Effect of AZD0530 on Cerebral Metabolic Decline in Alzheimer Disease: A Randomized Clinical Trial

Christopher H van Dyck, Haakon B Nygaard, Kewei Chen, Michael C Donohue, Rema Raman, Robert A Rissman, James B Brewer, Robert A Koeppe, Tiffany W Chow, Michael S Rafii, Devon Gessert, Jiyoon Choi, R Scott Turner, Jeffrey A Kaye, Seth A Gale, Eric M Reiman, Paul S Aisen, Stephen M Strittmatter, Christopher H van Dyck, Haakon B Nygaard, Kewei Chen, Michael C Donohue, Rema Raman, Robert A Rissman, James B Brewer, Robert A Koeppe, Tiffany W Chow, Michael S Rafii, Devon Gessert, Jiyoon Choi, R Scott Turner, Jeffrey A Kaye, Seth A Gale, Eric M Reiman, Paul S Aisen, Stephen M Strittmatter

Abstract

Importance: Oligomeric amyloid-β peptide binds to cellular prion protein on the neuronal cell surface, activating intracellular fyn kinase to mediate synaptotoxicity and tauopathy. AZD0530 is an investigational kinase inhibitor specific for the Src family, including fyn, that has been repurposed for the treatment of Alzheimer disease.

Objective: To determine whether AZD0530 treatment slows the decline in cerebral metabolic rate for glucose (CMRgl) and is safe and well tolerated.

Design, setting, and participants: This multicenter phase 2a randomized clinical trial enrolled participants between December 23, 2014, and November 30, 2016. Participants (n = 159) had mild Alzheimer dementia and positron emission tomography (PET) evidence of elevated levels of amyloid-β peptide. Efficacy analyses of all primary and secondary outcomes were conducted in a modified intention-to-treat population. Final analyses were conducted from February 9, 2018, to July 25, 2018.

Interventions: AZD0530 (100 mg or 125 mg daily) vs placebo for 52 weeks.

Main outcomes and measures: Primary outcome was the reduction in relative CMRgl, as measured by 18F-fluorodeoxyglucose (18F-FDG) PET, at 52 weeks in an Alzheimer disease-associated prespecified statistical region of interest. Secondary end points included change in cognition, function, and other biomarkers.

Results: Among the 159 participants, 79 were randomized to receive AZD0530 and 80 to receive placebo. Of the 159 participants, 87 (54.7%) were male, with a mean (SD) age of 71.0 (7.7) years. Based on a week-2 plasma drug level (target = 180 ng/mL; 30nM free), 15 participants (19.2%) had their AZD0530 dose escalated from 100 mg to 125 mg. Mean plasma levels from weeks 13 to 52 were 220 ng/mL and 36nM free. More participants discontinued treatment with AZD0530 than with placebo (21 vs 11), most commonly because of adverse events. The most frequent adverse events were gastrointestinal disorders (primarily diarrhea), which occurred in 38 participants (48.1%) who received AZD0530 and in 23 (28.8%) who received placebo. In the primary outcome, the treatment groups did not differ in 52-week decline in relative CMRgl (mean difference: -0.006 units/y; 95% CI, -0.017 to 0.006; P = .34). The treatment groups also did not differ in the rate of change in Alzheimer's Disease Assessment Scale-Cognitive Subscale, Alzheimer's Disease Cooperative Study-Activities of Daily Living, Clinical Dementia Rating, Neuropsychiatric Inventory, or Mini-Mental State Examination scores. Secondary volumetric magnetic resonance imaging analyses revealed no treatment effect on total brain or ventricular volume but did show trends for slowing the reduction in hippocampal volume and entorhinal thickness.

Conclusions and relevance: Statistically significant effects of AZD0530 treatment were not found on relative CMRgl reduction in an Alzheimer disease-associated region of interest or on secondary clinical or biomarker measures.

Trial registration: ClinicalTrials.gov identifier: NCT02167256.

Conflict of interest statement

Conflict of Interest Disclosures: Dr van Dyck reported receiving consulting fees from Roche, Eisai, Kyowa Kirin, Merck, Eli Lilly, and Janssen as well as grants for clinical trials from Biogen, Novartis, Eli Lilly, Merck, Eisai, Janssen, Roche, Genentech, Biohaven, Toyama, and TauRx outside of the submitted work. Dr Nygaard reported receiving 18F-Florbetapir from Avid Radiopharmaceuticals for the Canadian site (University of British Columbia). Dr Donohue reported personal fees from Eli Lilly and Neurotrack and other from Janssen outside the submitted work. Dr Chen reported current full-time employment at Green Valley Pharmaceuticals, Shanghai, China, and past employment at Banner Alzheimer’s Institute during the conduct of the study. Dr Raman reported receiving grants from Eli Lilly, Janssen, and the National Institute on Aging through the Alzheimer’s Therapeutic Research Institute. Dr Brewer reported receiving consulting fees from Elan, Bristol-Myers Squibb, Avanir, Novartis, Genentech, and Eli Lilly as well as stock options in CorTechs Labs Inc and Human Longevity Inc. Dr Koeppe reported receiving grants from the National Institutes of Health during the conduct of the study. Dr Kaye reported receiving grants from Alzheimer's Therapeutic Research Institute during the conduct of the study. Dr Turner reported receiving consulting fees from Eli Lilly and grants for clinical trials from Acadia, Biogen, Novartis, Eli Lilly, Merck, Eisai, Janssen, Roche, Genentech, and Toyama. Dr Gale reported receiving consulting fees from Phillips and grants for clinical trials from Eisai, Roche, Eli Lilly, Novartis, and Genentech. Dr Reiman reported receiving fees as a scientific advisor to Alkahest, Alzheon, Aural Analytics, Denali, Green Valley, Roche (expenses only), United Neuroscience, and Zinfandel and grants for clinical trials from Genentech/Roche, Novartis/Amgen, and Avid/Lilly. Dr Aisen reported receiving consulting fees from Proclara, Merck, Biogen, Roche, Eisai, Storz, and ImmunobrainCheckpoint as well as grants for clinical trials from Eli Lilly and Janssen. Dr Strittmatter reported receiving founder shares and consulting fees from ReNetX Bio as well as being an inventor on Yale patents and patent applications related to Alzheimer treatment. No other disclosures were reported.

Figures

Figure 1.. CONSORT Diagram
Figure 1.. CONSORT Diagram
Figure 2.. Primary Outcome
Figure 2.. Primary Outcome
A, A 52-week cerebral metabolic rate for glucose (CMRgl) decline statistical region of interest (sROI; in the red-to-yellow color scale) and a spared sROI (in the blue-to-green color scale) were generated using baseline and follow-up. Shown are 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) images acquired in an Alzheimer’s Disease Neuroimaging Initiative study and updated for amyloid-positive participants with Alzheimer disease, as previously described., B. The y-axis represents relative CMRgl-derived 18F-FDG PET in the Alzheimer disease–associated sROI normalized to the spared sROI. The treatment groups did not differ in the 52-week decline in CMRgl (difference, −0.006 units/y; 95% CI, −0.017 to 0.006; P = .34). The mean (SD) 12-month decrease in 18F-FDG PET CMRgl was .0525 (.0340) in the placebo (control) group (n = 72), which was close to the pilot estimates used in the power analysis, and 0.0569 (0.0303) in the AZD0530 group (n = 59). Blue represents the placebo group; orange, the AZD0530 group.
Figure 3.. Secondary Outcomes
Figure 3.. Secondary Outcomes
A, Analyses of clinical variables used a mixed model of repeated measures to estimate the mean group difference at each follow-up time, with change from baseline as the outcome, controlling for baseline score, age, and apolipoprotein E (APOE) ε4 status. B. Analyses of magnetic resonance imaging (MRI) variables used an analysis of covariance model with percentage of deformation per year from baseline as the outcome, adjusted for mean baseline volume or thickness of brain region, age, and APOE ε4 status. Blue represents the placebo group; orange, the AZD0530 group. aAlzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-Cog11) score range: 0 (indicating best) to 70 (worst). bAlzheimer’s Disease Cooperative Study–Activities of Daily Living (ADCS-ADL) score range: 0 (worst) to 78 (best). cClinical Dementia Rating–Sum of Boxes (CDR-SB) score range: 0 (best) to 18 (worst). dNeuropsychiatric Inventory score range: 0 (best) to 144 (worst).

References

    1. Voytyuk I, De Strooper B, Chávez-Gutiérrez L. Modulation of γ- and β-secretases as early prevention against Alzheimer’s disease. Biol Psychiatry. 2018;83(4):320-327. doi:10.1016/j.biopsych.2017.08.001
    1. van Dyck CH. Anti-amyloid-β monoclonal antibodies for Alzheimer’s disease: pitfalls and promise. Biol Psychiatry. 2018;83(4):311-319. doi:10.1016/j.biopsych.2017.08.010
    1. Berman DE, Dall’Armi C, Voronov SV, et al. . Oligomeric amyloid-beta peptide disrupts phosphatidylinositol-4,5-bisphosphate metabolism. Nat Neurosci. 2008;11(5):547-554. doi:10.1038/nn.2100
    1. Cleary JP, Walsh DM, Hofmeister JJ, et al. . Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function. Nat Neurosci. 2005;8(1):79-84. doi:10.1038/nn1372
    1. Hong S, Beja-Glasser VF, Nfonoyim BM, et al. . Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352(6286):712-716. doi:10.1126/science.aad8373
    1. Lacor PN, Buniel MC, Furlow PW, et al. . Aβ oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci. 2007;27(4):796-807. doi:10.1523/JNEUROSCI.3501-06.2007
    1. Lambert MP, Barlow AK, Chromy BA, et al. . Diffusible, nonfibrillar ligands derived from Aβ1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95(11):6448-6453. doi:10.1073/pnas.95.11.6448
    1. Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457(7233):1128-1132. doi:10.1038/nature07761
    1. Lesné S, Koh MT, Kotilinek L, et al. . A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440(7082):352-357. doi:10.1038/nature04533
    1. Li S, Hong S, Shepardson NE, Walsh DM, Shankar GM, Selkoe D. Soluble oligomers of amyloid beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron. 2009;62(6):788-801. doi:10.1016/j.neuron.2009.05.012
    1. Palop JJ, Mucke L. Amyloid-beta-induced neuronal dysfunction in Alzheimer’s disease: from synapses toward neural networks. Nat Neurosci. 2010;13(7):812-818. doi:10.1038/nn.2583
    1. Shankar GM, Li S, Mehta TH, et al. . Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837-842. doi:10.1038/nm1782
    1. Walsh DM, Klyubin I, Fadeeva JV, et al. . Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416(6880):535-539. doi:10.1038/416535a
    1. Purro SA, Nicoll AJ, Collinge J. Prion protein as a toxic acceptor of amyloid-β oligomers. Biol Psychiatry. 2018;83(4):358-368. doi:10.1016/j.biopsych.2017.11.020
    1. Haas LT, Salazar SV, Kostylev MA, Um JW, Kaufman AC, Strittmatter SM. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer’s disease. Brain. 2016;139(pt 2):526-546. doi:10.1093/brain/awv356
    1. Um JW, Kaufman AC, Kostylev M, et al. . Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer aβ oligomer bound to cellular prion protein. Neuron. 2013;79(5):887-902. doi:10.1016/j.neuron.2013.06.036
    1. Larson M, Sherman MA, Amar F, et al. . The complex PrP(c)-fyn couples human oligomeric Aβ with pathological tau changes in Alzheimer’s disease. J Neurosci. 2012;32(47):16857-16871. doi:10.1523/JNEUROSCI.1858-12.2012
    1. Um JW, Nygaard HB, Heiss JK, et al. . Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15(9):1227-1235. doi:10.1038/nn.3178
    1. Beecham GW, Hamilton K, Naj AC, et al. ; Alzheimer’s Disease Genetics Consortium (ADGC) . Genome-wide association meta-analysis of neuropathologic features of Alzheimer’s disease and related dementias. PLoS Genet. 2014;10(9):e1004606. doi:10.1371/journal.pgen.1004606
    1. Bhaskar K, Hobbs GA, Yen SH, Lee G. Tyrosine phosphorylation of tau accompanies disease progression in transgenic mouse models of tauopathy. Neuropathol Appl Neurobiol. 2010;36(6):462-477. doi:10.1111/j.1365-2990.2010.01103.x
    1. Bhaskar K, Yen SH, Lee G. Disease-related modifications in tau affect the interaction between fyn and tau. J Biol Chem. 2005;280(42):35119-35125. doi:10.1074/jbc.M505895200
    1. Chin J, Palop JJ, Puoliväli J, et al. . Fyn kinase induces synaptic and cognitive impairments in a transgenic mouse model of Alzheimer’s disease. J Neurosci. 2005;25(42):9694-9703. doi:10.1523/JNEUROSCI.2980-05.2005
    1. Chin J, Palop JJ, Yu GQ, Kojima N, Masliah E, Mucke L. Fyn kinase modulates synaptotoxicity, but not aberrant sprouting, in human amyloid precursor protein transgenic mice. J Neurosci. 2004;24(19):4692-4697. doi:10.1523/JNEUROSCI.0277-04.2004
    1. Dourlen P, Fernandez-Gomez FJ, Dupont C, et al. . Functional screening of Alzheimer risk loci identifies PTK2B as an in vivo modulator and early marker of tau pathology. Mol Psychiatry. 2017;22(6):874-883. doi:10.1038/mp.2016.59
    1. Haas LT, Strittmatter SM. Oligomers of amyloid β prevent physiological activation of the cellular prion protein-metabotropic glutamate receptor 5 complex by glutamate in Alzheimer disease. J Biol Chem. 2016;291(33):17112-17121. doi:10.1074/jbc.M116.720664
    1. Ittner LM, Ke YD, Delerue F, et al. . Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell. 2010;142(3):387-397. doi:10.1016/j.cell.2010.06.036
    1. Kaufman AC, Salazar SV, Haas LT, et al. . Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol. 2015;77(6):953-971. doi:10.1002/ana.24394
    1. Lambert JC, Ibrahim-Verbaas CA, Harold D, et al. ; European Alzheimer’s Disease Initiative (EADI); Genetic and Environmental Risk in Alzheimer’s Disease; Alzheimer’s Disease Genetic Consortium; Cohorts for Heart and Aging Research in Genomic Epidemiology . Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45(12):1452-1458. doi:10.1038/ng.2802
    1. Lee G, Newman ST, Gard DL, Band H, Panchamoorthy G. Tau interacts with src-family non-receptor tyrosine kinases. J Cell Sci. 1998;111(pt 21):3167-3177.
    1. Lee G, Thangavel R, Sharma VM, et al. . Phosphorylation of tau by fyn: implications for Alzheimer’s disease. J Neurosci. 2004;24(9):2304-2312. doi:10.1523/JNEUROSCI.4162-03.2004
    1. Roberson ED, Halabisky B, Yoo JW, et al. . Amyloid-β/fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J Neurosci. 2011;31(2):700-711. doi:10.1523/JNEUROSCI.4152-10.2011
    1. Nygaard HB. Targeting fyn kinase in Alzheimer’s disease. Biol Psychiatry. 2018;83(4):369-376. doi:10.1016/j.biopsych.2017.06.004
    1. Nygaard HB, van Dyck CH, Strittmatter SM. Fyn kinase inhibition as a novel therapy for Alzheimer’s disease. Alzheimers Res Ther. 2014;6(1):8. doi:10.1186/alzrt238
    1. Reiman EM, Langbaum JBS. Brain imaging in the evaluation of putative Alzheimer’s disease slowing, risk-reducing and prevention therapies In: Jagust WJ, D’Esposito M, eds. Imaging the Aging Brain. New York: Oxford University Press; 2009:319-350. doi:10.1093/acprof:oso/9780195328875.003.0020
    1. Jones RW, Schwam E, Wilkinson D, et al. . Rates of cognitive change in Alzheimer disease: observations across a decade of placebo-controlled clinical trials with donepezil. Alzheimer Dis Assoc Disord. 2009;23(4):357-364. doi:10.1097/WAD.0b013e31819cd4be
    1. Landau SM, Harvey D, Madison CM, et al. ; Alzheimer’s Disease Neuroimaging Initiative . Associations between cognitive, functional, and FDG-PET measures of decline in AD and MCI. Neurobiol Aging. 2011;32(7):1207-1218. doi:10.1016/j.neurobiolaging.2009.07.002
    1. Alexander GE, Chen K, Pietrini P, Rapoport SI, Reiman EM. Longitudinal PET evaluation of cerebral metabolic decline in dementia: a potential outcome measure in Alzheimer’s disease treatment studies. Am J Psychiatry. 2002;159(5):738-745. doi:10.1176/appi.ajp.159.5.738
    1. Reiman EM, Caselli RJ, Chen K, Alexander GE, Bandy D, Frost J. Declining brain activity in cognitively normal apolipoprotein E epsilon 4 heterozygotes: a foundation for using positron emission tomography to efficiently test treatments to prevent Alzheimer’s disease. Proc Natl Acad Sci U S A. 2001;98(6):3334-3339. doi:10.1073/pnas.061509598
    1. Chen K, Langbaum JB, Fleisher AS, et al. ; Alzheimer’s Disease Neuroimaging Initiative . Twelve-month metabolic declines in probable Alzheimer’s disease and amnestic mild cognitive impairment assessed using an empirically pre-defined statistical region-of-interest: findings from the Alzheimer’s Disease Neuroimaging Initiative. Neuroimage. 2010;51(2):654-664. doi:10.1016/j.neuroimage.2010.02.064
    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. Rosen WG, Terry RD, Fuld PA, Katzman R, Peck A. Pathological verification of ischemic score in differentiation of dementias. Ann Neurol. 1980;7(5):486-488. doi:10.1002/ana.410070516
    1. Sheikh J, Yesavage J. Geriatric Depression Scale (GDS): recent evidence and development of a shorter version In: Brind TL, ed. Clinical Gerontology: A Guide to Assessment and Intervention. New York: Haworth Press; 1986:165-173.
    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. Baselga J, Cervantes A, Martinelli E, et al. . Phase I safety, pharmacokinetics, and inhibition of SRC activity study of saracatinib in patients with solid tumors. Clin Cancer Res. 2010;16(19):4876-4883. doi:10.1158/1078-0432.CCR-10-0748
    1. Chen K, Lee W, Kuang X, et al. Twelve-month glucose metabolism declines in an empirically pre-defined statistical region-of-interest in amyloid-positive persons with Alzheimer's dementia and mild cognitive impairment: updated ADNI findings. Paper presented at: Human Amyloid Imaging; January 19, 2018; Miami, FL.
    1. Mohs RC, Knopman D, Petersen RC, et al. ; the Alzheimer’s Disease Cooperative Study . Development of cognitive instruments for use in clinical trials of antidementia drugs: additions to the Alzheimer’s Disease Assessment Scale that broaden its scope. 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. Galasko D, Bennett D, Sano M, et al. ; the Alzheimer’s Disease Cooperative Study . An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. Alzheimer Dis Assoc Disord. 1997;11(suppl 2):S33-S39. doi:10.1097/00002093-199700112-00005
    1. Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology. 1993;43(11):2412-2414. doi:10.1212/WNL.43.11.2412-a
    1. Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994;44(12):2308-2314. doi:10.1212/WNL.44.12.2308
    1. Cummings JL. The Neuropsychiatric Inventory: assessing psychopathology in dementia patients. Neurology. 1997;48(5)(suppl 6):S10-S16. doi:10.1212/WNL.48.5_Suppl_6.10S
    1. Holland D, Dale AM; Alzheimer’s Disease Neuroimaging Initiative . Nonlinear registration of longitudinal images and measurement of change in regions of interest. Med Image Anal. 2011;15(4):489-497. doi:10.1016/j.media.2011.02.005
    1. Desikan RS, Ségonne F, Fischl B, et al. . An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. Neuroimage. 2006;31(3):968-980. doi:10.1016/j.neuroimage.2006.01.021
    1. Nygaard HB, Wagner AF, Bowen GS, et al. . A phase Ib multiple ascending dose study of the safety, tolerability, and central nervous system availability of AZD0530 (saracatinib) in Alzheimer’s disease. Alzheimers Res Ther. 2015;7(1):35. doi:10.1186/s13195-015-0119-0

Source: PubMed

3
Subscribe