Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-2xdlg Total loading time: 0 Render date: 2024-06-17T08:58:43.012Z Has data issue: false hasContentIssue false

12 - The Importance of Phase 2 in Drug Development for Alzheimer’s Disease

from 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

Phase 2 in drug development is a crucial phase that can make or break success. The goals in Phase 2 are to determine safety, dosage and efficacy. In this chapter elements of planning, design, biomarker use and clinical outcomes are highlighted and some good and bad examples are given, emphasizing the importance of conducting a proper Phase 2.

Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 150 - 161
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

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.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.CrossRefGoogle Scholar
Cummings, J, Feldman, HH, Scheltens, P. The “rights” of precision drug development for Alzheimer’s disease. Alzheimers Res Ther 2019; 11: 76.CrossRefGoogle 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.Google Scholar
Molinuevo, JL, Ayton, S, Batrla, R, et al. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol 2018; 136: 821–53.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle Scholar
Sevigny, J, Chiao, P, Bussière, T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016; 537: 50–6.CrossRefGoogle ScholarPubMed
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 ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Soininen, H, Solomon, A, Visser, PJ, et al. 36-month LipiDiDiet multinutrient clinical trial in prodromal Alzheimer’s disease. Alzheimers Dement 2021; 17: 2940.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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 ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
Bilder, RM, Reise, SP. Neuropsychological tests of the future: how do we get there from here? Clin Neuropsychologist 2019; 33: 220–45.CrossRefGoogle 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 ScholarPubMed
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 ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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 ScholarPubMed
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.CrossRefGoogle ScholarPubMed

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
×