UB-311, a novel UBITh® amyloid β peptide vaccine for mild Alzheimer's disease

Chang Yi Wang, Pei-Ning Wang, Ming-Jang Chiu, Connie L Finstad, Feng Lin, Shugene Lynn, Yuan-Hung Tai, Xin De Fang, Kesheng Zhao, Chung-Ho Hung, Yiting Tseng, Wen-Jiun Peng, Jason Wang, Chih-Chieh Yu, Be-Sheng Kuo, Paul A Frohna, Chang Yi Wang, Pei-Ning Wang, Ming-Jang Chiu, Connie L Finstad, Feng Lin, Shugene Lynn, Yuan-Hung Tai, Xin De Fang, Kesheng Zhao, Chung-Ho Hung, Yiting Tseng, Wen-Jiun Peng, Jason Wang, Chih-Chieh Yu, Be-Sheng Kuo, Paul A Frohna

Abstract

Introduction: A novel amyloid β (Aβ) synthetic peptide vaccine (UB-311) has been evaluated in a first-in-human trial with patients of mild-to-moderate Alzheimer's disease. We describe translational research covering vaccine design, preclinical characterization, and phase-I clinical trial with supportive outcome that advances UB-311 into an ongoing phase-II trial.

Methods: UB-311 is constructed with two synthetic Aβ1-14-targeting peptides (B-cell epitope), each linked to different helper T-cell peptide epitopes (UBITh®) and formulated in a Th2-biased delivery system. The hAPP751 transgenic mouse model was used to perform the proof-of-concept study. Baboons and macaques were used for preclinical safety, tolerability, and immunogenicity evaluation. Patients with mild-to-moderate Alzheimer's disease (AD) were immunized by intramuscular route with 3 doses of UB-311 at weeks 0, 4, and 12, and monitored until week 48. Safety and immunogenicity were assessed per protocol, and preliminary efficacy was analyzed by Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog), Mini-Mental State Examination (MMSE), and Alzheimer's Disease Cooperative Study-Clinician's Global Impression of Change (ADCS-CGIC).

Results: UB-311 covers a diverse genetic background and facilitates strong immune response with high responder rate. UB-311 reduced the levels of Aβ1-42 oligomers, protofibrils, and plaque load in hAPP751 transgenic mice. Safe and well-tolerated UB-311 generated considerable site-specific (Aβ1-10) antibodies across all animal species examined. In AD patients, UB-311 induced a 100% responder rate; injection site swelling and agitation were the most common adverse events (4/19 each). A slower rate of increase in ADAS-Cog from baseline to week 48 was observed in the subgroup of mild AD patients (MMSE ≥ 20) compared with the moderate AD subgroup, suggesting that UB-311 may have a potential of cognition improvement in patients with early stage of Alzheimer's dementia.

Discussion: The UBITh® platform can generate a high-precision molecular vaccine with high responder rate, strong on-target immunogenicity, and a potential of cognition improvement, which support UB-311 for active immunotherapy in early-to-mild AD patients currently enrolled in a phase-II trial (NCT02551809).

Keywords: Alzheimer's disease; Amyloid β vaccine; FIH clinical trial; UB-311; UBITh® platform.

Figures

Fig. 1
Fig. 1
Patient disposition and study design. The first-in-human (FIH) clinical trial enrolled 19 patients (50–80 years old) with mild-to-moderate Alzheimer's disease in a 24-week interventional study, and all 19 subjects received three UB-311 vaccine doses by intramuscular injection (300-μg immunogen peptide) at 0, 4, and 12 weeks and completed the treatment study (identifier no.: NCT00965588). After the first five subjects passed the safety evaluation at week 48, the remaining 14 subjects enrolled in and completed a 24-week observational extension study added to the end of the interventional study to monitor long-term safety, tolerability, immunogenicity, and efficacy (identifier no.: NCT01189084).
Fig. 2
Fig. 2
Serum anti-Aβ antibody titers assayed by ELISA and preferable targeted Aβ species visualized by dot plot after UB-311 immunization in AD patients. (A) Mean antibody response during the 24-week interventional study (solid line) in 19 AD patients treated with UB-311; 14 of 19 patients (dashed line) were followed up in an additional 24-week observational study, whose mean ADAS-Cog score was 4.9 (improvement). The mean baseline titer (pretreatment) was 1.0 log10; and 4 weeks after last vaccine boost at week 16, the titer peaked at 2.7 log10 (range: 1.8–3.7 log10). At week 48, all patients had decreasing but still positive antibody titers, measured by Aβ1–28 ELISA test. At week 16, serum samples recognized Aβ1–42 monomers (B) and oligomers (C); preimmune serum samples collected at week 0 had anti-Aβ1–42 antibody levels below quantification limit (not shown on log scale), except subjects P109 (monomer) and P108 (oligomer). (D) At week 16, analysis of serum dot plot (left panel), flanked by the positive control 6E10 mAb, reveals that the vaccine-induced anti-Aβ antibodies bind preferentially to Aβ fibrils, followed by oligomers, and the least to monomers; the densitometric measures (right panel) for the three Aβ species are the mean scales from three representative AD subjects, P105, P108, and P206. Abbreviations: AD, Alzheimer's disease; ADAS-Cog, Alzheimer's Disease Assessment Scale–Cognitive Subscale; ELISA, enzyme-linked immunosorbent assay.
Fig. 3
Fig. 3
Mean change over time in ADAS-Cog and MMSE scores by age, MMSE at baseline, or gender. For assessment of ADAS-cog and MMSE scores, UB-311-treated patients were stratified by (A and D) age in years (P < .0002 for the difference in the rate of disease progression. Abbreviations: ADAS-Cog, Alzheimer's Disease Assessment Scale–Cognitive Subscale; MMSE, Mini–Mental State Examination.

References

    1. Hardy J., Selkoe D.J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356.
    1. Kayed R., Lasagna-Reeves C.A. Molecular mechanisms of amyloid oligomers toxicity. J Alzheimers Dis. 2013;33:S67–S78.
    1. Kumar D.K., Choi S.H., Washicosky K.J., Eimer W.A., Ghofrani J., Leftkowitz A. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med. 2016;8:340ra72.
    1. Soscia S.J., Kirby J.E., Washicosky K.J., Tucker S.M., Ingelsson M., Hyman B. The Alzheimer's disease-associated amyloid-beta-protein is an antimicrobial peptide. PLoS One. 2010;5:e9505.
    1. Qian X.X., Hamad B., Dias-Lalcaca G. The Alzheimer disease market. Nat Rev Drug Discov. 2015;14:675–676.
    1. Schenk D., Barbour R., Dunn W., Gordon G., Grajeda H., Guido T. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999;400:173–177.
    1. Orgogozo J.M., Gilman S., Dartigues J.F., Laurent B., Puel M., Kirby L.C. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology. 2003;61:46–54.
    1. Gilman S., Koller M., Black R.S., Jenkins L., Griffith S.G., Fox N.C. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64:1553–1562.
    1. Pasquier F., Sadowsky C., Holstein A., Leterme G.L., Peng Y., Jackson N. Two phase 2 multiple ascending-dose studies of vautide cridificar (ACC-001) and QS-21 adjuvant in mild-to-moderate Alzheimer's disease. J Alzheimers Dis. 2016;51:1131–1143.
    1. Winblad B., Andreasen N., Minthon L., Floesser A., Imbert G., Dumortier T. Safety, tolerability and antibody response of active Aβ immunotherapy with CAD106 in patients with Alzheimer's disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol. 2012;11:597–604.
    1. Savage M.J., Wu G., McCampbell A., Wessner K.R., Citron M., Ling X. A novel multivalent Abeta peptide vaccine with preclinical evidence of a central immune response that generates antisera recognizing a wide range of Abeta peptide species. Alzheimers Dement. 2010;6:S142. Abstract No. O3-07-03.
    1. Hendrix S., Ellison N., Stanworth S., Tierney L., Mattner F., Schmidt W. Methodological aspects of the phase II study AFF006 evaluating amyloid-beta-targeting vaccine AFFITOPE AD02 in early Alzheimer's disease—prospective use of novel composite scales. J Prev Alzheimers Dis. 2015;2:91–102.
    1. Marciani D.J., Ellison N., Stanworth S., Tierney L., Mattner F., Schmidt W. A retrospective analysis of the Alzheimer's disease vaccine progress—the critical need for new development strategies. J Neurochem. 2016;137:687–700.
    1. Agadjanyan M.G., 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;11:1246–1259.
    1. Farlow M.R., Andreasen N., Riviere M.E., Vostiar I., Vitaliti A., Sovago J. Long-term treatment with Aβ immunotherapy with CAD106 in mild Alzheimer's disease. Alzheimers Res Ther. 2015;7:23.
    1. Vandenberghe R., Riviere M.E., Caputo A., Sovago J., Maguire R.P., Farlow M. Active Aβ immunotherapy CAD106 in Alzheimer's disease: A phase 2b study. Alzheimers Dement (N Y) 2017;3:10–22.
    1. Cummings J., Morstort T., Lee G. Alzheimer's drug development pipeline: 2016. Alzheimers Dement (N Y) 2016;2:222–232.
    1. Wang C.Y., Finstad C.L., Walfield A.M., Sia C., Sokoll K.K., Chang T.Y. Site-specific UBITh® amyloid-β vaccine for immunotherapy of Alzheimer's disease. Vaccine. 2007;25:3041–3052.
    1. Monsonego A., Zota V., Karni A., Krieger J.I., Bar-Or A., Bitan G. Increased T cell reactivity to amyloid beta protein in older humans and patients with Alzheimer disease. J Clin Invest. 2003;112:415–422.
    1. Greenberg S.M., Bacskai B.J., Hyman B.T. Alzheimer disease's double edged vaccine. Nat Med. 2003;9:389–390.
    1. Ghochikyan A., Mkrtichyan M., Petrushina I., Movsesyan N., Karapetyan A., Cribbs D.H. Prototype Alzheimer's disease epitope vaccine induced strong Th2-type anti-Abeta antibody response with Alum to Quil A adjuvant switch. Vaccine. 2006;24:2275–2282.
    1. Pellicano M., Bulati M., Buffa S., Barbagallo M., Di Prima A., Misiano G. Systemic immune responses in Alzheimer's disease: in vitro mononuclear cell activation and cytokine production. J Alzheimers Dis. 2010;21:181–192.
    1. Marciani D.J. Alzheimer's disease vaccine development: a new strategy focusing on immune modulation. J Neuroimmunol. 2015;287:54–63.
    1. Giunta B., Fernandez F., Nikolic W.V., Obregon D., Rrapo E., Town T. Inflammaging as a prodrome to Alzheimer's disease. J Neuroinflammation. 2008;5:51.
    1. Reale M., Iarlori C., Feliciani C., Gambi D. Peripheral chemokine receptors, their ligands, cytokines and Alzheimer's disease. J Alzheimers Dis. 2008;14:147–159.
    1. Montagne A., Barnes S.R., Sweeney M.D., Halliday M.R., Sagare A.P., Zhao Z. Blood-brain barrier breakdown in the aging human hippocampus. Neuron. 2015;85:296–302.
    1. Wang C.Y., Chang T.Y., Walfield A.M., Ye J., Shen M., Chen S.P. Effective synthetic peptide vaccine for foot-and-mouth disease in swine. Vaccine. 2002;20:2603–2610.
    1. Wang C.Y., Shen M., Tam G., Fang X.D., Ye J., Shen F. Synthetic AIDS vaccine by targeting HIV receptor. Vaccine. 2002;21:89–97.
    1. Wang C.Y., Walfield A.M., Fang X.D., Hammerberg B., Ye J., Li M.L. Synthetic IgE peptide vaccine for immunotherapy of allergy. Vaccine. 2003;21:1580–1590.
    1. Wang C.Y., Walfield A.M. Site-specific peptide vaccines for immunotherapy and immunization against chronic diseases, cancer, infectious diseases, and for veterinary applications. Vaccine. 2005;23:2049–2056.
    1. Bayer A.J., Bullock R., Jones R.W., Wilkinson D., Paterson K.R., Jenkins L. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology. 2005;64:94–101.
    1. Ghiso J., Frangione B. Amyloidosis and Alzheimer's disease. Adv Drug Deliv Rev. 2002;54:1539–1551.
    1. Zlokovic B.V. Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders. Nat Rev Neurosci. 2011;12:723–738.
    1. Masters C.L., Simms G., Weinman N.A., Multhaup G., McDonald B.L., Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A. 1985;82:4245–4249.
    1. DeMattos R.B., Bales K.R., Cummins D.J., Dodart J.C., Paul S.M., Holtzman D.M. Peripheral anti-Abeta antibody alters CNS and plasma Abeta clearance and decreases brain A beta burden in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A. 2001;98:8850–8855.
    1. Zhao L.N., Long H., Mu Y., Chew L.Y. The toxicity of amyloid oligomers. Int J Mol Sci. 2012;13:7303–7327.
    1. Kayed R., Head E., Thompson J.L., McIntire T.M., Milton S.C., Cotman C.W. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300:486–489.
    1. Shankar G.M., Li S., Mehta T.H., Garcia-Munoz A., Shepardson N.E., Smith I. Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–842.
    1. Stancu I.C., Vasconcelos B., Terwel D., Dewachter I. Models of β-amyloid induced Tau-pathology: the long and “folded” road to understand the mechanism. Mol Neurodegener. 2014;9:51.
    1. Parajuli B., Sonobe Y., Horiuchi H., Takeuchi H., Mizuno T., Suzumara A. Oligomeric amyloid beta induces IL-1β processing via production of ROS: implication in Alzheimer's disease. Cell Death Dis. 2013;4:e975.
    1. Wisniewski T., Goni F. Immunotherapeutic approaches for Alzheimer's disease. Neuron. 2015;85:1162–1176.
    1. Wisniewski T., Drummond E. Developing therapeutic vaccines against Alzheimer's disease. Expert Rev Vaccines. 2016;15:401–415.
    1. Salloway S., Sperling R., Fox N.C., Blennow K., Klunk W., Raskind M. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med. 2014;370:322–333.
    1. Siemers E.R., Sundell K.L., Carlson C., Case M., Sethuraman G., Liu-Seifert H. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer's disease patients. Alzheimers Dement. 2016;12:110–120.
    1. Sperling R., Salloway S., Brooks D.J., Tampieri D., Barakos J., Fox N.C. Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis. Lancet Neurol. 2012;11:241–249.
    1. Sevigny J., Chiao P., Bussiere T., Weinreb P.H., Williams L., Maier M. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature. 2016;537:50–56.
    1. Bouter Y., Lopez Noguerola J.S., Tucholla P., Crespi G.A., Parker M.W., Wiltfang J. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against N-truncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol. 2015;130:713–729.
    1. Reiman E.M. Attack on amyloid-β protein. Nature. 2016;537:36–37.
    1. Zempel H., Luedtke J., Kumar Y., Biermat J., Dawson H., Mandelkow E. Amyloid-β oligomers induce synaptic damage via tau-dependent microtubule severing by TTLL6 and spastin. EMBO J. 2013;32:2920–2937.
    1. Seward M.E., Swanson E., Norambuena A., Reimann A., Cochran J.N., Li R. Amyloid-β signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer's disease. J Cell Sci. 2013;126:1278–1286.
    1. Wang Y., Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17:5–21.

Source: PubMed

3
Se inscrever