Novel single-chain antibodies against highly conserved epitopes in the hemagglutinin of influenza A viruses: Promising agents for universal therapies

Document Type : Original Article

Authors

1 Recombinant Antibody Laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran

2 Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver BC, Canada

3 Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran

4 HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

Objective(s): Development of new antibodies with broad activity would provide anti-influenza prophylaxis and treatment. Human single-chain variable fragments (scFvs) are considered effective agents against viruses. In this study specific human scFvs against highly conserved epitopes in the hemagglutinin (HA) of influenza A viruses were selected and their neutralizing activity was evaluated.
Materials and Methods: Bioinformatic methods were used to evaluate HA epitopes. The panning process selected specific clones from a scFv library. PCR and DNA fingerprinting differentiated the common patterns. Soluble forms of scFvs were produced and evaluated using Western blot analysis. The neutralizing effects of anti-HA scFvs were assessed by microneutralization assay using MDCK cells. Real-time PCR was done to determine the exact copy number of the virus following neutralization.
Results: Bioinformatic evaluation confirmed the antigenicity and accessibility of the epitopes. Four specific anti-HA scFvs, scFvs I, II, I’, and II’ were selected. The scFvs neutralized 2009 H1N1 pandemic and 83.34%, 79.17%, 75%, and 62.5% reduction in the virus titers were obtained following treatments with scFv-II′, I, I′, and II, respectively. Real-time PCR demonstrated 98.6%, 95.7%, 95.26%, and 91.19% reductions in virus numbers following neutralization with scFv-II′, I, I′, and II, respectively.
Conclusion: Anti-HA scFvs selected against highly conserved HA of influenza A virus with high neutralizing effects, offer novel human antibodies for prophylaxis and treatment of a wide range of influenza viruses including different subtypes of H1N1, H3N2, and H5N1 influenza A virus. The antibodies have the potential to be used for universal therapy.

Keywords


1.    Zhou F, Trieu MC, Davies R, Cox RJ. Improving influenza vaccines: challenges to effective implementation. Curr Opin Immunol 2018; 53:88-95.
2.    Engels G, Hierweger AM, Hoffmann J, Thieme R, Thiele S, Bertram S, et al. Pregnancy-related immune adaptation promotes the emergence of highly virulent H1N1 influenza virus strains in allogenically pregnant mice. Cell Host Microbe 2017; 21: 321-333.
3.    Ravindran M, Gowtham S, Mehta P, Narayanan P. Retrospective analysis of maternal and foetal outcome of H1N1 influenza amongst antenatal mothers at a tertiary care hospital. J Patient Saf  2017; 5: 69-72.
4.    Huang Q, Sivaramakrishna RP, Ludwig K, Korte T, Böttcher C, Herrmann A. Early steps of the conformational change of influenza virus hemagglutinin to a fusion active state: stability and energetics of the hemagglutinin. Biochim Biophys Acta 2003; 1614: 3-13.
5.    Wiley D, Wilson I, Skehel J. Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature 1981; 289: 373-378.
6.    Kubota-Koketsu R, Mizuta H, Oshita M, Ideno S, Yunoki M, Kuhara M, et al. Broad neutralizing human monoclonal antibodies against influenza virus from vaccinated healthy donors. Biochem Biophys Res Commun 2009; 387:180-185.
7.    Yamashita A, Kawashita N, Kubota-Koketsu R, Inoue Y, Watanabe Y, Ibrahim MS, et al. Highly conserved sequences for human neutralization epitope on hemagglutinin of influenza A viruses H3N2, H1N1 and H5N1: Implication for human monoclonal antibody recognition. Biochem Biophys Res Commun  2010; 393: 614-618.
8.    Hu W. Highly conserved domains in hemagglutinin of influenza viruses characterizing dual receptor binding. Nat Sci 2010; 2: 1005-1014.
9.    Moattari A, Emami A,  Amini E. Influenza A virus (A/Shiraz/8/2010(H1N1)) segment 4 hemagglutinin (HA) gene, complete cds [Internet]. 2010. Available from: https://www.ncbi.nlm.nih.gov/nuccore/309401688.
10.    Mohammadi S, Hosseinzadeh F, Nejatollahi F. Production of specific anti-EGFR single chain antibodies: a promising strategy in the immunotherapy of EGFR expressing tumor tissues. Int J Cancer Manag 2017; 10: e6666.
11.    Ehsaei B, Nejatollahi F, Mohammadi M. Specific single chain antibodies against a neuronal growth inhibitor receptor, nogo receptor 1: Promising new antibodies for the immunotherapy of Multiple Sclerosis. Shiraz E-Med J 2017;18: e45358.
12.    Nejatollahi F, Silakhori S, Moazen B. Isolation and evaluation of specific human recombinant antibodies from a phage display library against her3 cancer signaling antigen. Middle East J Cancer 2014; 5: 137-144.
13.    Bagheri V, Nejatollahi F, Esmaeili SA, Momtazi AA, Motamedifar M, Sahebkar A. Neutralizing human recombinant antibodies against herpes simplex virus type 1 glycoproteins B from a phage-displayed scFv antibody library. Life Sci 2017; 169:1-5.
14.    Moazen B, Ebrahimi E, Nejatollahi F. Single chain antibodies against gp55 of human cytomegalovirus (HCMV) for prophylaxis and treatment of HCMV infections. Jundishapur J Microbiol 2016; 9: e16241.
15.    Foster LH, Lum LG. Treatment of hematological malignancies with T cell redirected bispecific antibodies: current status and future needs. Expert Opin Biol Ther 2019; 19: 707-720.
16.    Nejatollahi F, Bayat P, Moazen B. Cell growth inhibition and apoptotic effects of a specific anti-RTFscFv antibody on prostate cancer, but not glioblastoma, cells. F1000Research 2017; 6:156-167.
17.    Hosseinzadeh F, Mohammadi S, Nejatollahi F. Production and evaluation of specific single-chain antibodies against CTLA-4 for cancer-targeted therapy. Rep Biochem Mol Biol 2017; 6: 8–14.
18.    Zarei N, Fazeli M, Mohammadi M, Nejatollahi F. Cell growth inhibition and apoptosis in breast cancer cells induced by anti-FZD7 scfvs: Involvement of bioinformatics-based design of novel epitopes. Breast Cancer Res and Treat 2018;169:427-436.
19.     Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019;7:4000-4016.
20.    Nejatollahi F, Hodgetts SJ, Vallely PJ, Burnie JP. Neutralising human recombinant antibodies to human cytomegalovirus glycoproteins gB and gH. FEMS FEMS Immunol Med Microbiol 2002; 34: 237-44.
21.    Truelove S, Zhu H, Lessler J, Riley S, Read JM, Wang S, et al. A comparison of hemagglutination inhibition and neutralization assays for characterizing immunity to seasonal influenza A. Influenza Other Respir Viruses 2016; 10: 518-524.
22.     Balcioglu BK, Ozdemir-Bahadir A, Hinc D, Tamerler C, Erdag B. Cost effective filamentous phage based immunization nanoparticles displaying a full-length hepatitis B virus surface antigen. Adv Biosci Biotechnol 2014; 5: 46-53
23.    Rudraraju R, Subbarao K. Passive immunization with influenza haemagglutinin specific monoclonal antibodies. Hum Vaccin Immunother 2018; 14:2728-2736.
24.    Sparrow E, Friede M, Sheikh M, Torvaldsen S, Newall AT. Passive immunization for influenza through antibody therapies, a review of the pipeline, challenges and potential applications. Vaccine 2016; 34: 5442-5448.
25.    Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov 2006; 5: 997-1014.
26.    Le Gall F, Reusch U, Little M, Kipriyanov SM et al. Effect of linker sequences between the antibody variable domains on the formation, stability and biological activity of a bispecific tandem diabody. Protein Eng Des Sel 2004; 17: 357-366.
27.     Pissawong T, Maneewatch S, Thueng-in K, Srimanote P, Thanongsaksrikul J, Songserm T, et al. Human monoclonal ScFv that bind to different functional domains of M2 and inhibit H5N1 influenza virus replication. Virol J 2013; 10:148-161.
28.    Zhang X, Qi X, Zhang Q, Zeng X, Shi Z, Jin Q, et al. Human 4F5 single-chain Fv antibody recognizing a conserved HA1 epitope has broad neutralizing potency against H5N1 influenza A viruses of different clades. Antiviral Res 2013; 99 :91-99.
29.    He FT, Nie YZ, Chen BJ, Qiao TD, Fan DM, Li RF, et al. Expression and identification of recombinant soluble single-chain variable fragment of monoclonal antibody MC3. World J Gastroenterol 2002; 8: 258-262.
30.    Cheng Y, Li Z, Xi H, Gu T, Yuan R, Chen X, et al. A VL-linker-VH orientation dependent single chain variable antibody fragment against rabies virus G protein with enhanced neutralizing potency in vivo. Protein Pept Lett  2016; 23: 24-32.
31.    Li TW, Cheng SF, Tseng YT, Yang YC, Liu WC, Wang SC, et al. Development of single-chain variable fragments (scFv) against influenza virus targeting hemagglutinin subunit 2 (HA2). Arch Virol 2016; 161:19-31.
32.    Teferedegne B, Lewis AM, Peden K., Murata H. Development of a neutralization assay for influenza virus using an endpoint assessment based on quantitative reverse-transcription PCR. PLoS One 2013; 8:e56023.
33.    Ellis J, Iturriza M, Allan R, Bermingham A, Brown K, Gray J, Brown D. Evaluation of four real-time PCR assays for detection of influenza A (H1N1) v viruses. Euro Surveill 2009; 14: pii: 19230.
34.    Faye O, Faye O, Diallo D, Diallo M, Weidmann M. Quantitative real-time PCR detection of Zika virus and evaluation with field-caught mosquitoes. Virol J 2013; 22:311-318.
35.    Mackay M. Real-time PCR in the microbiology laboratory. Clin Microbiol Infect 2004; 10:190-212.
36.    Greninger AL, Bateman AC, Atienza EE, Wendt S, Makhsous N, Jerome KR, et al. Copy number heterogeneity of JC virus standards. J Clin Microbiol 2017; 55:824–831.
37.    Zhu W, Xie K, Xu Y, Wang L, Chen K, Zhang L, et al. CRISPR/Cas9 produces anti-hepatitis B virus effect in hepatoma cells and transgenic mouse. Virus Res 2016; 217:125-132.
38.    Inazawa N, Hori T, Yamamoto M, Hatakeyama N, Yoto Y, Nojima M, et al. HHV‐6 encephalitis may complicate the early phase after allogeneic hematopoietic stem cell transplantation: Detection by qualitative multiplex PCR and subsequent quantitative real‐time PCR. J Med Virol 2016; 88: 319-323.
39.    Kyriakis CS, Zhang M, Wolf S, Jones LP, Shim BS, Chocallo AH, et al. Molecular epidemiology of swine influenza A viruses in the Southeastern United States, highlights regional differences in circulating strains. Vet Microbiol 2017; 211:174-179.
40.    Itoh Y, Yoshida R, Shichinohe S, Higuchi M, Ishigaki H, Nakayama M, Ishida H, Kitano M, Arikata M, Kitagawa N. Protective efficacy of passive immunization with monoclonal antibodies in animal models of H5N1 highly pathogenic avian influenza virus infection. PLoS Pathog 2014; 10: e1004192.
41.    Liu Q, Zhou Yh, Yang Zq. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol 2016; 13: 3-10.
42.    Krammer F, Palese P. Influenza virus hemagglutinin stalk-based antibodies and vaccines. Curr Opin Virol  2013; 3: 521-530.
43.    Ahmad ZA, Yeap SK, Ali AM, Ho WY, Alitheen NBM, Hamid M. scFv antibody: principles and clinical application. Clin Dev Immunol 2012; 980250.