Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-24T22:24:55.466Z Has data issue: false hasContentIssue false

11 - Phase 1 Trials in Alzheimer’s Disease Drug Development

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 1 clinical trials are the entrance to the further clinical development of new compounds. The chapter describes the regulatory background and highlights most important issues about selection of the maximum recommended starting dose, dose escalation steps, and definition of maximum tolerated dose, or maximum applied dose in a study considering actual guidelines. There is an overview about selection of subject populations and frequently used trial designs. The principles of single-ascending-dose and multiple-ascending-dose tolerance studies are described with a few examples of studies in Alzheimer’s disease (AD). The safety assessment is important in clinical practice, as AD drugs will be used over many years, so excellent tolerability is a must! In Phase 1, a careful assessment of pharmacokinetic (PK) properties of a new compound forms the basis for dose selection in Phase 2 and 3 studies and supports the decision on the treatment regimen. The importance of inclusion of different biomarkers in these studies to allow assessment of pharmacodynamic and PK relationship and to potentially identify first signals in human studies indicating therapeutic usefulness in the indication.

Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 135 - 149
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

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.Google 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.Google Scholar
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.Google 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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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 Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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 Scholar
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.CrossRefGoogle 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 ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Ebert, U, Kirch, W. Scopolamine model of dementia: electroencephalogram findings and cognitive performance. Eur J Clin Invest 1998; 28: 944–9.Google Scholar
Adler, G, Brassen, S. Short-term rivastigmine treatment reduces EEG slow-wave power in Alzheimer patients. Neuropsychobiology 2001; 43: 273–6.Google Scholar
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 Scholar
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.CrossRefGoogle ScholarPubMed
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

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
×