Evaluating the Safety and Efficacy of Crenezumab vs Placebo in Adults With Early Alzheimer Disease: Two Phase 3 Randomized Placebo-Controlled Trials

Susanne Ostrowitzki, Tobias Bittner, Kaycee M Sink, Howard Mackey, Christina Rabe, Lawrence S Honig, Emanuele Cassetta, Michael Woodward, Mercè Boada, Christopher H van Dyck, Timo Grimmer, Dennis J Selkoe, Andres Schneider, Kathleen Blondeau, Nan Hu, Angelica Quartino, David Clayton, Michael Dolton, Yifan Dang, Beth Ostaszewski, Sandra M Sanabria-Bohórquez, Michael Rabbia, Balazs Toth, Udo Eichenlaub, Jillian Smith, Lee A Honigberg, Rachelle S Doody, Susanne Ostrowitzki, Tobias Bittner, Kaycee M Sink, Howard Mackey, Christina Rabe, Lawrence S Honig, Emanuele Cassetta, Michael Woodward, Mercè Boada, Christopher H van Dyck, Timo Grimmer, Dennis J Selkoe, Andres Schneider, Kathleen Blondeau, Nan Hu, Angelica Quartino, David Clayton, Michael Dolton, Yifan Dang, Beth Ostaszewski, Sandra M Sanabria-Bohórquez, Michael Rabbia, Balazs Toth, Udo Eichenlaub, Jillian Smith, Lee A Honigberg, Rachelle S Doody

Abstract

Importance: Alzheimer disease (AD), a neurodegenerative disease characterized by β-amyloid plaques and τ tangles in the brain, represents an unmet medical need with no fully approved therapeutics to modify disease progression.

Objective: To investigate the safety and efficacy of crenezumab, a humanized monoclonal immunoglobulin G4 antibody targeting β-amyloid oligomers, in participants with prodromal to mild (early) AD.

Design, setting, and participants: Two phase 3 multicenter randomized double-blind placebo-controlled parallel-group efficacy and safety studies of crenezumab in participants with early AD, CREAD and CREAD2, were initiated in 2016 and 2017, respectively, and were designed to evaluate the efficacy and safety of crenezumab in participants with early AD. CREAD (194 sites in 30 countries) and CREAD2 (209 sites in 27 countries) were global multicenter studies. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. Both trials enrolled individuals aged 50 to 85 years with early AD. Participants with some comorbidities and evidence of cerebral infarction or more than 4 microbleeds or areas of leptomeningeal hemosiderosis on magnetic resonance imaging were excluded. After 2923 and 2858 were excluded, respectively, 813 participants in CREAD and 806 in CREAD2 were randomly assigned in a 1:1 ratio to either placebo or crenezumab. In the final analysis, there were 409 participants in the placebo group and 404 in the crenezumab group in CREAD and 399 in the placebo group and 407 in the crenezumab group in CREAD2. Data were analyzed up until January 2019 and August 2019, respectively.

Interventions: Participants received placebo or 60 mg/kg crenezumab intravenously every 4 weeks for up to 100 weeks.

Main outcomes and measures: The primary outcome was change from baseline to week 105 in Clinical Dementia Rating-Sum of Boxes (CDR-SB) score.

Results: There were 813 participants in CREAD (mean [SD] age, 70.7 [8.2] years; 483 female and 330 male) and 806 in CREAD2 (mean [SD] age, 70.9 [7.7] years; 456 female and 350 male). Baseline characteristics were balanced between both groups. The between-group difference in mean change from baseline in CDR-SB score (placebo minus crenezumab) was -0.17 (95% CI, -0.86 to 0.53; P = .63) at week 105 in the CREAD study (88 placebo; 86 crenezumab). Compared with previous trials, no new safety signals were identified, and amyloid-related imaging abnormalities with edema were rare, mild, and transient. No meaningful changes in AD biomarkers were observed. Both studies were discontinued following a preplanned interim analysis indicating that CREAD was unlikely to meet the primary end point.

Conclusions and relevance: Crenezumab was well tolerated but did not reduce clinical decline in participants with early AD.

Trial registration: ClinicalTrials.gov Identifiers: CREAD, NCT02670083; CREAD2, NCT03114657.

Conflict of interest statement

Conflict of Interest Disclosures: Drs Ostrowitzki, Bittner, Schneider, Blondeau, and Hu and Ms Smith are full-time employees of F. Hoffmann-La Roche and stockholders in F. Hoffmann-La Roche. Drs Sink, Mackey, Rabe, Hu, Clayton, and Sanabria-Bohórquez; Mr Rabbia, Dr Toth; and Drs Eichenlaub, Honigberg, and Doody are full-time employees of Genentech, part of F. Hoffmann-La Roche, and stockholders in F. Hoffmann-La Roche. Dr Honig reports consultant fees from Cortexyme, Eisai, and Prevail; study research funding from AbbVie, Alector, Biogen, Eisai, Genentech, F. Hoffmann-LaRoche, and Eli Lilly; grants from Roche during the conduct of the study; and grants from Abbvie, Eisai, Alector, Acumen, Genentech, Union Chimique Belge, Vaccinex, Biogen, Aprinoia, Cyclerion, Johnson & Johnson, Transposon, and Neurokine Therapeutics and personal fees from Eisai, Prevail, Cortexyme, and Biogen outside the submitted work. Dr Woodward has received research funding and speaker/consultancy fees from F. Hoffmann-La Roche, Eli Lilly, Johnson & Johnson/Janssen Cilag, AbbVie, Actinogen, Axovant, Anavex, Cognition Therapies, Pfizer, Novartis, Biogen, Nutricia, Mundipharma, Abbott, Merck, Eisai, and GlaxoSmithKline. Dr Boada reports grants from Merck, AbbVie, Araclon, Biogen, Bioibérica, Grifols, Kyowa Hakko Kirin, Eli Lilly; grants and other support from Merck Sharp & Dohme, Nutricia, Oryzon Genomics, Piramal Imaging Limited, and Schwabe Farma Iberica outside the submitted work; and for Caixabank, but does not receive them directly; fees from Servier and Eli Lilly for consulting; from Eli Lilly, Nutricia, Roche, and Schwabe for lectures; from Eli Lilly, Biogen, Roche, Bioiberica, Eisai, and Schwabe for serving on advisory boards; from Grifols, Roche, Biogen Araclon Biotech for consulting, advisory boards, and lectures; from Merck and Zambon for consulting and advisory boards; from Novo-Nordisk and Servier for advisory boards and lectures; grants from Grifols, La Caixa, Ciberned, European Federation of Pharmaceutical Industries and Associations, the EuroNanoMed III program, the Innovative Medicines Initiative Eranet Call 2012, Programa Misiones IA-2021—Spain, the EU Joint Programme – Neurodegenerative Disease Research PRE-ADAPT project, European Marie Sklodowska Curie, Instituto de Salud Carlos III, and Fondo Europeo Desarrollo Regional during the conduct of the study.Dr van Dyck has received recent speaker/consultancy fees from F. Hoffmann-La Roche, Eisai, Ono, and Cerevel and recent study research funding from Biogen, Biohaven, Eisai, Genentech, F. Hoffmann-La Roche, Johnson & Johnson/Janssen, Eli Lilly, Merck, Novartis, Union Chimique Belge, and Cerevel as well as grants from Biogen, Eli Lilly, Biohaven, Janssen, Union Chimique Belge, Roche, Genentech, Eisai, and Cerevel and personal fees from Roche (scientific advisor), Eisai (scientific advisor), Ono (consultant), and Cerevel (consultant) outside the submitted work. Dr Grimmer has received consulting fees from AbbVie,Alector, Anavex, Biogen, Bracket, Eli Lilly, Functional Neuromodulation, Grifols, Iqvia/Quintiles, Life Molecular Imaging, Novartis, Novo Nordisk, Noselab, NuiCare, Roche Pharma, Toyama, and Vivoryon; lecture fees from Actelion, B.Braun, Biogen, Eli Lilly, Life Molecular Imaging, Novartis, Parexel, Roche Diagnostics, Roche Pharma, Schwabe, Toyama, Union Chimique Belge, and Vivoryon; and grants to his institution from Actelion and Novartis. Dr Selkoe is a director and consultant to Prothena Biosciences. Dr Quartino was a full-time employee of Genentech, part of F. Hoffmann-La Roche, at the time of this work and is now a full-time employee of Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Gothenburg, Sweden. Dr Dolton was a full-time employee of Genentech, part of F. Hoffmann-La Roche, at the time of this work and is now a full-time employee of Roche Products and owns stock or stock options in Roche. No other disclosures were reported.

Figures

Figure 1.. Enrollment, Randomization, and Study Completion…
Figure 1.. Enrollment, Randomization, and Study Completion in CREAD and CREAD2
CREAD indicates A Phase III, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Efficacy and Safety Study of Crenezumab in Patients With Prodromal to Mild Alzheimer’s Disease. aThe number of participant deaths does not represent the total number of deaths in the studies but only those where the death itself led to study discontinuation. A total of 13 deaths occurred (CREAD: 5 in placebo and 8 in crenezumab; CREAD2: 6 in placebo and 0 in crenezumab).
Figure 2.. Mean Change in Clinical Dementia…
Figure 2.. Mean Change in Clinical Dementia Rating–Sum of Boxes Scores for CREAD
CREAD indicates A Phase III, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Efficacy and Safety Study of Crenezumab in Patients With Prodromal to Mild Alzheimer’s Disease.

References

    1. Panza F, Lozupone M, Logroscino G, Imbimbo BP. A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease. Nat Rev Neurol. 2019;15(2):73-88. doi:10.1038/s41582-018-0116-6
    1. Meilandt WJ, Maloney JA, Imperio J, et al. . Characterization of the selective in vitro and in vivo binding properties of crenezumab to oligomeric Aβ. Alzheimers Res Ther. 2019;11(1):97. doi:10.1186/s13195-019-0553-5
    1. Cummings JL, Cohen S, van Dyck CH, et al. . ABBY: a phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology. 2018;90(21):e1889-e1897. doi:10.1212/WNL.0000000000005550
    1. Salloway S, Honigberg LA, Cho W, et al. . Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer’s disease (BLAZE). Alzheimers Res Ther. 2018;10(1):96. doi:10.1186/s13195-018-0424-5
    1. Guthrie H, Honig LS, Lin H, et al. . Safety, tolerability, and pharmacokinetics of crenezumab in patients with mild-to-moderate Alzheimer’s disease treated with escalating doses for up to 133 weeks. J Alzheimers Dis. 2020;76(3):967-979. doi:10.3233/JAD-200134
    1. 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(3):263-269. doi:10.1016/j.jalz.2011.03.005
    1. Albert MS, DeKosky ST, Dickson D, et al. . The diagnosis of mild cognitive impairment 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(3):270-279. doi:10.1016/j.jalz.2011.03.008
    1. O’Bryant SE, Waring SC, Cullum CM, et al. ; Texas Alzheimer’s Research Consortium . Staging dementia using Clinical Dementia Rating Scale Sum of Boxes scores: a Texas Alzheimer’s research consortium study. Arch Neurol. 2008;65(8):1091-1095. doi:10.1001/archneur.65.8.1091
    1. 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(suppl 2):S13-S21. doi:10.1097/00002093-199700112-00003
    1. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry. 1984;141(11):1356-1364. doi:10.1176/ajp.141.11.1356
    1. 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(3):189-198. doi:10.1016/0022-3956(75)90026-6
    1. 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(suppl 2):S33-S39. doi:10.1097/00002093-199700112-00005
    1. Hughes CP, Berg L, Danziger WL, Coben LA, Martin RL. A new clinical scale for the staging of dementia. Br J Psychiatry. 1982;140:566-572. doi:10.1192/bjp.140.6.566
    1. Kaufer DI, Cummings JL, Ketchel P, et al. . Validation of the NPI-Q, a brief clinical form of the Neuropsychiatric Inventory. J Neuropsychiatry Clin Neurosci. 2000;12(2):233-239. doi:10.1176/jnp.12.2.233
    1. Logsdon RG, Gibbons LE, McCurry SM, Teri L. Quality of life in Alzheimer’s disease: patient and caregiver reports. J Ment Health Aging. 1999;5(1):21-32.
    1. Le Scouiller S, Edgar C, Rylands A, et al. . Cross-cultural adaptation of the Zarit Caregiver Interview for Alzheimer’s Disease (ZCI-AD) and the Caregiver Global Impression scales in 13 languages. Value Health. 2016;19(7):PA389. doi:10.1016/j.jval.2016.09.244
    1. Zarit SH, Orr NK, Zarit JM. The Hidden Victims of Alzheimer’s Disease: Families Under Stress. NYU Press; 1985.
    1. Herdman M, Gudex C, Lloyd A, et al. . Development and preliminary testing of the new five-level version of EQ-5D (EQ-5D-5L). Qual Life Res. 2011;20(10):1727-1736. doi:10.1007/s11136-011-9903-x
    1. Blennow K, Dage J, Johnson SC, et al. . Exploring the need for robust biomarker assays in Alzheimer’s disease and other neurodegenerative diseases. Alzheimers Dement. 2019;15(75):2-231. doi:10.1016/j.jalz.2019.06.2638
    1. Yang T, Dang Y, Ostaszewski B, et al. . Target engagement in an Alzheimer trial: crenezumab lowers amyloid β oligomers in cerebrospinal fluid. Ann Neurol. 2019;86(2):215-224. doi:10.1002/ana.25513
    1. Posner K, Brown GK, Stanley B, et al. . The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168(12):1266-1277. doi:10.1176/appi.ajp.2011.10111704
    1. Penninkilampi R, Brothers HM, Eslick GD. Safety and efficacy of anti-amyloid-β immunotherapy in Alzheimer’s disease: a systematic review and meta-analysis. J Neuroimmune Pharmacol. 2017;12(1):194-203. doi:10.1007/s11481-016-9722-5
    1. Swanson C, Zhang Y, Dhadda S, et al. . DT-01-07: treatment of early AD subjects with BAN2401, an anti-Aβ protofibril monoclonal antibody, significantly clears amyloid plaque and reduces clinical decline. Alzheimer’s & Dementia. 2018;14(7S):P1668. doi:10.1016/j.jalz.2018.07.009
    1. Mintun MA, Lo AC, Duggan Evans C, et al. . Donanemab in early Alzheimer’s disease. N Engl J Med. 2021;384(18):1691-1704. doi:10.1056/NEJMoa2100708
    1. Dunn B, Stein P, Cavazzoni P. Approval of aducanumab for Alzheimer disease—the FDA’s perspective. JAMA Intern Med. 2021;181(10):1276-1278. doi:10.1001/jamainternmed.2021.4607
    1. Wessels AM, Lines C, Stern RA, et al. . Cognitive outcomes in trials of two BACE inhibitors in Alzheimer’s disease. Alzheimers Dement. 2020;16(11):1483-1492. doi:10.1002/alz.12164
    1. Ultsch M, Li B, Maurer T, et al. . Structure of crenezumab complex with Aβ shows loss of β-hairpin. Sci Rep. 2016;6:39374. doi:10.1038/srep39374
    1. Yoshida K, Moein A, Bittner T, et al. . Pharmacokinetics and pharmacodynamic effect of crenezumab on plasma and cerebrospinal fluid beta-amyloid in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Res Ther. 2020;12(1):16. doi:10.1186/s13195-020-0580-2
    1. Ferl GZ, Fuji RN, Atwal JK, Sun T, Ramanujan S, Quartino AL. Mechanistic modeling of soluble Aβ dynamics and target engagement in the brain by anti-Aβ mAbs in Alzheimer’s disease. Curr Alzheimer Res. 2020;17(4):393-406. doi:10.2174/1567205017666200302122307

Source: PubMed

3
Abonnieren