment-Guidance-for-Industy.pdf (accessed January 20, 2021).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 January 20, 2021).Google Scholar

References

Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Babulal, GM, Quiroz, YT, Albensi, BC, et al. Perspectives on ethnic and racial disparities in Alzheimer’s disease and related dementias: update and areas of immediate need. Alzheimers Dement 2019; 15: 292312.Google Scholar
Wong, R, Amano, T, Lin, S-Y, Zhou, Y, Morrow-Howell, N. Strategies for the recruitment and retention of racial/ethnic minorities in Alzheimer disease and dementia clinical research. Curr Alzheimer Res 2019; 16: 458–71.Google Scholar
Ashford, MT, Eichenbaum, J, Williams, T, et al. Effects of sex, race, ethnicity, and education on online aging research participation. Alzheimers Dement (N Y) 2020; 6: 19.Google Scholar
McDonald, A, Knight, R, Cambell, M, et al. What influences recruitment to randomized controlled trials? A review of trials funded by two UK funding agencies. Trials 2006; 7: 18.Google Scholar
Dowling, NM, Olson, N, Mish, T, Kaprakattu, P, Gleason, C. A model for the design and implementation of a participant recruitment registry for clinical studies of older adults. Clin Trials 2012; 9: 204–14.Google Scholar
Hsu, D, Marshall, GA. Primary and secondary prevention trials in Alzheimer disease: looking back, moving forward. Curr Alzheimer Res 2016; 14: 426–40.Google Scholar
Aisen, PS, Sperling, RA, Cummings, J, et al. The Trial-Ready Cohort for Preclinical/Prodromal Alzheimer’s Disease (TRC-PAD) project: an overview. J Prev Alzheimers Dis 2020; 7: 208–12.Google Scholar
Rafii, MS, Aisen, PS. Alzheimer’s disease clinical trials: moving toward successful prevention. CNS Drugs 2019; 33: 99106. https://doi.org/10.1007/s40263-018-0598-1.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug-development pipeline: 2020. Alzheimers Dement (N Y) 2020; 6: e12050.Google Scholar
US Food and Drug Administration. Enhancing the diversity of clinical trial populations: eligibility criteria, enrollment practices, and trial designs guidance for industry. November 2020. www.fda.gov/regulatory-information/search-fda-guidance-documents/enhancing-diversity-clinical-trial-populations-eligibility-criteria-enrollment-practices-and-trial. Available at: www.fda.gov/media/127712/download November 17, 2020).Google Scholar
National Institute on Aging. Alzheimer’s disease and related dementias: clinical studies recruitment planning guide. Available at: www.nia.nih.gov/sites/default/files/2019-05/ADEAR-recruitment-guide-508.pdf (accessed June 17, 2020).Google Scholar
Heredia, NI, Strong, LL, Hatten, V. Community perceptions of biobanking participation: a qualitative study among Mexican-Americans in three Texas cities. Public Health Genom 2017; 20: 4657.Google Scholar
Nuño, MM, Gillen, DL, Dosanjh, KK, et al. Attitudes toward clinical trials across the Alzheimer ’s disease spectrum. Alzheimers Res Ther 2017; 9: 8190.Google Scholar
Zhou, Y, Elashoff, D, Kremen, S, et al. African Americans are less likely to enroll in preclinical Alzheimer’s disease clinical trials. Alzheimers Dement (N Y) 2017; 3: 5764.Google Scholar
Barnes, LL, Bennett, DA. Alzheimer’s disease in African Americans: risk factors and challenges for the future. Health Aff 2014; 33: 580–6.Google Scholar
Matthews, KA, Xu, W, Gaglioti, AH, et al. Racial and ethnic estimates of Alzheimer’s disease and related dementias in the United States (2015–2060) in adults aged ≥65 years. Alzheimers Dement 2019; 15: 1724.Google Scholar
Faison, WE, Schultz, SK, Aerssens, J, et al. Potential ethnic modifiers in the assessment and treatment of Alzheimer’s disease: challenges for the future. Int Psychogeriatr 2007; 19: 539–58.Google Scholar
Olin, JT, Dagerman, KS, Fox, LS, Bowers, B, Schneider, SL. Increasing ethnic minority participation in Alzheimer’s disease research. Alzheimer Dis Assoc Disord 2002; 16: S82–5.Google Scholar
Grill, JD, Kwon, J, Teylan, MA, et al. Retention of Alzheimer disease research participants. Alzheimer Dis Assoc Disord 2019; 33: 299306.Google Scholar
Chêne, G, Beiser, A, Au, R, et al. Gender and incidence of dementia in the Framingham Heart Study from mid-adult life. Alzheimers Dement 2015; 11: 310–20.Google Scholar
Hebert, L, Weuve, J, Scherr, PA, Evans, D. Alzheimer’s disease in the United States (2010–2050) estimated using the 2010 Census. Neurology 2013; 80: 1778–83.Google Scholar
Plassman, BL, Langa, KM, Fisher, GG, et al. Prevalence of dementia in the United States: the aging, demographics, and memory study. Neuroepidemiology 2007; 29: 125–32.Google Scholar
Mehta, K, Yeo, G. Systematic review of dementia prevalence and incidence in US racial/ethnic populations. Alzheimers Dement 2017; 13: 7283.Google Scholar
Weden, MM. Secular trends in dementia and cognitive impairment of U.S. rural and urban older adults. Am J Prev Med 2018; 54: 164–72.Google Scholar
Jaffe, S. Aging in rural America. Health Aff 2015; 34: 710.Google Scholar
Singh, GK, Siahpush, M. Widening rural–urban disparities in all-cause mortality and mortality from major causes of death in the USA, 1969–2009. J Urban Health 2014; 91: 272–92.Google Scholar
Borak, J, Salipante-Zaidel, C, Slade, M, Fields, C. Mortality disparities in Appalachia: reassessment of major risk factors. J Occup Environ Med 2012; 54: 146–56.Google Scholar
Galvin, J, Fu, Q, Nguyen, J, Glasheen, C, Scharff, D. Psychosocial determinants of intention to screen for Alzheimer’s disease. Alzheimers Dement 2009; 4: 353–60.Google Scholar
Wiese, LK, Williams, CL, Tappen, RM. Analysis of barriers to cognitive screening in rural populations in the United States. Adv Nurs Sci 2014; 37: 327–39.Google Scholar
National Institute on Aging. Together we make the difference: national strategy for recruitment and participation in Alzheimer’s and related dementias clinical research. Available at: www.nia.nih.gov/research/recruitment-strategy (accessed October 22, 2020).Google Scholar
Marquez, DX, Glover, CM, Lamar, M, et al. Representation of older Latinxs in cohort studies at the Rush Alzheimer’s Disease Center. Neuroepidemiology 2020; 54: 404–18.Google Scholar
Williams, MM, Meisel, MM, Williams, J, Morris, JC. An interdisciplinary outreach model of African American recruitment for Alzheimer’s disease research. Gerontologist 2011; 51: 134–41.Google Scholar
Howell, JC, Parker, MW, Watts, KD, et al. Research lumbar punctures among African Americans and Caucasians: perception predicts experience. Front Aging Neurosci 2016; 8: 17.Google Scholar
Boise, L, Hinton, L, Rosen, HJ, Ruhl, M. Will my soul go to heaven if they take my brain? Beliefs and worries about brain donation among four ethnic groups. Gerontologist 2017; 57: 719–34.Google Scholar
Walter, S, Clanton, TB, Langford, OG, et al. Recruitment into the Alzheimer Prevention Trials (APT) Webstudy for a Trial-Ready Cohort for Preclinical and Prodromal Alzheimer’s Disease (TRC–PAD). J Prev Alzheimers Dis 2020; 7: 219–25.Google Scholar
Jimenez-Maggiora, GA, Bruschi, S, Raman, R, et al. TRC-PAD: accelerating recruitment of AD clinical trials through innovative information technology. J Prev Alzheimers Dis 2020; 7: 226–33.Google Scholar
Grill, JD, Monsell, SE. Choosing Alzheimer’s disease prevention clinical trial populations. Neurobiol Aging 2014; 35: 116.Google Scholar
Weuve, J, Sagiv, SK, Fox, MP. Quantitative bias analysis for collaborative science. Epidemiology 2018; 29: 627–30.Google Scholar
Burke, SL, Hu, T, Naseh, M, et al. Factors influencing attrition in 35 Alzheimer’s disease centers across the USA: a longitudinal examination of the National Alzheimer’s Coordinating Center’s Uniform Data Set. Aging Clin Exp Res 2019; 31: 1283–97.Google Scholar
Glymour, MM, Chěne, G, Tzourio, C, Brain, Dufouil C. MRI markers and dropout in a longitudinal study of cognitive aging: the three-city Dijon study. Neurology 2012; 79: 1340–8.Google Scholar
Crocker, JC, Ricci-Cabello, I, Parker, A, et al. Impact of patient and public involvement on enrolment and retention in clinical trials: systematic review and meta-analysis. BMJ 2018; 363: 117.Google Scholar

References

Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Fargo, KN, Carrillo, MC, Weiner, MW, Potter, WZ, Khachaturian, Z. The crisis in recruitment for clinical trials in Alzheimer’s and dementia: an action plan for solutions. Alzheimers Dement 2016; 12: 1113–15.Google Scholar
Grill, JD, Karlawish, J. Addressing the challenges to successful recruitment and retention in Alzheimer’s disease clinical trials. Alzheimers Res Ther 2010; 2: 34.Google Scholar
Tang, M-X, Cross, P, Andrews, H, et al. Incidence of AD in African-Americans, Caribbean Hispanics, and Caucasians in northern Manhattan. Neurology 2001; 56: 4956.Google Scholar
Sperling, RA, Aisen, PS, Beckett, LA, 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.Google Scholar
Norton, S, Matthews, FE, Barnes, DE, Yaffe, K, Brayne, C. Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol 2014; 13: 788–94.Google Scholar
Ovod, V, Ramsey, KN, Mawuenyega, KG, et al. Amyloid beta concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement 2017; 13: 841–9.Google Scholar
Mattsson-Carlgren, N, Janelidze, S, Palmqvist, S, et al. Longitudinal plasma p-tau217 is increased in early stages of Alzheimer’s disease. Brain 2020; 143: 3234–41.Google Scholar
Aisen, P, Touchon, J, Andrieu, S, et al. Registries and cohorts to accelerate early phase Alzheimer’s trials. A report from the E.U./U.S. Clinical Trials in Alzheimer’s Disease Task Force. J Prev Alzheimers Dis 2016; 3: 68–4.Google Scholar
Kluding, PM, Denton, J, Jamison, TR, et al. Frontiers: integration of a research participant registry with medical clinic registration and electronic health records. Clin Transl Sci 2015; 8: 405–11.Google Scholar
Grill, JD. Recruiting to preclinical Alzheimer’s disease clinical trials through registries. Alzheimers Dement (N Y) 2017; 3: 205–12.Google Scholar
Jeon, YH, Fargo, K, Smith, A, et al. Comparison of digital platforms for participant recruitment in dementia research: lessons and future directions from a global collaborative: epidemiology/innovative methods in epidemiology (i.e., assessment methods, design, recruitment strategies, statistical methods, etc.). Alzheimers Dement 2020; 16: e046401.Google Scholar
Weiner, MW, Nosheny, R, Camacho, M, et al. The Brain Health Registry: an internet-based platform for recruitment, assessment, and longitudinal monitoring of participants for neuroscience studies. Alzheimers Dement 2018; 14: 1063–76.Google Scholar
Mackin, RS, Insel, PS, Truran, D, et al. Unsupervised online neuropsychological test performance for individuals with mild cognitive impairment and dementia: results from the Brain Health Registry. Alzheimers Dement (Amst) 2018 10: 573–82.Google Scholar
Nosheny, RL, Camacho, MR, Insel, PS, et al. Online study partner-reported cognitive decline in the Brain Health Registry. Alzheimers Dement 2018; 4: 565–74.Google Scholar
Nosheny, RL, Camacho, MR, Jin, C, et al. Validation of online functional measures in cognitively impaired older adults. Alzheimers Dement 2020; 16: 1426–37.Google Scholar
Ashford, MT, Neuhaus, J, Jin, C, et al. Predicting amyloid status using self-report information from an online research and recruitment registry: the Brain Health Registry. Alzheimers Dement (Amst) 2020; 12: e12102.Google Scholar
Ashford, MT, Eichenbaum, J, Williams, T, et al. Effects of sex, race, ethnicity, and education on online aging research participation. Alzheimers Dement (N Y) 2020; 6: e12028.Google Scholar
Fockler, JK, Ashford, MT, Flenniken, D, et al. Brain Health Registry GenePool study: a novel approach to online genetics research. Alzheimers Dement (N Y) 2021; 7: e12118.Google Scholar
Langbaum, JB, High, N, Nichols, J, et al. The Alzheimer’s Prevention Registry: a large internet-based participant recruitment registry to accelerate referrals to Alzheimer’s-focused studies. J Prev Alzheimers Dis 2020; 7: 242–50.Google Scholar
Reiman, EM, Langbaum, JB, Tariot, PN. Alzheimer’s Prevention Initiative: a proposal to evaluate presymptomatic treatments as quickly as possible. Biomark Med 2010; 4: 314.Google Scholar
Langbaum, JB, Karlawish, J, Roberts, JS, et al. GeneMatch: a novel recruitment registry using at-home APoE genotyping to enhance referrals to Alzheimer’s prevention studies. Alzheimers Dement 2019; 15: 515–24.Google Scholar
Zhong, K, Cummings, J. Healthybrains.org: from registry to randomization. J Prev Alzheimers Dis 2016; 3: 123–6.Google Scholar
Amariglio, RE, Donohue, MC, Marshall, GA, et al. Tracking early decline in cognitive function in older individuals at risk for Alzheimer disease dementia. JAMA Neurol 2015; 72: 446.Google Scholar
Walter, S, Clanton, TB, Langford, OG, et al. Recruitment into the Alzheimer Prevention Trials (APT) Webstudy for a Trial-Ready Cohort for Preclinical and Prodromal Alzheimer’s Disease (TRC–PAD). J Prev Alzheimers Dis 2020; 7: 219–25.Google Scholar
Sperling, RA, Donohue, MC, Raman, R, et al. Association of factors with elevated amyloid burden in clinically normal older individuals. JAMA Neurol 2020; 77: 735.Google Scholar
Aisen, PS, Sperling, RA, Cummings, J, et al. The Trial-Ready Cohort for Preclinical/Prodromal Alzheimer’s Disease (TRC-PAD) project: an overview. J Prev Alzheimers Dis 2020; 7: 208–12.Google Scholar
Langford, O, Raman, R, Sperling, RA, et al. Predicting amyloid burden to accelerate recruitment of secondary prevention clinical trials. J Prev Alzheimers Dis 2020; 7: 213–18.Google Scholar
Walter, S, Langford, OG, Clanton, TB, et al. The Trial-Ready Cohort for Preclinical and Prodromal Alzheimer’s Disease (TRC-PAD): experience from the first 3 years. J Prev Alzheimers Dis 2020; 4: 234–41.Google Scholar
Perrin, A, Turner, E. Smartphones help blacks, Hispanics bridge some – but not all – digital gaps with whites. Available at: www.pewresearch.org/fact-tank/2019/08/20/smartphones-help-blacks-hispanics-bridge-some-but-not-all-digital-gaps-with-whites/ (accessed April 2020).Google Scholar

References

Heart Special Project Committee. Organization, review, and administration of cooperative studies (Greenberg Report): a report from the Heart Special Project Committee to the National Advisory Heart Council, May 1967. Control Clin Trials 1988; 9: 137–48.Google Scholar
DeMets, DL, Ellenberg, SS. Data monitoring committees expect the unexpected. N Engl J Med 2016; 375: 1365–71.Google Scholar
National Institutes of Health. NIH guide for grants and contracts. 1979. Available at: https://grants.nih.gov/grants/guide/historical/1979_06_05_Vol_08_No_08.pdf (accessed November 14, 2020).Google Scholar
National Institutes of Health. NIH policy for data and safety monitoring. 1998. Available at: https://grants.nih.gov/grants/guide/notice-files/not98-084.html (accessed November 14, 2020).Google Scholar
International Conference on Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. Integrated addendum to ICH E6(R1): guideline for good clinical practice. 2016. Available at: https://ichgcp.net/ (accessed November 14, 2020).Google Scholar
International Conference on Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. Statistical principles for clinical trials. 1998. Available at: https://database.ich.org/sites/default/files/E9_Guideline.pdf (accessed November 14, 2020).Google Scholar
Food and Drug Administration. Guidance for clinical trial sponsors: establishment and operation of clinical trial data monitoring committees. March 2006. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/establishment-and-operation-clinical-trial-data-monitoring-committees (accessed November 18, 2020).Google Scholar
Gewandter, JS, Kitt, RA, Hunsinger, MR, et al. Reporting of data monitoring boards in publications of randomized clinical trials is often deficient: ACTTION systematic review. J Clin Epidemiol 2017; 83: 101–7.Google Scholar
Calis, KA, Archdeacon, P, Bain, RP, et al. Understanding the functions and operations of data monitoring committees: survey and focus group findings. Clin Trials 2017; 14: 5966.Google Scholar
Sartor, O, Halabi, S. Independent data monitoring committees: an update and overview. Urol Oncol 2015; 33: 143–8.Google Scholar
National Institutes of Health (NIH). Further guidance on data and safety monitoring for Phase I and Phase II trials. 2000. Available at: https://grants.nih.gov/grants/guide/notice-files/not-od-00-038.html (accessed November 18, 2020).Google Scholar
US Department of Veterans Affairs. Clinical Science Research & Development (CSRD), data monitoring committee guidance. 2015. Available at: www.research.va.gov/services/csrd/data-monitoring.cfm (accessed December 3, 2020).Google Scholar
Calis, KA, Archdeacon, P, Bain, R, et al. Recommendations for data monitoring committees from the Clinical Trials Transformation Initiative. Clin Trials 2017; 14: 342–8.Google Scholar
DMC Project. CTTI recommendations: data monitoring committees. 2016. Available at: www.ctti-clinicaltrials.org/files/recommendations/dmc-recommendations.pdf (accessed November 18, 2020).Google Scholar
CTSA Collaborative DSMB Workgroup. DSMB training manual. 2018. Available at: https://tuftsctsi.wpengine.com/research-services/regulatory/data-and-safety-monitoring-board-training-manual-for-investigator-initiated-studies (accessed December 3, 2020).Google Scholar
Grant, AM, Altman, DG, Babiker, AG, et al. A proposed charter for clinical trial data monitoring committees: helping them to do their job well. Lancet 2005; 365: 711–22.Google Scholar
Neaton, JD, Grund, B, Wentworth, D. How to construct an optimal interim report: what the data monitoring committee does and doesn’t need to know. Clin Trials 2018; 15: 359–65.Google Scholar
Ellenberg, SS, Fleming, TR, DeMets, DL. Data Monitoring Committees in Clinical Trials : A Practical Perspective. Newark, NJ: John Wiley & Sons Inc.; 2019.Google Scholar
Kim, K. Sequential designs for clinical trials. In Designs for Clinical Trials: Perspectives on Current Issues, Harrington, D (ed.). New York, NY: Springer; 2012: 5780.Google Scholar
Doody, RS, Raman, R, Farlow, M, et al. A Phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med 2013; 369: 341–50.Google Scholar
Yunusa, I, El Helou, ML, Alsahali, S. Pimavanserin: a novel antipsychotic with potentials to address an unmet need of older adults with dementia-related psychosis. Front Pharmacol 2020; 11: 15.Google Scholar
Wilhelmsen, L. Role of the data and safety monitoring committee (DSMC). Stat Med 2002; 21: 2823–9.Google Scholar
Acadia Pharmaceuticals. ACADIA pharmaceuticals presents positive top-line results from pivotal Phase 3 harmony trial of pimavanserin in patients with dementia-related psychosis. 12th Clinical Trials on Alzheimer’s Disease (CTAD) Meeting, December 4-7, 2019. Available at: https://ir.acadia-pharm.com/news-releases/news-release-details/acadia-pharmaceuticals-presents-positive-top-line-results?field_nir_news_date_value (accessed December 3, 2020).Google Scholar
Alzforum.org. Biogen/Eisai halt Phase 3 aducanumab trials. Available at: www.alzforum.org/news/research-news/biogeneisai-halt-phase-3-aducanumab-trials (accessed December 10, 2020).Google Scholar
Alzforum.org. Aducanumab still needs to prove itself, researchers say. Available at: www.alzforum.org/news/research-news/aducanumab-still-needs-prove-itself-researchers-say (accessed December 10, 2020).Google Scholar
Karlawish, J. If the FDA approved Biogen’s Alzheimer’s treatment, I won’t prescribe it. Available at: www.statnews.com/2021/05/30/if-the-fda-approves-biogens-alzheimers-treatment-i-wont-prescribe-it/ (accessed July 8, 2021).Google Scholar

References

Wimo, A. The worldwide costs of dementia 2015 and comparisons with 2010. Alzheimers Dement 2017; 13: 17.Google Scholar
Gustavsson, A, Green, C, Jones, RW, et al. Current issues and future research priorities for health economic modelling across the full continuum of Alzheimer’s disease. Alzheimers Dement 2017; 13: 312–21.Google Scholar
Prince, M, Ali, G, Guerchet, M, et al. Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimers Res Ther 2016; 8: 23.Google Scholar
Thiers, FA, Sinskey, AJ, Berndt, ER. Trends in the globalization of clinical trials. Nat Rev Drug Discov 2008; 7: 1314.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug-development pipeline: 2020. Alzheimers Dement (N Y) 2020; 6: e12050.Google Scholar
Nichols, E, Szoeke, CEI, Vollset, SE, et al. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2019; 18: 88106.Google Scholar
Qiao, Y, Alexander, GC, Moore, TJ. Globalization of clinical trials: variation in estimated regional costs of pivotal trials, 2015–2016. Clin Trials 2019; 16: 329–33.Google Scholar
Alzheimer’s Disease International. Dementia statistics. Available at: www.alzint.org/about/dementia-facts-figures/dementia-statistics/ (accessed June 29, 2021).Google Scholar
Jia, L, Du, Y, Chu, L, et al. Prevalence, risk factors, and management of dementia and mild cognitive impairment in adults aged 60 years or older in China: a cross-sectional study. Lancet Public Health 2020; 5: e66171.Google Scholar
Jia, L, Quan, M, Fu, Y, et al. Dementia in China: epidemiology, clinical management, and research advances. Lancet Neurol 2019; 4422: 112.Google Scholar
Jones, CW, Handler, L, Crowell, KE, et al. Non-publication of large randomized clinical trials: cross sectional analysis. BMJ 2013; 347: 19.Google Scholar
Gauthier, S, Albert, M, Fox, N, et al. Why has therapy development for dementia failed in the last two decades? Alzheimers Dement 2016; 12: 60–4.Google Scholar
Goldman, DP, Fillit, H, Neumann, P. Accelerating Alzheimer’s disease drug innovations from the research pipeline to patients. Alzheimers Dement 2018; 14: 833–6.Google Scholar
Jia, J, Zuo, X, Jia, XF, et al. Diagnosis and treatment of dementia in neurology outpatient departments of general hospitals in China. Alzheimers Dement 2016; 12: 446–53.Google Scholar
Wang, H, Xie, H, Qu, Q, et al. The continuum of care for dementia: needs, resources and practice in China. J Glob Health 2019; 9: 020321.Google Scholar
Long, JM, Holtzman, DM. Alzheimer disease: an update on pathobiology and treatment strategies. Cell 2019; 179: 312–39.Google Scholar
Wang, X, Wang, H, Li, H, Li, T, Yu, X. Frequency of the apolipoprotein E ε4 allele in a memory clinic cohort in Beijing: a naturalistic descriptive study. PLoS One 2014; 9: e99130.Google Scholar
Zhang, M, Wang, H, Li, T, Yu, X. Prevalence of neuropsychiatric symptoms across the declining memory continuum: an observational study in a memory clinic setting. Dement Geriatr Cogn Dis Extra 2012; 2: 200–8.Google Scholar
Petersen, RC, Thomas, RG, Aisen, PS, et al. Randomized controlled trials in mild cognitive impairment: sources of variability. Neurology 2017; 88: 1751–8.Google Scholar
Zhao, M, Lv, X, Tuerxun, M, et al. Delayed help seeking behavior in dementia care: preliminary findings from the Clinical Pathway for Alzheimer’s Disease in China (CPAD) study. Int Psychogeriatr 2016; 28: 211–19.Google Scholar
Paulino Ramirez, Diaz S, Gil Gregório, P, Manuel Ribera Casado, J, et al. The need for a consensus in the use of assessment tools for Alzheimer’s disease: the Feasibility Study (assessment tools for dementia in Alzheimer centres across Europe), a European Alzheimer’s Disease Consortium’s (EADC) survey. Int J Geriatr Psychiatry 2005; 20: 744–8.Google Scholar
Shen, JHQ, Shen, Q, Yu, H, et al. Validation of an Alzheimer’s disease assessment battery in Asian participants with mild to moderate Alzheimer’s disease. Am J Neurodegener Dis 2014; 3: 158–69.Google Scholar
Pocock, S, Calvo, G, Marrugat, J, et al. International differences in treatment effect: do they really exist and why? Eur Heart J 2013; 34: 1846–52.Google Scholar
Mentz, RJ, Kaski, J-C, Dan, G-A, et al. Implications of geographical variation on clinical outcomes of cardiovascular trials. Am Heart J 2012; 164: 303–12.Google Scholar
Kristensen, SL, Martinez, F, Jhund, PS, et al. Geographic variations in the PARADIGM-HF heart failure trial. Eur Heart J 2016; 37: 3167–74.Google Scholar
Cummings, JL, Atri, A, Ballard, C, et al. Insights into globalization: comparison of patient characteristics and disease progression among geographic regions in a multinational Alzheimer’s disease clinical program. Alzheimers Res Ther 2018; 10: 116.Google Scholar
Cummings, J, Reynders, R, Zhong, K. Globalization of Alzheimer’s disease clinical trials. Alzheimers Res Ther 2011; 3: 24.Google Scholar
Wang, H. Nexus between cognitive reserve and modifiable risk factors of dementia. Int Psychogeriatr 2020; 32: 559–62.Google Scholar
Wang, J, Gu, Y, Dong, W, et al. Lower small-worldness of intrinsic brain networks facilitates the cognitive protection of intellectual engagement in elderly people without dementia: a near-infrared spectroscopy study. Am J Geriatr Psychiatry 2020; 28: 722–31.Google Scholar
Henley, DB, Dowsett, SA, Chen, YF, et al. Alzheimer’s disease progression by geographical region in a clinical trial setting. Alzheimers Res Ther 2015; 7: 43.Google Scholar
Jack, CR, Bennett, DA, Blennow, K, et al. NIA–AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14: 535–62.Google Scholar
Hampel, H, Vergallo, A, Flores Aguilar, L, et al. Precision pharmacology for Alzheimer’s disease for the Alzheimer Precision Medicine Initiative (APMI). Pharmacol Res 2018; 130: 331–65.Google Scholar
Wang, X, Sun, G, Feng, T, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res 2019; 29: 787803.Google Scholar
Xiao, S, Chan, P, Wang, T, et al. A 36-week multicenter, randomized, double-blind, placebo-controlled, parallel-group, Phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimers Res Ther 2021; 13: 62.Google Scholar
Editorial. Alzheimer’s disease research enterprise in the era of COVID-19/SARS-CoV-2. Alzheimers Dement 2020; 16: 587–8.Google Scholar

References

Rosen, WG, Mohs, RC, Davis, KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry 1984; 141: 1356–64.Google Scholar
Folstein, MF, Folstein, SE, McHugh, PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975; 12: 189–98.Google Scholar
Wesnes, KA, Harrison, JE. The evaluation of cognitive function in the dementias: methodological and regulatory considerations. Dialog Clin Neurosci 2003; 5: 7788.Google Scholar
Hobart, J, Cano, S, Posner, H, et al. Putting the Alzheimer’s cognitive test to the test I: traditional psychometric methods. Alzheimers Dement 2013; 9: S49.Google Scholar
Mohs, RC, Knopman, D, Petersen, RC, et al. Development of cognitive instruments for use in clinical trials of antidementia drugs: additions to the Alzheimer’s Disease Assessment Scale that broaden its scope. The Alzheimer’s Disease Cooperative Study. Alzheimer Dis Assoc Disord 1997; 11: 1321.Google Scholar
Vellas, B, Andrieu, S, Sampaio, C, et al. Endpoints for trials in Alzheimer’s disease: a European task force consensus. Lancet Neurol 2008; 7: 436–50.Google Scholar
Ritchie, K, Ropacki, M, Albala, B, et al. Recommended cognitive outcomes in pre-clinical Alzheimer’s disease: consensus statement from the European Prevention of Alzheimer’s Dementia (EPAD) project. Alzheimers Dement 2017; 13: 186–95.Google Scholar
Podhorna, J, Krahnke, T, Shear, T, et al. Alzheimer’s Disease Assessment Scale – cognitive subscale variants in mild cognitive impairment and mild Alzheimer’s disease: change over time and the effects of enrichment strategies. Alzheimers Res Ther 2016; 8: 113.Google Scholar
Hendrix, S, Ellison, N. Are Alzheimer’s treatment failures due to inactive compounds or are we doing something wrong? Alzheimers Dement 2017; 13: P617.Google Scholar
Sevigny, JJ, Peng, Y, Liu, L, et al. Item analysis of ADAS-cog: effect of baseline cognitive impairment in a clinical AD trial. Am J Alzheimers Dis Other Demen 2010; 25: 119–24.Google Scholar
Winblad, B, Gauthier, S, Scinto, L, et al. Safety and efficacy of galantamine in subjects with mild cognitive impairment. Neurology 2008; 70: 2024–35.Google Scholar
Hort, J, Andel, R, Mokrisova, I, et al. Effect of donepezil in Alzheimer disease can be measured by a computerized human analog of the Morris water maze. Neurodegener Dis 2014; 13: 192–6.Google Scholar
Hendrix, SB, Wells, BM, and the Alzheimer’s Disease Neuroimaging Initiative (ADNI).Time course of cognitive decline in subjects with mild Alzheimer’s disease based on ADAS-cog subscales and neuropsychological tests measured in ADNI. Alzheimers Dement 2010; 6: e50.Google Scholar
Gilman, S, Koller, M, Black, RS, et al. Clinical effects of Abeta immunisation (AN1792) in patients with AD in an interrupted trial. Neurology 2005; 64: 1553–62.Google Scholar
Rosenberg, A, Ngandu, T, Rusanen, M, et al. Multidomain lifestyle intervention benefits a large elderly population at risk for cognitive decline and dementia regardless of baseline characteristics: the FINGER trial. Alzheimers Dement 2018; 14: 263–70.Google Scholar
Harrison, JE, Rentz, D, Brashear, HR, et al. Psychometric evaluation of the Neuropsychological Test Battery in individuals with normal cognition, mild cognitive impairment, or mild to moderate Alzheimer’s disease: results from a longitudinal study. J Prev Alzheimers Dis 2018; 5: 236–44.Google Scholar
European Medicines Agency. Guideline on the clinical investigation of medicines for the treatment of Alzheimer’s disease. CPMP/EWP/553/95 Rev.2 2018. Avaialable at: www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-investigation-medicines-treatment-alzheimers-disease-revision-2_en.pdf (accessed December 31, 2020).Google Scholar
Food and Drug Administration. Early Alzheimer’s Disease: Developing Drugs for Treatment. Guidance for Industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER); 2018.Google Scholar
Leber, P. What is the evidence that a dementia treatment works? Criteria used by drug regulatory authorities. In Evidence-Based Dementia Practice, Qizilbash, N, Schneider, LS, Chui, H, et al. (eds.). Oxford: Blackwells; 2003: 376–87.Google Scholar
Food and Drug Administration. Guidance for Industry Alzheimer’s Disease: Developing Drugs for the Treatment of Early Stage Disease (FDA-2013-D-0077) Draft; 2013.Google Scholar
Harrison, JE. Cognition comes of age: comments on the new FDA draft guidance for early Alzheimer’s disease. Alzheimers Res Ther 2018; 10: 61.Google Scholar
Szlyk, JP, Myers, L, Zhang, YX, et al. Development and assessment of a neuropsychological battery to aid in predicting driving performance. J Rehabil Res Dev 2002; 39: 483–96.Google Scholar
Liu-Seifert, H, Siemers, E, Selzler, K, et al. Correlation between cognition and function across the spectrum of Alzheimer’s disease. J Prev Alzheimers Dis 2016; 3: 138–44.Google Scholar
Katz, R. Presentation to the Alzheimer’s Association Research Roundtable Scales for Alzheimer’s Disease Meeting. April 2008, Washington, reported in R. Black, B. Greenberg, J.M. Ryan et al. Perspectives scales as outcome measures for Alzheimer’s disease. Alzheimers Dement 2009; 5: 324–39.Google Scholar
Harrison, J, Minassian, S, Jenkins, L, et al. A neuropsychological test battery for use in Alzheimer disease clinical trials. Arch Neurol 2007; 64: 1323–9.Google Scholar
Wessels, AM, Edgar, CJ, Nathan, PJ, et al. Cognitive go/no-go decision-making criteria in Alzheimer’s disease drug development. Drug Discov Today 2021; 26: 1330–6.Google Scholar
Harrison, JE, Bradford, E, Edgar, C, et al. Testing compounds which facilitate learning: a new proof of principle paradigm from CDR. XXV CINP Congress, Chicago, July 9–13, 2006.Google Scholar
McIntyre, RS, Harrison, JE, Loft, H, et al. The effects of vortioxetine on cognitive function in patients with major depressive disorder (MDD): a meta-analysis of three randomized controlled trials. Int J Neuropsychopharmacol 2016; 19: pyw055.Google Scholar
Harrison, JE, Lophaven, S, Olsen, CK. Which cognitive domains are improved by treatment with vortioxetine? Int J Neuropsychopharmacol 2016; 9: pyw054.Google Scholar
Schneider, L, Goldberg, TE. Composite cognitive and functional measures for early stage Alzheimer’s disease trials. Alzheimers Dement 2020; 12: e12017.Google Scholar
Harrison, JE. Commentary: composite cognitive and functional measures for early-stage Alzheimer’s disease trials. Alzheimers Dement: Cogn Behav Assess 2020; 12: 13.Google Scholar
Harrison, JE, Hendrix, SB. The assessment of cognition in translational medicine: A contrast between the approaches used in Alzheimer’s disease and major depressive disorder. In Translational Medicine in CNS Drug Development. Handbook of Behavioral Neuroscience, Volume 25, Nomikos G (ed.). Amsterdam: Elsevier; 2019: 297308.Google Scholar
Wang, J, Logovinsky, V, Hendrix, SB, et al. ADCOMS: a composite clinical outcome for prodromal Alzheimer’s disease trials. J Neurol Neurosurg Psychiatry 2016; 87: 993–9.Google Scholar
Jutten, RJ, Harrison, JE, Brunner, AJ, et al. The Cognitive–Functional Composite is sensitive to clinical progression in early dementia: longitudinal findings from the Catch–Cog study cohort. Alzheimers Dement (N Y) 2020; 6: e12020.Google Scholar
Jutten, RS, Harrison, JE, Lee Meeuw Kjoeet, PR, et al. Assessing cognition and daily function in early dementia using the Cognitive–Functional Composite: findings from the Catch–Cog study cohort. Alzheimers Res Ther 2019; 11: 45.Google Scholar
Ferris, SH, Lucca, U, Mohs, R, et al. Objective psychometric tests in clinical trials of dementia drugs. Position paper from the International Working Group on Harmonization of Dementia Drug Guidelines. Alzheimers Dis Assoc Disord 1997; 11: 34–8.Google Scholar
Lannfelt, L, Blennow, K, Zetterberg, H, et al. Targeting Aβ as a modifying therapy of Alzheimer’s disease: safety, efficacy and biomarker findings of a Phase IIa randomised, double-blind placebo-controlled trial of PBT2. Lancet Neurol 2008; 7: 779–86.Google Scholar
Hilt, D, Gawryl, M, Koenig, G, et al. EVP-6124: safety, tolerability and cognitive effects of a novel A7 nicotinic receptor agonist in Alzheimer’s disease patients on stable donepezil or rivastigmine therapy. Alzheimers Dement 2009; 5: P4-348.Google Scholar
Scheltens, P, Twisk, JWR, Blesa, R, et al. Efficacy of souvenaid in mild Alzheimer’s disease: results from a randomized, controlled trial. J Alzheimers Dis 2012; 31: 225–36.Google Scholar
Nathan, PJ, Boardley, R, Scott, N, et al. The safety, tolerability, pharmacokinetics and cognitive effects of GSK239512, a selective histamine H₃ receptor antagonist in patients with mild to moderate Alzheimer’s disease: a preliminary investigation. Curr Alzheimer Res 2013; 10: 240–51.Google Scholar
Grove, RA, Harrington, CM, Mahler, A, et al. A randomized, double-blind, placebo-controlled, 16-week study of the H3 receptor antagonist, GSK239512 as a monotherapy in subjects with mild-to-moderate Alzheimer’s disease. Curr Alzheimer Res 2014; 11: 4758.Google Scholar
Scheltens, P, Hallikainen, M, Grimmer, T, et al. Safety, tolerability and efficacy of the glutaminyl-cyclase inhibitor PQ912 in Alzheimer’s disease: results of a randomized, double-blind, placebo-controlled Phase 2a study. Alzheimers Res Ther 2018; 10: 107.Google Scholar
Rockwood, K. Size of the treatment on cognition of cholinesterase inhibitors for Alzheimer’s disease. J Neurol Neurosurg Psychiatry 2004; 75: 677–85.Google Scholar
Cummings, J, Scheltens, P, McKeith, I, et al. Effect size analyses of souvenaid in patients with Alzheimer’s disease. J Alzheimers Dis 2016; 55: 1131–9.Google Scholar
Rockwood, K, Black, SE, Robillard, A, et al. Potential treatment effects of donepezil not detected in Alzheimer’s disease clinical trials: a physician survey. Int J Geriatr Psychiatry 2004; 19: 854–60.Google Scholar
Harrison, JE, Lam, R, Baune, BT, et al. Selection of cognitive tests for trials of therapeutic agents. Lancet Psychiatry 2016; 8: 113.Google Scholar

References

Food and Drug Administration. Digital Health Innovation Action Plan. Available at: www.fda.gov/media/106331/download (accessed 2018).Google Scholar
Food and Drug Administration. Software as a medical device (SAMD): clinical evaluation. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/software-medical-device-samd-clinical-evaluation (accessed 2017).Google Scholar
FDA–NIH Biomarker Working Group. BEST (Biomarkers, Endpoints, and Other Tools) Resource. Silver Spring, MD and Bethesda, MD: Food and Drug Administration and National Institutes of Health; 2016.Google Scholar
Chinner, A, Blane, J, Lancaster, C, Hinds, C, Koychev, I. Digital technologies for the assessment of cognition: a clinical review. Evid-Based Ment Health 2018; 21: 6771.Google Scholar
Koo, BM, Vizer, LM. Mobile technology for cognitive assessment of older adults: a scoping review. Innov Aging 2018; 3: igy038.Google Scholar
Lussier, M, Lavoie, M, Giroux, S, et al. Early detection of mild cognitive impairment with in-home monitoring sensor technologies using functional measures: a systematic review. IEEE J Biomed Health Inform 2019; 23: 838–47.Google Scholar
Thabtah, F, Mampusti, E, Peebles, D, Herradura, R, Varghese, J. A mobile-based screening system for data analyses of early dementia traits detection. J Med Syst 2019; 44: 24.Google Scholar
Piau, A, Wild, K, Mattek, N, Kaye, J. Current state of digital biomarker technologies for real-life, home-based monitoring of cognitive function for mild cognitive impairment to mild Alzheimer disease and implications for clinical care: systematic review. J Med Internet Res 2019, 21: e12785.Google Scholar
Kourtis, LC, Regele, OB, Wright, JM, Jones, GB. Digital biomarkers for Alzheimer’s disease: the mobile/wearable devices opportunity. NPJ Digit Med 2019; 2: 9.Google Scholar
Piers, RJ, Devlin, KN, Ning, B, et al. Age and graphomotor decision making assessed with the digital clock drawing test: the Framingham Heart Study. J Alzheimers Dis 2017; 60: 1611–20.Google Scholar
Thomas, JA, Burkhardt, HA, Chaudhry, S, et al. Assessing the utility of language and voice biomarkers to predict cognitive impairment in the Framingham Heart Study cognitive aging cohort data. J Alzheimers Dis 2020; 76: 905–22.Google Scholar
Moon, S, Song, HJ, Sharma, VD, et al. Classification of Parkinson’s disease and essential tremor based on balance and gait characteristics from wearable motion sensors via machine learning techniques: a data-driven approach. J Neuroeng Rehabil 2020; 17: 125.Google Scholar
Leach, JM, Mancini, M, Kaye, JA, Hayes, TL, Horak, FB. Day-to-day variability of postural sway and its association with cognitive function in older adults: a pilot study. Front Aging Neurosci 2018; 10: 126.Google Scholar
Tulipani, LJ, Meyer, B, Larie, D, Solomon, AJ, McGinnis, RS. Metrics extracted from a single wearable sensor during sit–stand transitions relate to mobility impairment and fall risk in people with multiple sclerosis. Gait Post 2020; 80: 361–6.Google Scholar
Dorsey, ER, Omberg, L, Waddell, E. Deep phenotyping of Parkinson’s disease. J Parkinsons Dis 2020; 10: 855–73.Google Scholar
Hunfalvay, M, Roberts, CM, Murray, NP. Vertical smooth pursuit as a diagnostic marker of traumatic brain injury. Concussion 2020; 5: CNC69.Google Scholar
Regalia, G, Gerboni, G, Migliorini, M, et al. Sleep assessment by means of a wrist actigraphy-based algorithm: agreement with polysomnography in an ambulatory study on older adults. Chronobiol Int 2021; 38: 400–14.Google Scholar
Kuwabara, M, Harada, K, Hishiki, Y, Kario, K. Validation of two watch-type wearable blood pressure monitors according to the ANSI/AAMI/ISO81060-2:2013 guidelines: Omron HEM-6410 T-ZM and HEM-6410 T-ZL. J Clin Hypertens (Greenwich) 2019; 21: 853–8.Google Scholar
Collier, SR, McCraw, C, Campany, M, et al. Withings body cardio versus gold standards of pulse-wave velocity and body composition. J Pers Med 2020; 10: 17.Google Scholar
Mosnaim, GS, Stempel, DA, Gonzalez, C, et al. The impact of patient self-monitoring via electronic medication monitor and mobile app plus remote clinician feedback on adherence to inhaled corticosteroids: a randomized controlled trial. J Allergy Clin Immunol Pract 2021; 9: 1586–94.Google Scholar
Jafri, RZ, Balliro, CA, El-Khatib, F, A three-way accuracy comparison of the Dexcom G5, Abbott Freestyle Libre Pro, and Senseonics Eversense continuous glucose monitoring devices in a home-use study of subjects with type 1 diabetes. Diabetes Technol Therapeut 2020; 22: 846–52.Google Scholar
Vandenberk, T, Storms, V, Lanssens, D, et al. A vendor-independent mobile health monitoring platform for digital health studies: development and usability study. JMIR mHealth uHealth 2019; 7: e12586.Google Scholar
Seelye, A, Mattek, N, Sharma, N. Weekly observations of online survey metadata obtained through home computer use allow for detection of changes in everyday cognition before transition to mild cognitive impairment. Alzheimers Dement 2018; 14: 187–94.Google Scholar
Sano, M, Zhu, CW, Kaye, J, et al. A randomized clinical trial to evaluate home-based assessment of people over 75 years old. Alzheimers Dement 2019; 15: 615–24.Google Scholar
Thomas, N, Beattie, Z, Marcoe, J, et al. An ecologically valid, longitudinal, and unbiased assessment of treatment efficacy in Alzheimer disease (the EVALUATE-AD trial): proof-of-concept study. JMIR Res Protoc 2020; 9: e17603.Google Scholar
Ahamed, F, Shahrestani, S, Cheung, H. Internet of things and machine learning for healthy ageing: identifying the early signs of dementia. Sensors (Basel) 2020; 20: E6031.Google Scholar
Cavuoto, MG, Kinsella, GJ, Ong, B, Pike, KE, Nicholas, CL. Naturalistic measurement of sleep in older adults with amnestic mild cognitive impairment: anxiety symptoms do not explain sleep disturbance. Curr Alzheimer Res 2019; 16: 233–42.Google Scholar
Øksnebjerg, L, Woods, B, Ruth, K, et al. A tablet app supporting self-management for people with dementia: explorative study of adoption and use patterns. JMIR mHealth uHealth 2020; 8: e14694.Google Scholar
Buchman, AS, Dawe, RJ, Leurgans, SE. Different combinations of mobility metrics derived from a wearable sensor are associated with distinct health outcomes in older adults. J Gerontol A Biol Sci Med Sci 2020; 75: 1176–83.Google Scholar
Mueller, A, Hoefling, HA, Muaremi, A, et al. Continuous digital monitoring of walking speed in frail elderly patients: noninterventional validation study and longitudinal clinical trial. JMIR mHealth uHealth 2019; 7: e15191.Google Scholar
De Vito, AN, Sawyer, RJ 2nd, LaRoche, A, et al. Acceptability and feasibility of a multicomponent telehealth care management program in older adults with advanced dementia in a residential memory care unit. Gerontol Geriatr Med 2020; 6: 2333721420924988.Google Scholar
Seelye, A, Leese, MI, Dorociak, K, et al. Feasibility of in-home sensor monitoring to detect mild cognitive impairment in aging military veterans: prospective observational study. JMIR Form Res 2020; 4: e16371.Google Scholar
Lyons, BE, Austin, D, Seelye, A, et al. Pervasive computing technologies to continuously assess Alzheimer’s disease progression and intervention efficacy. Front Aging Neurosci 2015; 7: 102.Google Scholar
Dodge, HH, Zhu, J, Mattek, NC, et al. Use of high-frequency in-home monitoring data may reduce sample sizes needed in clinical trials. PloS One 2015; 10: e0138095.Google Scholar
Food and Drug Administration. Digital Health Center of Excellence. Available at: www.fda.gov/medical-devices/digital-health-center-excellence (accessed November 2020).Google Scholar
Xue, C, Karjadi, C, Paschalidis, IC, Au, R, Kolachalama, VB. Detection of dementia on voice recordings using deep learning: a Framingham Heart Study. Alzheimers Res Ther 2021; 13: 146.Google Scholar
Inan, OT, Tenaerts, P, Prindiville, SA, et al. Digitizing clinical trials. NPJ Digit Med 2020; 3: 17.Google Scholar
Steinhubl, SR, Wolff-Hughes, DL, Nilsen, W, Iturriaga, E, Califf, RM. Digital clinical trials: creating a vision for the future. NPJ Digit Med 2019; 2: 13.Google Scholar
National Academies of Sciences, Engineering, and Medicine. The Role of Digital Health Technologies in Drug Development: Proceedings of a Workshop. Washington, DC: The National Academies Press; 2020.Google Scholar

References

Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement (N Y) 2019; 5: 272–93.Google Scholar
Food and Drug Administration. Project Facilitate: assisting healthcare providers with expanded access requests for investigational oncology products. Available at: www.fda.gov/about-fda/oncology-center-excellence/project-facilitate (accessed October 31, 2020).Google Scholar
Jarlow, JP, Moscicki, R. Impact of expanded access on FDA regulatory action and product labeling. Ther Innov Regul Sci 2017; 51: 12.Google Scholar
Food and Drug Administration. Expanded access to investigational drugs for treatment use: final rule. Fed Regist 2009; 74: 40900–45.Google Scholar
Food and Drug Administration. FDA backgrounder on FDAMA. Available at: www.fda.gov/regulatory-information/food-and-drug-administration-modernization-act-fdama-1997/fda-backgrounder-fdama (accessed October 31, 2020).Google Scholar
Food and Drug Administration. Expanded Access Program Report. 2018. Available at: www.fda.gov/media/119971/download (accessed October 31, 2020).Google Scholar
Center for Drug Evaluation and Research. CDER NextGen Portal. Available at: https://edm.fda.gov/ (accessed October 31, 2020).Google Scholar
Food and Drug Administration. 21st Century Cures Act. Available at: www.fda.gov/regulatory-information/selected-amendments-fdc-act/21st-century-cures-act (accessed October 31, 2020).Google Scholar
Center for Drug Evaluation and Research. Guidance for Industry: Expanded Access to Investigational Drugs for Treatment Use-Questions and Answers, Maryland: FDA; 2017.Google Scholar
Jarow, JP, Lurie, P, Crowley Ikenberry, S, Lemery, S. Overview of FDA’s expanded access program for investigational drugs. Ther Innov Regul Sci 2017; 51: 177–9.Google Scholar
Borysowski, J, Ehni, H-J, Górski, A. Ethics review in compassionate use. BMC Med 2017; 15: 136–42.Google Scholar
Food and Drug Administration. IRB review of individual patient expanded access requests for investigational drugs and biological products during the COVID-19 public health emergency. June 2020. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/institutional-review-board-irb-review-individual-patient-expanded-access-requests-investigational (accessed October 31, 2020).Google Scholar
Chapman, CR, Eckman, J, Bateman-House, AS. Oversight of right-to-try and expanded access requests for off-trial access to investigational drugs. Ethics Hum Res 2020; 42: 213.Google Scholar
Porcino, A, Shamseer, L, Chan, A-W, et al. SPIRIT extension and elaboration for n-of-1 trials: SPENT 2019 checklist. BMJ 2020; 368: m122.Google Scholar
Jarow, JP, Lemery, S, Bugin, K, Khozin, S, Moscicki, R. Expanded access of investigational drugs: the experience of the center of drug evaluation and research over a 10-year period. Ther Innov Regul Sci 2016; 50: 705–9.Google Scholar
McKee, AE, Markon, AO, Chan-Tack, KM, Lurie, P. How often are drugs made available under the Food and Drug Administration’s expanded access process approved? J Clin Pharmacol 2017; 57: S136–42.Google Scholar

References

Oxford, AE, Stewart, ES, Rohn, TT. Clinical trials in Alzheimer’s disease: a hurdle in the path of remedy. Int J Alzheimers Dis 2020; 2020: 5380346.Google Scholar
Murphy, MF, Sramek, JJ, Kurtz, NM, et al. Alzheimer’s Disease: Optimizing the Development of the Next Generation of Therapeutic Compounds. Oxford: Oxford University Press; 1998.Google Scholar
Cummings, J, Lee, G, Ritter, A, et al. AD drug development pipeline: 2019. Alzheimers Dement (NY) 2019; 5: 272–93.Google Scholar
Cummings, J, Aisen, P, DuBois, B, et al. Drug development in AD: the path to 2015. Alzheimers Res Ther 2016; 8: 39.Google Scholar
Satlin, A, Wang, J, Logovinsky, V, et al. Design of a Bayesian adaptive Phase 2 proof-of-concept trial for BAN2401, a putative disease-modifying monoclonal antibody for the treatment of Alzheimer’s disease. Alzheimers Dement (N Y) 2016; 2: 112.Google Scholar
House, A, Drosopoulou, NE, Riordan, HJ. Technology-assisted cohort optimization of early phase multi-centre patient studies. Int Pharmaceut Ind 2017; 9: 46–9.Google Scholar
Krafcik, BM, Doros, G, Malikova, MA. A single center analysis of factors influencing study start-up timeline in clinical trials. Future Sci OA 2017; 3: FS0223.Google Scholar
Babic, T, Riordan, HJ. Improving screen failure and recruitment rates in AD clinical trials. J Clin Stud 2016; 8: 3840.Google Scholar
Watson, JL, Ryan, L, Silverberg, N, et al. Obstacles and opportunities in Alzheimer’s clinical trial recruitment. Health Aff 2014; 33: 574–9.Google Scholar
Zupancic, B. Driving patient engagement in alzheimer disease clinical research to achieve trial success. World Pharma Today 2017. Available at: www.worldpharmatoday.com/Articles/driving-patient-engagement-in-alzheimer-s-disease-clinical-research-to-achieve-trial-success/ (accessed December 9, 2020).Google Scholar
Carbo, MA, Rock, C, Doyle, K, et al. An evaluation of independent subject eligibility review to ensure enrollment of high-quality appropriate subjects in mild Alzheimer’s disease. Alzheimer’s Association International Conference, Chicago, IL, July 2018.Google Scholar
World Health Organization. Handbook for good clinical research practice. Available at: https://apps.who.int/iris/handle/10665/43392 (accessed December 9, 2020).Google Scholar
Food and Drug Administration. Guidance for industry. Oversight of clinical investigations: a risk-based approach to monitoring. August 2013. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/oversight-clinical-investigations-risk-based-approach-monitoring (accessed December 9, 2020).Google Scholar
Kornsey, E, Friedmann, B, Bartolic, E, et al. An examination into the effects of five rater training modalities on the project conduct in international AD trials. International Society for CNS Clinical Trials and Methodology Meeting, Marina Del Ray, CA, Oct 1–3, 2012.Google Scholar
Avrumson, R, Carbo, MA, Riordan, HJ, et al. Effectiveness of rater training and data surveillance in Alzheimer disease clinical trials. Clinical Trials in Alzheimer’s Disease (CTAD) Congress, Boston, MA, November 2017.Google Scholar
Riordan, HJ, Perakslis, E, Roosz, S, et al. Utilizing large data sets and extended trial observation to close the AD evidence gap. J Clin Stud 2019; 11: 4650.Google Scholar
Perakslis, E, Riordan, H, Friedhoff, L, et al. A call for a global ‘bigger’ data approach to Alzheimer disease. Nat Rev Drug Discov 2019; 18: 319–20.Google Scholar

References

Food and Drug Administration. Peripheral and Central Nervous System Drugs Advisory Committee meeting. Available at: www.fda.gov/advisory-committees/human-drug-advisory-committees/peripheral-and-central-nervous-system-drugs-advisory-committee (accessed January 2021).Google Scholar
European Medicines Agency. Ebixa (memantine). Available at: www.ema.europa.eu/en/medicines/human/EPAR/ebixa (accessed January 2021).Google Scholar
Chinese National Medical Products Agency. Treatment for Alzheimer’s available by end of 2019. Available at: http://english.nmpa.gov.cn/2019-11/04/c_422037.htm (accessed January 2021).Google Scholar
Food and Drug Administration. Alzheimer’s disease: developing drugs for treatment. Guidance for industry. 2018. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/alzheimers-disease-developing-drugs-treatment-guidance-industy (accessed January 2021).Google Scholar
European Medicines Agency. Guideline on the clinical investigation of medicines to treat Alzheimer’s disease, 2018. Available at: www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-investigation-medicines-treatment-alzheimers-disease-revision-2_en.pdf (accessed January 2021).Google Scholar
The University of Tokyo Hospital. Project to promote the development of innovative pharmaceuticals, medical devices, and regenerative medical products (Ministry of Health, Labour, and Welfare) regulatory science research for the establishment of criteria for clinical evaluation of drugs for Alzheimer’s disease, issues to consider in the clinical evaluation and development of drugs for Alzheimer’s disease. Available at: www.pmda.go.jp/files/000221585.pdf (accessed January 2021).Google Scholar
Jack, CR, Bennett, DA, Blennow, K, et al. NIA–AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14: 535–62.Google Scholar
European Medicines Agency. Connecting the dots towards global knowledge of the international medicine regulatory landscape: mapping of international initiatives. Available at: www.ema.europa.eu/en/documents/leaflet/connecting-dots-towards-global-knowledge-international-medicine-regulatory-landscape-mapping_en.pdf (accessed January 2021).Google Scholar
Food and Drug Administration. 21st Century Cures Act. Available at: www.fda.gov/regulatory-information/selected-amendments-fdc-act/21st-century-cures-act (accessed January 2021).Google Scholar
European Medicines Agency. EU Medicines Agencies network strategy to 2020, working together to improve health. Available at: www.ema.europa.eu/en/documents/other/eu-medicines-agencies-network-strategy-2020-working-together-improve-health_en.pdf (accessed January 2021).Google Scholar
European Medicines Agency. Regulatory science strategy 2025. Available at: www.ema.europa.eu/en/about-us/how-we-work/regulatory-science-strategy#regulatory-science-strategy-to-2025-section (accessed January 2021).Google Scholar
National Medical Products Administration (NMPA). http://english.nmpa.gov.cn/drugs.html (accessed January 2021).Google Scholar
Engen, S. The improving Japanese pharmaceutical regulatory environment. Available at: Locustwalk.com/the-improving-japanese-pharmaceutical-regulatory-environment/ (accessed January 2021).Google Scholar
Food and Drug Administration. Investigational new drug (IND) application. Available at: www.fda.gov/drugs/types-applications/investigational-new-drug-ind-application (accessed January 2021).Google Scholar
Food and Drug Administration. Formal meetings between the FDA and sponsors or applicants of PDUFA products guidance for industry, December 2017. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/formal-meetings-between-fda-and-sponsors-or-applicants-pdufa-products-guidance-industry (accessed January 2021).Google Scholar
Food and Drug Administration. Special protocol assessment guidance for industry, April 2018. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/special-protocol-assessment-guidance-industry (accessed January 2021).Google Scholar
Food and Drug Administration. Expedited programs for serious conditions: drugs and biologics, May 2014. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/expedited-programs-serious-conditions-drugs-and-biologics (accessed January 2021).Google Scholar
European Medicines Agency. Clinical trial regulation. Available at: www.ema.europa.eu/en/human-regulatory/research-development/clinical-trials/clinical-trial-regulation (accessed January 2021).Google Scholar
European Medicines Agency. Scientific advice and protocol assistance. Available at: www.ema.europa.eu/en/human-regulatory/research-development/scientific-advice-protocol-assistance (accessed January 2021).Google Scholar
European Medicines Agency. Adaptive pathways. Available at: www.ema.europa.eu/en/human-regulatory/research-development/adaptive-pathways (accessed January 2021).Google Scholar
European Medicines Agency. PRIME: priority medicines. Available at: www.ema.europa.eu/en/human-regulatory/research-development/prime-priority-medicines (accessed January 2021).Google Scholar
European Medicines Agency. Conditional marketing authorization. Available at: www.ema.europa.eu/en/human-regulatory/marketing-authorisation/conditional-marketing-authorisation (accessed January 2021).Google Scholar
European Medicines Agency. Exceptional circumstances. Available at: www.ema.europa.eu/en/glossary/exceptional-circumstances (accessed January 2021).Google Scholar
Japan Pharmaceutical Manufacturers Association. Pharmaceutical Administration and Regulations in Japan. Available at: www.jpma.or.jp/english/parj/ (accessed January 2021).Google Scholar
Kajiwara, E, Shikano, M. Considerations and regulatory challenges for innovative medicines in expedited approval programs: breakthrough therapy and Sakigake designation. Ther Innov Regul Sci 2020; 54: 814–20.Google Scholar
Kondo, H, Sugita, T, Ida, N, Fukushima, H, Yasuda, N. A comparison of PMDA and EMA consultations for regulatory and scientific matters in drugs and regenerative medicine products. Ther Innov Regul Sci 2017; 51: 355–9.Google Scholar
Zhuxing, Y, Haixue, W. The regulatory requirements and critical points of drug clinical trials registration in China. Available at: www.appliedclinicaltrialsonline.com/view/regulatory-requirements-and-key-points-drug-clinical-trials-registration-china (accessed January 2021).Google Scholar
Jianqing, C. What’s new for conditional approval of drugs in China? Tigermed Insight. Available at: https://tigermedgrp.com/whats-new-for-conditional-approval-of-drugs-in-china/ (accessed January 2021).Google Scholar
Covington and Burling. China promulgates revised drug registration regulation, August 2020. Available at: www.cov.com/-/media/files/corporate/publications/2020/04/china-promulgates-revised-drug-registration-regulation.pdf (accessed January 2021).Google Scholar
Centre for Innovation and Regulatory Science. CIRS RD Briefing 77: new drug approvals in six major authorities, June 2020. Available at: www.cirsci.org/publications/cirs-rd-briefing-77-new-drug-approvals-in-six-major-authorities/ (accessed January 2021).Google Scholar
Xu, W, Yuanyuan, D. New drug approvals in China in 2019. Available at: https://globalforum.diaglobal.org/issue/may-2020/new-drug-approvals-in-china-in-2019/ (accessed January 2021).Google Scholar
Food and Drug Administration. Qualification process for drug development tools guidance for industry and FDA staff, November, 2020. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/qualification-process-drug-development-tools-guidance-industry-and-fda-staff (accessed January 2021).Google Scholar
European Medicines Agency. Qualification of novel methodologies for medicine development. Available at: www.ema.europa.eu/en/human-regulatory/research-development/scientific-advice-protocol-assistance/qualification-novel-methodologies-medicine-development-0 (accessed January 2021).Google Scholar
Food and Drug Administration. About biomarkers and qualification. Available at: www.fda.gov/drugs/biomarker-qualification-program/about-biomarkers-and-qualification (accessed January 2021).Google Scholar
European Medicines Agency. Opinions and letters of support on the qualification of novel methodologies for medicine development. Available at: www.ema.europa.eu/en/human-regulatory/research-development/scientific-advice-protocol-assistance/novel-methodologies-biomarkers/opinions-letters-support-qualification-novel-methodologies-medicine-development (accessed January 2021).Google Scholar
Arnerić, SP, Batrla-Utermann, R, Beckett, L, et al. Cerebrospinal fluid biomarkers for Alzheimer’s disease: a view of the regulatory science qualification landscape from the Coalition Against Major Diseases CSF Biomarker Team. J Alzheimers Dis 2017; 55: 1935.Google Scholar
Sauer, JM, Porter, AC; Biomarker Programs, Predictive Safety Testing Consortium. Preclinical biomarker qualification. Exp Biol Med (Maywood) 2018; 243: 2227.Google Scholar
Pacifici, E, Bain, S (eds.). An Overview of FDA Regulated Products: From Drugs and Cosmetics to Food and Tobacco. Cambridge, MA: Academic Press; 2018.Google Scholar
Van Norman, GA. Drugs and devices: comparison of European and US approval processes. J Am Coll Cardiol Basic Transl Sci 2016; 1: 399412.Google Scholar
Food and Drug Administration. Benefit–risk assessment in drug regulatory decision making. Draft PDUFA VI implementation plan (FY 2018-2022). Available at: www.fda.gov/files/about%20fda/published/Benefit-Risk-Assessment-in-Drug-Regulatory-Decision-Making.pdf (accessed January 2021).Google Scholar
Juhaeri, J. Benefit–risk evaluation: the past, present and future. Ther Adv Drug Saf 2019; 10: 2042098619871180;DOI: http://doi.org/10.1177/2042098619871180.Google Scholar
European Medicines Agency. Benefit–risk methodology. Available at: www.ema.europa.eu/en/about-us/support-research/benefit-risk-methodology (accessed January 2021).Google Scholar
Morant, AV, Vestergaard, HT, Blædel-Lassen, A, Navikas, V. US, EU, and Japanese regulatory guidelines for development of drugs for treatment of Alzheimer’s disease: implications for global drug development. Clin Transl Sci 2020; 13: 652–64.Google Scholar
Cummings, J, Lee, G, Ritter, A, Sabbagh, M, Zhong, K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (N Y) 2020; 6: e12050.Google Scholar

References

Hurd, MD, Martorell, P, Delavande, A, Mullen, KJ, Langa, KM. Monetary costs of dementia in the United States. N Engl J Med 2013; 368: 1326–34.Google Scholar
Largent, EA, Karlawish, J, Grill, JD. Study partners: essential collaborators in discovering treatments for Alzheimer’s disease. Alzheimers Res Ther 2018; 10: 101.Google Scholar
Knopman, DS. Clinical trial design issues in mild to moderate Alzheimer disease. Cogn Behav Neurol 2008; 21: 197201.Google Scholar
Grill, JD, Karlawish, J. Addressing the challenges to successful recruitment and retention in Alzheimer’s disease clinical trials. Alzheimers Res Ther 2010; 2: 34.Google Scholar
Schneider, LS, Olin, JT, Lyness, SA, Chui, HC. Eligibility of Alzheimer’s disease clinic patients for clinical trials. J Am Geriatr Soc 1997; 45: 923–8.Google Scholar
Karlawish, JH, Casarett, D, Klocinski, J, Sankar, P. How do AD patients and their caregivers decide whether to enroll in a clinical trial? Neurology 2001; 56: 789–92.Google Scholar
Karlawish, JH, Casarett, D, Propert, KJ, James, BD, Clark, CM. Relationship between Alzheimer’s disease severity and patient participation in decisions about their medical care. J Am Geriatr Soc Neurol 2002; 15: 6872.Google Scholar
Cox, CG, Ryan, BAM, Gillen, DL, Grill, JD. preliminary, A study of clinical trial enrollment decisions among people with mild cognitive impairment and their study partners. Am J Geriatr Psychiatry 2019; 27: 322–32.Google Scholar
Cox, CG, Ryan, MM, Gillen, D, Grill, JD. Is reluctance to share Alzheimer’s disease biomarker status with a study partner a barrier to preclinical trial recruitment? J Prev Alzheimers Dis 2021; 8: 52–8.Google Scholar
10. Karlawish, JH, Casarett, DJ, James, BD. Alzheimer’s disease patients’ and caregivers’ capacity, competency, and reasons to enroll in an early-phase Alzheimer’s disease clinical trial. J Am Geriatr Soc 2002; 50: 2019–24.Google Scholar
Okonkwo, O, Griffith, HR, Belue, K, et al. Medical decision-making capacity in patients with mild cognitive impairment. Neurology 2007; 69: 1528–35.Google Scholar
Jefferson, AL, Lambe, S, Moser, DJ, et al. Decisional capacity for research participation in individuals with mild cognitive impairment. J Am Geriatr Soc 2008; 56: 1236–43.Google Scholar
Okonkwo, OC, Griffith, HR, Copeland, JN, et al. Medical decision-making capacity in mild cognitive impairment: a 3-year longitudinal study. Neurology 2008; 71: 1474–80.Google Scholar
National Research Council (US) Panel on Handling Missing Data in Clinical Trials. The Prevention and Treatment of Missing Data in Clinical Trials. Washington, DC: National Academies Press; 2010.Google Scholar
Grill, JD, Kwon, J, Teylan, MA, et al. Retention of Alzheimer disease research participants. Alzheimer Dis Assoc Disord 2019; 33: 299306.Google Scholar
Robinson, KA, Dinglas, VD, Sukrithan, V, et al. Updated systematic review identifies substantial number of retention strategies: using more strategies retains more study participants. J Clin Epidemiol 2015; 68: 1481–7.Google Scholar
Kozauer, N, Katz, R. Regulatory innovation and drug development for early-stage Alzheimer’s disease. N Engl J Med 2013; 368: 1169–71.Google Scholar
Leber, P. Observations and suggestions on antidementia drug development. Alzheimer Dis Assoc Disord 1996; 10: 31–5.Google Scholar
Amariglio, RE, Donohue, MC, Marshall, GA, et al. Tracking early decline in cognitive function in older individuals at risk for Alzheimer disease dementia: the Alzheimer’s Disease Cooperative Study Cognitive Function Instrument. JAMA Neurol 2015; 72: 446–54.Google Scholar
Ryan, MM, Grill, JD, Gillen, DL, Alzheimer’s Disease Neuroimaging Initiative. Participant and study partner prediction and identification of cognitive impairment in preclinical Alzheimer’s disease: study partner vs. participant accuracy. Alzheimers Res Ther 2019; 11: 85.Google Scholar
Grill, JD, Karlawish, J. Study partners should be required in preclinical Alzheimer’s disease trials. Alzheimers Res Ther 2017; 9: 93.Google Scholar
Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Grill, JD, Raman, R, Ernstrom, K, Aisen, P, Karlawish, J. Effect of study partner on the conduct of Alzheimer disease clinical trials. Neurology 2013; 80: 282–8.Google Scholar
Grill, JD, Raman, R, Ernstrom, K, et al. Comparing recruitment, retention, and safety reporting among geographic regions in multinational Alzheimer’s disease clinical trials. Alzheimers Res Ther 2015; 7: 39.Google Scholar
Grill, JD, Monsell, S, Karlawish, J. Are patients whose study partners are spouses more likely to be eligible for Alzheimer’s disease clinical trials? Dement Geriatr Cogn Disord 2012; 33: 334–40.Google Scholar
Bernard, MA, Clayton, JA, Lauer, MS. Inclusion across the lifespan: NIH policy for clinical research. JAMA 2018; 320: 1535–6.Google Scholar
Conde-Sala, JL, Garre-Olmo, J, Turro-Garriga, O, Vilalta-Franch, J, Lopez-Pousa, S. Differential features of burden between spouse and adult-child caregivers of patients with Alzheimer’s disease: an exploratory comparative design. Int J Nurs Stud 2010; 47: 1262–73.Google Scholar
Cary, MS, Rubright, JD, Grill, JD, Karlawish, J. Why are spousal caregivers more prevalent than nonspousal caregivers as study partners in AD dementia clinical trials? Alzheimer Dis Assoc Disord 2015; 29: 70–4.Google Scholar
Zhou, Y, Elashoff, D, Kremen, S, et al. African Americans are less likely to enroll in preclinical Alzheimer’s disease clinical trials. Alzheimers Dement (N Y) 2017; 3: 5764.Google Scholar
Sperling, RA, Rentz, DM, Johnson, KA, et al. The A4 study: stopping AD before symptoms begin? Sci Transl Med 2014; 6: 228fs13.Google Scholar
Berstein, OM, Grill, JD, Gillen, DL. Recruitment and retention of participant and study partner dyads in two multinational Alzheimer’s disease registration trials. Alzheimers Res Ther 2021; 13: 16.Google Scholar
Edland, SD, Emond, JA, Aisen, PS, Petersen, RC. NIA-funded Alzheimer centers are more efficient than commercial clinical recruitment sites for conducting secondary prevention trials of dementia. Alzheimer Dis Assoc Disord 2010; 24: 159–64.Google Scholar
Conde-Sala, JL, Garre-Olmo, J, Turro-Garriga, O, Vilalta-Franch, J, Lopez-Pousa, S. Quality of life of patients with Alzheimer’s disease: differential perceptions between spouse and adult child caregivers. Dement Geriatr Cogn Disord 2010; 29: 97108.Google Scholar
Grill, JD, Karlawish, J. Consider the source: the implications of informant type on outcome assessments. Alzheimer Dis Assoc Disord 2015; 29: 364.Google Scholar
Ready, RE, Ott, BR, Grace, J. Validity of informant reports about AD and MCI patients’ memory. Alzheimer Dis Assoc Disord 2004; 18: 11–16.Google Scholar
Cacchione, PZ, Powlishta, KK, Grant, EA, Buckles, VD, Morris, JC. Accuracy of collateral source reports in very mild to mild dementia of the Alzheimer type. J Am Geriatr Soc 2003; 51: 819–23.Google Scholar
Grill, JD, Zhou, Y, Karlawish, J, Elashoff, D. Frequency and impact of informant replacement in Alzheimer disease research. Alzheimer Dis Assoc Disord 2015; 29: 242–8.Google Scholar
Nuno, MM, Gillen, DL, Grill, JD, et al. Study partner types and prediction of cognitive performance: implications to preclinical Alzheimer’s trials. Alzheimers Res Ther 2019; 11: 92.Google Scholar
Karlawish, J, Cary, MS, Rubright, J, Tenhave, T. How redesigning AD clinical trials might increase study partners’ willingness to participate. Neurology 2008; 71: 1883–8.Google Scholar
Gelinas, L, Largent, EA, Cohen, IG, et al. Framework for ethical payment to research participants. N Engl J Med 2018; 378: 766–71.Google Scholar

References

Jack, CR, Bennett, DA, Blennow, K, et al. NIA–AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14: 535–62.Google Scholar
Petersen, RC, Lopez, O, Armstrong, MJ, et al. Practice guideline update summary: mild cognitive impairment: report of the Guideline Development, Dissemination, and Implementation subcommittee of the American Academy of Neurology. Neurology 2018; 90: 126–35.Google Scholar
Liu, JL, Hlávka, JP, Hillestad, R, Mattke, S. Assessing the Preparedness of the U.S. health care system infrastructure for an Alzheimer’s treatment. Available at: www.rand.org/pubs/research_reports/RR2272.html (accessed July 9, 2021).Google Scholar
Hlavka, JP, Mattke, S, Liu, JL. Assessing the preparedness of the health care system infrastructure in six European countries for an Alzheimer’s treatment. Rand Health Q 2019; 8: 2.Google Scholar
Hebert, LE, Weuve, J, Scherr, PA, Evans, DA. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology 2013; 80: 1778–83.Google Scholar
Sado, M, Ninomiya, A, Shikimoto, R, et al. The estimated cost of dementia in Japan, the most aged society in the world. PLoS One 2018; 13: e0206508.Google Scholar
Lang, L, Clifford, A, Wei, L, et al. Prevalence and determinants of undetected dementia in the community: a systematic literature review and a meta-analysis. BMJ Open 2017; 7: e011146.Google Scholar
Sperling, RA, Rentz, DM, Johnson, KA, et al. The A4 study: stopping AD before symptoms begin? Sci Transl Med 2014; 6: 228fs13.Google Scholar
Baxi, SM, Girosi, F, Liu, JL. Assessing the preparedness of the Australian health care system infrastructure for an Alzheimer’s disease-modifying therapy. Available at: www.rand.org/pubs/research_reports/RR2891.html (accessed July 9, 2021).Google Scholar
Liu, JL, Hlavka, JP, Coulter, DT, et al. Assessing the preparedness of the Canadian health care system infrastructure for an Alzheimer’s treatment. Available at: www.rand.org/pubs/research_reports/RR2744.html (accessed July 9, 2021).Google Scholar
Mattke, HJ, Yoong, J, Wang, M, Goto, R. Assessing the preparedness of the Japanese health care system infrastructure for an Alzheimer’s treatment. Available at: https://cesr.usc.edu/sites/default/files/CESR%202019-101.pdf (accessed July 9, 2021) .Google Scholar
Hankyung Jun, SKC, Yoong, J, Mattke, S. Assessing the preparedness of the Korean healthcare system infrastructure for an Alzheimer’s treatment. Available at: https://cesr.usc.edu/sites/default/files/Korea_Infrastructure_Projection_final.pdf (accessed July 9, 2021) .Google Scholar
Hankyung Jun, SKC, Yoong, J, Mattke, S. Assessing the preparedness of the Taiwanese healthcare system infrastructure for an Alzheimer’s treatment. Available at: https://cesr.usc.edu/sites/default/files/Taiwan_Infrastructure_Projection_final.pdf (accessed July 9, 2021).Google Scholar
Ritchie, CW, Russ, TC, Banerjee, S, et al. The Edinburgh Consensus: preparing for the advent of disease-modifying therapies for Alzheimer’s disease. Alzheimers Res Ther 2017; 9: 85.Google Scholar
Martínez-Lagea, P, Martín-Carrascob, M, Arrietad, E, Rodrigof, J, Formigac, F. Map of Alzheimer’s disease and other dementias in Spain. MapEA Project. Rev Esp Geriatr Gerontol 2018; 53: 2637.Google Scholar
Jun, H, Cho, SK, Aliyev, ER, Mattke, S, Suen, S-C. How much value would a treatment for Alzheimer’s disease offer? Cost-effectiveness thresholds for pricing a disease-modifying therapy. Curr Alzheimer Res 2020; 17 : 819–22.Google Scholar
Mattke, S, Cho, SK, Bittner, T, Hlavka, J, Hanson, M. Blood-based biomarkers for Alzheimer’s pathology and the diagnostic process for a disease-modifying treatment: projecting the impact on the cost and wait times. Alzheimers Dement (Amst) 2020; 12: e12081.Google Scholar
Ngandu, T, Lehtisalo, J, Solomon, A, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet 2015; 385: 2255–63.Google Scholar
Rabinovici, GD, Gatsonis, C, Apgar, C, et al. Association of amyloid positron emission tomography with subsequent change in clinical management among Medicare beneficiaries with mild cognitive impairment or dementia. JAMA 2019; 321: 1286–94.Google Scholar
Russell-Babin, K, Wurmser, T. Transforming care through top-of-license practice. Nurs Manage 2016; 47: 24–8.Google Scholar
Mattke, S, Hoch, E. Borrowing for the cure. Available at: www.rand.org/pubs/perspectives/PE141.html (accessed July 9, 2021).Google Scholar
Lam, J, Jun, H, Cho, S K, Hanson, M, Mattke, S. Projection of budgetary savings to US state Medicaid programs from reduced nursing home use due to an Alzheimer’s disease treatment. Alzheimers Dement (Amst) 2021; 13: e12159.Google Scholar
Greaves, I, Greaves, N, Walker, E, et al. Gnosall Primary Care Memory Clinic: eldercare facilitator role description and development. Dementia (London) 2015; 14: 389408.Google Scholar
Engedal, K, Gausdal, M, Gjora, L, Haugen, PK. Assessment of dementia by a primary health care dementia team cooperating with the family doctor: the Norwegian model. Dement Geriatr Cogn Disord 2012; 34: 263–70.Google Scholar
Lee, L, Hillier, LM, Heckman, G, et al. Primary care-based memory clinics: expanding capacity for dementia care. Can J Aging 2014; 33: 307–19.Google Scholar
Alzheimer’s Association. The Alzheimer’s and Dementia Care ECHO® Program. Available at: www.alz.org/professionals/professional-providers/echo-alzheimers-dementia-care-program (accessed June 6, 2019).Google Scholar
Lam, J, Hlávka, J, Mattke, S. The potential emergence of disease-modifying treatments for Alzheimer disease: the role of primary care in managing the patient journey. J Am Board Fam Med 2019; 32: 931–40.Google Scholar
Palmqvist, S, Janelidze, S, Stomrud, E, et al. Performance of fully automated plasma assays as screening tests for Alzheimer disease-related beta-amyloid status. JAMA Neurol 2019; 76: 1060–9.Google Scholar
Palmqvist, S, Janelidze, S, Quiroz, YT, et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 2020; 324: 772.Google Scholar
Kourtis, LC, Regele, OB, Wright, JM, Jones, GB. Digital biomarkers for Alzheimer’s disease: the mobile/wearable devices opportunity. NPJ Digit Med 2019; 2: 9.Google Scholar
Buegler, M, Harms, RL, Balasa, M, et al. Digital biomarker-based individualized prognosis for people at risk of dementia. Alzheimers Dement (Amst) 2020; 12: e12073.Google Scholar
Biogen. Biogen to launch pioneering study to develop digital biomarkers of cognitive health using apple watch and iphone [press release]. Available at: https://investors.biogen.com/news-releases/news-release-details/biogen-launch-pioneering-study-develop-digital-biomarkers (accessed January 2021).Google Scholar
Arnett, DK, Blumenthal, RS, Albert, MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease. J Am Coll Cardiol 2019; 74: e177232.Google Scholar
Progetto Interceptor. Interceptor: the project. Available at: www.interceptorproject.com/en/lo-studio-interceptor/ (accessed July 9, 2021).Google Scholar
Belleville, S, Leblanc, AC, Kergoat, M-J, et al. The Consortium for the early identification of Alzheimer’s disease – Quebec (CIMA-Q). Alzheimers Dement (Amst) 2019; 11: 787–96.Google Scholar
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
Mattke, S, Klautzer, L, Mengistu, T. Medicines as a service. Available at: www.rand.org/pubs/occasional_papers/OP381.html (accessed July 9, 2021).Google Scholar

References

World Health Organization. WHO mortality database. Available at: www.who.int/healthinfo/mortality_data/en/ (accessed June 1, 2021).Google Scholar
Alzheimer’s Association. 2020 Alzheimer’s disease facts and figures. Alzheimers Dement 2020; 16: 391460.Google Scholar
Hebert, LE, Weuve, J, Scherr, PA, et al. Alzheimer disease in the United States (2010–2050) estimated using the 2010 Census. Neurology 2013; 80: 1778–83.Google Scholar
Lao, K, Ji, N, Zhang, X, et al. Drug development for Alzheimer’s disease: review. J Drug Target 2019; 27: 164–73.Google Scholar
Cummings, J, Ritter, A, Zhong, K. Clinical trials for disease-modifying therapies in Alzheimer’s disease: a primer, lessons learned, and a blueprint for the future. J Alzheimers Dis 2018; 64: S322.Google Scholar
Cummings, J, Feldman, HH, Scheltens, P, et al. The “rights” of precision drug development for Alzheimer’s disease. Alzheimers Res Ther 2019; 11: 76.Google Scholar
Tuttle, KR. Impact of the COVID-19 pandemic on clinical research. Nat Rev Nephrol 2020; 16: 562–4.Google Scholar
Asaad, M, Khan Habibullah, K, Butler, CE, et al. The impact of COVID-19 on clinical trials. Ann Surg 2020; 272: e222–3.Google Scholar
Applied Clinical Trials. Findings from a Tufts study examining the effects of COVID-19 on clinical trials. Available at:www.appliedclinicaltrialsonline.com/view/covid-19-and-its-impact-on-the-future-of-clinical-trial-execution (accessed December 20, 2020).Google Scholar
Sathian, B, Asim, M, Banerjee, I, et al. Impact of COVID-19 on clinical trials and clinical research: a systematic review. Nepal J Epidemiol 2020; 10: 878–87.Google Scholar
Food and Drug Administration. FDA guidance on conduct of clinical trials of medical products during COVID-19 public health emergency. March 2020. Available at: www.fda.gov/regulatory-information/search-fda-guidance-documents/fda-guidance-conduct-clinical-trials-medical-products-during-covid-19-public-health-emergency (accessed May 20, 2020).Google Scholar
Medicines and Healthcare products Regulatory Agency Inspectorate. Advice for management of clinical trials in relation to Coronavirus. March 12, 2020. Available at: https://mhrainspectorate.blog.gov.uk/2020/03/12/advice-for-management-of-clinical-trials-in-relation-to-coronavirus/ (accessed June 1, 2021).Google Scholar
European Medicines Agency. Guidance on the management of clinical trials during the COVID-19 (Coronavirus) pandemic. Version 3. April 28, 2020. Available at: https://ecrin.org (accessed June 1, 2021).Google Scholar
Australian Government Agency:Therapeutic goods administration. Clinical trial processes: information relating to COVID-19. March 31, 2020. Available at: www.tga.gov.au/clinical-trial-processes (accessed May 30, 2021).Google Scholar
Comas-Herrera, A, Zalakain, J. Mortality associated with COVID-19 outbreaks in care homes: early international evidence. April 12, 2020. Available at: https://ltccovid.org/2020/04/12/mortality-associated-with-covid-19-outbreaks-in-care-homes-early-international-evidence/ (accessed May 30, 2021).Google Scholar
Altieri, M, Santangelo, G. The psychological impact of COVID-19 pandemic and lockdown on caregivers of people with dementia. Am J Geriatr Psychiatry 2021; 29: 2734.Google Scholar
UsAgainstAlzheimer’s. UsAgainstAlzheimer’s survey on COVID-19 and Alzheimer’s community summary of findings for June 2020 survey (Survey #4). Available at: www.usagainstalzheimers.org/sites/default/files/2020-06/UsA2%20COVID%20Survey%204%20Summary%206.25.20.pdf (accessed June 1, 2021).Google Scholar
Anker, SD, Butler, J, Khan, MS, et al. Conducting clinical trials in heart failure during (and after) the COVID-19 pandemic: an Expert Consensus Position Paper from the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur Heart J 2020; 41: 2109–17.Google Scholar
Medidata. COVID-19 and clinical trials: the Medidata perspective. Available at: www.medidata.com/wp-content/uploads/2020/08/COVID19-Response8.0_Clinical-Trials_2020824_v1.pdf (accessed Nov 29, 2020).Google Scholar
Kennedy, M, Helfand, BKI, Gou, RY, et al. Delirium in older patients with COVID-19 presenting to the emergency department. JAMA Netw Open 2020; 3: e2029540.Google Scholar
Lai, FH-Y, Yan, EW, Yu, KK, et al. The protective impact of telemedicine on persons with dementia and their caregivers during the COVID-19 pandemic. Am J Geriatr Psychiatry 2020; 28: 1175–84.Google Scholar
Saini, KS, de Las Heras, B, de Castro, J, et al. Effect of the COVID-19 pandemic on cancer treatment and research. Lancet Haematol 2020; 7: e4325.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
×