FUNDAMANT: an interventional 72-week phase 1 follow-up study of AADvac1, an active immunotherapy against tau protein pathology in Alzheimer's disease

Petr Novak, Reinhold Schmidt, Eva Kontsekova, Branislav Kovacech, Tomas Smolek, Stanislav Katina, Lubica Fialova, Michal Prcina, Vojtech Parrak, Peter Dal-Bianco, Martin Brunner, Wolfgang Staffen, Michael Rainer, Matej Ondrus, Stefan Ropele, Miroslav Smisek, Roman Sivak, Norbert Zilka, Bengt Winblad, Michal Novak, Petr Novak, Reinhold Schmidt, Eva Kontsekova, Branislav Kovacech, Tomas Smolek, Stanislav Katina, Lubica Fialova, Michal Prcina, Vojtech Parrak, Peter Dal-Bianco, Martin Brunner, Wolfgang Staffen, Michael Rainer, Matej Ondrus, Stefan Ropele, Miroslav Smisek, Roman Sivak, Norbert Zilka, Bengt Winblad, Michal Novak

Abstract

Background: Neurofibrillary pathology composed of tau protein is closely correlated with severity and phenotype of cognitive impairment in patients with Alzheimer's disease and non-Alzheimer's tauopathies. Targeting pathological tau proteins via immunotherapy is a promising strategy for disease-modifying treatment of Alzheimer's disease. Previously, we reported a 24-week phase 1 trial on the active vaccine AADvac1 against pathological tau protein; here, we present the results of a further 72 weeks of follow-up on those patients.

Methods: We did a phase 1, 72-week, open-label study of AADvac1 in patients with mild to moderate Alzheimer's disease who had completed the preceding phase 1 study. Patients who were previously treated with six doses of AADvac1 at monthly intervals received two booster doses at 24-week intervals. Patients who were previously treated with only three doses received another three doses at monthly intervals, and subsequently two boosters at 24-week intervals. The primary objective was the assessment of long-term safety of AADvac1 treatment. Secondary objectives included assessment of antibody titres, antibody isotype profile, capacity of the antibodies to bind to AD tau and AADvac1, development of titres of AADvac1-induced antibodies over time, and effect of booster doses; cognitive assessment via 11-item Alzheimer's Disease Assessment Scale cognitive assessment (ADAS-Cog), Category Fluency Test and Controlled Oral Word Association Test; assessment of brain atrophy via magnetic resonance imaging (MRI) volumetry; and assessment of lymphocyte populations via flow cytometry.

Results: The study was conducted between 18 March 2014 and 10 August 2016. Twenty-six patients who completed the previous study were enrolled. Five patients withdrew because of adverse events. One patient was withdrawn owing to noncompliance. The most common adverse events were injection site reactions (reported in 13 [50%] of vaccinated patients). No cases of meningoencephalitis or vasogenic oedema were observed. New micro-haemorrhages were observed only in one ApoE4 homozygote. All responders retained an immunoglobulin G (IgG) antibody response against the tau peptide component of AADvac1 over 6 months without administration, with titres regressing to a median 15.8% of titres attained after the initial six-dose vaccination regimen. Booster doses restored previous IgG levels. Hippocampal atrophy rate was lower in patients with high IgG levels; a similar relationship was observed in cognitive assessment.

Conclusions: AADvac1 displayed a benign safety profile. The evolution of IgG titres over vaccination-free periods warrants a more frequent booster dose regimen. The tendency towards slower atrophy in MRI evaluation and less of a decline in cognitive assessment in patients with high titres is encouraging. Further trials are required to expand the safety database and to establish proof of clinical efficacy of AADvac1.

Trial registration: The studies are registered with the EU Clinical Trials Register and ClinicalTrials.gov : the preceding first-in-human study under EudraCT 2012-003916-29 and NCT01850238 (registered on 9 May 2013) and the follow-up study under EudraCT 2013-004499-36 and NCT02031198 (registered 9 Jan 2014), respectively.

Keywords: Active immunotherapy; Alzheimer’s disease; Clinical trial; Immunotherapy; Neurofibrillary pathology; Tau; Vaccine.

Conflict of interest statement

Competing interests

Authors affiliated with AXON Neuroscience SE, AXON Neuroscience CRM Services SE and AXON Neuroscience R&D Services SE receive salary from the company. The investigators’ institutions have received payments on a per-patient per-visit basis. BW has received personal fees from AXON Neuroscience SE for advisory services.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Antibody response to AADvac1 over 96 weeks of treatment. Values shown indicate geometric mean titres and 95% CI of the geometric mean. Time points of AADvac1 administration indicated by bold treatment weeks. Light grey area indicates values obtained in the first-in-human study
Fig. 2
Fig. 2
The antibody response to AADvac1 is immunoglobulin G1-dominated. Values were obtained after the sixth dose of AADvac1
Fig. 3
Fig. 3
AADvac1-induced antibodies label tau extracts from corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) brains. Left: Patient R17, pre-treatment serum. Right: Serum of the same patient after six doses of AADvac1. The serum labels both high-molecular-weight aggregates and low-molecular-weight fragments of tau protein. Staining of Alzheimer’s disease (AD) brain extract is shown as a positive control
Fig. 4
Fig. 4
Sera of patients treated with AADvac1 recognise tau pathology in Alzheimer’s disease and Pick’s disease. af Alzheimer’s disease. gl: Pick’s disease. Sera of three different patients with different strengths of antibody responses (patient R17 with an anti-AD-tau titre of 1:30,999; patient R25 with 1:18,185; and patient R10 with 1:12,800) were used for staining. Staining with pre-treatment sera is shown as a negative control (df, jl)
Fig. 5
Fig. 5
Sera of patients treated with AADvac1 recognise tau pathology in progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). ad PSP. gl CBD. Sera of three different patients with different strengths of antibody response (patient R17 with an anti-AD-tau titre of 1:30,999; patient R25 with 1:18,185; and patient R10 with 1:12,800) were used for staining. Staining with pre-treatment sera is shown as a negative control (df, jl)
Fig. 6
Fig. 6
Change in cognition (Alzheimer‘s Disease Assessment Scale 11-item cognitive assessment [ADAS-Cog11], Controlled Oral Word Association Test [COWAT]) over 96 weeks, displayed as correlation with the immunoglobulin G (IgG) titre AUC. Results are shown for completers (left) and for patients with a positive biomarker profile (right). Results are shown with raw AUC values (ad) and with AUC values corrected for disease severity (eh). Category Fluency Test results were inconclusive (not shown)
Fig. 7
Fig. 7
Hippocampal atrophy over 96 weeks, displayed as correlation with the immunoglobulin G (IgG) titre AUC. Results are shown for completers (a, b) (n = 18) and for patients with a positive biomarker profile (c, d) (n = 9). Results are also shown with raw AUC values (a, c) and with AUC values corrected for disease severity (b, d). One patient with frontotemporal dementia and one patient with poor magnetic resonance imaging scan quality were excluded. HCV Hippocampal volume

References

    1. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011;7(3):137–152. doi: 10.1038/nrneurol.2011.2.
    1. Nelson PT, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362–381. doi: 10.1097/NEN.0b013e31825018f7.
    1. Schneider LS, et al. Clinical trials and late-stage drug development for Alzheimer’s disease: an appraisal from 1984 to 2014. J Intern Med. 2014;275(3):251–283. doi: 10.1111/joim.12191.
    1. Wimo A, et al. The economic impact of dementia in Europe in 2008—cost estimates from the Eurocode project. Int J Geriatr Psychiatry. 2011;26(8):825–832. doi: 10.1002/gps.2610.
    1. Bachurin SO, Bovina EV, Ustyugov AA. Drugs in clinical trials for Alzheimer’s disease: the major trends. Med Res Rev. 2017;37(5):1186–1225. doi: 10.1002/med.21434.
    1. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–259. doi: 10.1007/BF00308809.
    1. Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430(7000):631–639. doi: 10.1038/nature02621.
    1. Whitwell JL, et al. Neuroimaging correlates of pathologically defined subtypes of Alzheimer’s disease: a case-control study. Lancet Neurol. 2012;11(10):868–877. doi: 10.1016/S1474-4422(12)70200-4.
    1. Spillantini MG, Goedert M. Tau pathology and neurodegeneration. Lancet Neurol. 2013;12(6):609–622. doi: 10.1016/S1474-4422(13)70090-5.
    1. Iqbal K, Liu F, Gong CX. Tau and neurodegenerative disease: the story so far. Nat Rev Neurol. 2016;12(1):15–27. doi: 10.1038/nrneurol.2015.225.
    1. Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17(1):5–21. doi: 10.1038/nrn.2015.1.
    1. Zilka N, Korenova M, Novak M. Misfolded tau protein and disease modifying pathways in transgenic rodent models of human tauopathies. Acta Neuropathol. 2009;118(1):71–86. doi: 10.1007/s00401-009-0499-y.
    1. Cummings J, et al. Alzheimer’s disease drug development pipeline: 2017. Alzheimers Dement (N Y) 2017;3(3):367–384.
    1. Agadjanyan MG, Petrovsky N, Ghochikyan A. A fresh perspective from immunologists and vaccine researchers: active vaccination strategies to prevent and reverse Alzheimer’s disease. Alzheimers Dement. 2015;11(10):1246–1259. doi: 10.1016/j.jalz.2015.06.1884.
    1. Pineda C, et al. Assessing the immunogenicity of biopharmaceuticals. BioDrugs. 2016;30(3):195–206. doi: 10.1007/s40259-016-0174-5.
    1. Gilman S, et al. Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64(9):1553–1562. doi: 10.1212/01.WNL.0000159740.16984.3C.
    1. Novak P, 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(2):123–134. doi: 10.1016/S1474-4422(16)30331-3.
    1. Vandenberghe R, et al. Active Aβ immunotherapy CAD106 in Alzheimer’s disease: a phase 2b study. Alzheimers Dement (N Y) 2017;3(1):10–22.
    1. Kontsekova E, et al. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer’s disease model. Alzheimers Res Ther. 2014;6(4):44. doi: 10.1186/alzrt278.
    1. Kontsekova E, et al. Identification of structural determinants on tau protein essential for its pathological function: novel therapeutic target for tau immunotherapy in Alzheimer’s disease. Alzheimers Res Ther. 2014;6(4):45. doi: 10.1186/alzrt277.
    1. Sperling RA, et al. The A4 study: stopping AD before symptoms begin? Sci Transl Med. 2014;6(228):228fs13. doi: 10.1126/scitranslmed.3007941.
    1. Tariot PN, et al. The Alzheimer’s Prevention Initiative Autosomal-Dominant Alzheimer’s Disease Trial: a study of crenezumab versus placebo in preclinical PSEN1 E280A mutation carriers to evaluate efficacy and safety in the treatment of autosomal-dominant Alzheimer’s disease, including a placebo-treated noncarrier cohort. Alzheimers Dement (N Y) 2018;4:150–160.
    1. Yanamandra K, et al. Anti-tau antibody reduces insoluble tau and decreases brain atrophy. Ann Clin Transl Neurol. 2015;2(3):278–288. doi: 10.1002/acn3.176.
    1. Bright J, et al. Human secreted tau increases amyloid-β production. Neurobiol Aging. 2015;36(2):693–709. doi: 10.1016/j.neurobiolaging.2014.09.007.
    1. Theunis C, et al. Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy. PLoS One. 2013;8(8):e72301. doi: 10.1371/journal.pone.0072301.
    1. Collin L, et al. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer’s disease. Brain. 2014;137(Pt 10):2834–2846. doi: 10.1093/brain/awu213.
    1. Czerkowicz J, et al. Pan-tau antibody Biib076 exhibits promising safety and biomarker profile in cynomolgus monkey toxicity study [abstract P4-039] Alzheimers Dement. 2018;13(7 Suppl):P1271.
    1. Zilka N, et al. Truncated tau from sporadic Alzheimer’s disease suffices to drive neurofibrillary degeneration in vivo. FEBS Lett. 2006;580(15):3582–3588. doi: 10.1016/j.febslet.2006.05.029.
    1. Smith SM, et al. Accurate, robust, and automated longitudinal and cross-sectional brain change analysis. Neuroimage. 2002;17(1):479–489. doi: 10.1006/nimg.2002.1040.
    1. Smith SM, et al. Advances in functional and structural MR image analysis and implementation as FSL. Neuroimage. 2004;23(Suppl 1):S208–S219. doi: 10.1016/j.neuroimage.2004.07.051.
    1. Reuter M, et al. Within-subject template estimation for unbiased longitudinal image analysis. Neuroimage. 2012;61(4):1402–1418. doi: 10.1016/j.neuroimage.2012.02.084.
    1. Reuter M, Rosas HD, Fischl B. Highly accurate inverse consistent registration: a robust approach. Neuroimage. 2010;53(4):1181–1196. doi: 10.1016/j.neuroimage.2010.07.020.
    1. Zimova I, et al. Human truncated tau induces mature neurofibrillary pathology in a mouse model of human tauopathy. J Alzheimers Dis. 2016;54(2):831–843. doi: 10.3233/JAD-160347.
    1. Arrighi HM, et al. Amyloid-related imaging abnormalities-haemosiderin (ARIA-H) in patients with Alzheimer’s disease treated with bapineuzumab: a historical, prospective secondary analysis. J Neurol Neurosurg Psychiatry. 2016;87(1):106–112.
    1. McKhann GM, 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. Barnes J, et al. A comparison of methods for the automated calculation of volumes and atrophy rates in the hippocampus. Neuroimage. 2008;40(4):1655–1671. doi: 10.1016/j.neuroimage.2008.01.012.
    1. Cash DM, et al. Assessing atrophy measurement techniques in dementia: results from the MIRIAD atrophy challenge. Neuroimage. 2015;123:149–164. doi: 10.1016/j.neuroimage.2015.07.087.
    1. Grundke-Iqbal I, et al. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A. 1986;83(13):4913–4917. doi: 10.1073/pnas.83.13.4913.
    1. Novak M, et al. Difference between the tau protein of Alzheimer paired helical filament core and normal tau revealed by epitope analysis of monoclonal antibodies 423 and 7.51. Proc Natl Acad Sci U S A. 1991;88(13):5837–5841. doi: 10.1073/pnas.88.13.5837.
    1. Binder LI, et al. Tau, tangles, and Alzheimer’s disease. Biochim Biophys Acta. 2005;1739(2–3):216–223. doi: 10.1016/j.bbadis.2004.08.014.
    1. Horowitz PM, et al. Early N-terminal changes and caspase-6 cleavage of tau in Alzheimer’s disease. J Neurosci. 2004;24(36):7895–7902. doi: 10.1523/JNEUROSCI.1988-04.2004.
    1. Fitzpatrick AWP, et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature. 2017;547(7662):185–190. doi: 10.1038/nature23002.
    1. Zhou Y, et al. Relevance of phosphorylation and truncation of tau to the etiopathogenesis of Alzheimer’s disease. Front Aging Neurosci. 2018;10:27. doi: 10.3389/fnagi.2018.00027.
    1. Marciniak E, et al. Tau deletion promotes brain insulin resistance. J Exp Med. 2017;214(8):2257–2269. doi: 10.1084/jem.20161731.
    1. Evans LD, et al. Extracellular monomeric and aggregated tau efficiently enter human neurons through overlapping but distinct pathways. Cell Rep. 2018;22(13):3612–3624. doi: 10.1016/j.celrep.2018.03.021.
    1. Novak M, Kabat J, Wischik CM. Molecular characterization of the minimal protease resistant tau unit of the Alzheimer’s disease paired helical filament. EMBO J. 1993;12(1):365–370. doi: 10.1002/j.1460-2075.1993.tb05665.x.
    1. Taniguchi-Watanabe S, et al. Biochemical classification of tauopathies by immunoblot, protein sequence and mass spectrometric analyses of sarkosyl-insoluble and trypsin-resistant tau. Acta Neuropathol. 2016;131(2):267–280. doi: 10.1007/s00401-015-1503-3.
    1. Lazuardi L, et al. Age-related loss of naive T cells and dysregulation of T-cell/B-cell interactions in human lymph nodes. Immunology. 2005;114(1):37–43. doi: 10.1111/j.1365-2567.2004.02006.x.
    1. Vellas B, et al. Long-term follow-up of patients immunized with AN1792: reduced functional decline in antibody responders. Curr Alzheimer Res. 2009;6(2):144–151. doi: 10.2174/156720509787602852.
    1. Henley DB, et al. Safety profile of Alzheimer’s disease populations in Alzheimer’s Disease Neuroimaging Initiative and other 18-month studies. Alzheimers Dement. 2012;8(5):407–416. doi: 10.1016/j.jalz.2011.05.2413.
    1. Salloway S, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370(4):322–333. doi: 10.1056/NEJMoa1304839.

Source: PubMed

3
订阅