Multi-subunit BCG booster vaccine GamTBvac: Assessment of immunogenicity and protective efficacy in murine and guinea pig TB models

A P Tkachuk, V A Gushchin, V D Potapov, A V Demidenko, V G Lunin, A L Gintsburg, A P Tkachuk, V A Gushchin, V D Potapov, A V Demidenko, V G Lunin, A L Gintsburg

Abstract

New innovative vaccines are highly needed to combat the global threat posed by tuberculosis. Efficient components-antigens and adjuvants-are crucial for development of modern recombinant TB vaccines. This study describes a new vaccine (GamTBvac) consisting of two mycobacterial antigen fusions (Ag85A and ESAT6-CFP10)-with dextran-binding domain immobilized on dextran and mixed with an adjuvant consisting of DEAE-dextran core, and with CpG oligodeoxynucleotides (TLR9 agonists). GamTBvac and its components were assessed for immunogenicity and protective efficacy in GamTBvac-prime/boost and BCG-prime/ GamTBvac-boost in murine and guinea pig TB models. Results show that in both infectious models, GamTBvac has a strong immunogenicity and significant protective effect against Mycobacterium tuberculosis strain H37Rv under aerosol and intravenous challenges. GamTBvac showed a particularly strong protective effect as a BCG booster vaccine.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Expression and purification of GamTBvac…
Fig 1. Expression and purification of GamTBvac components.
Two antigen fusion proteins DBD-ESAT6-CFP10 and DBD-Ag85A (A) were induced by IPTG in E. coli BL21. Coomassie blue staining was used for evaluation of inclusion bodies washing results (B) and column purification process (C); M—molecular weight marker, IB—inclusion bodies, W1-2 –wash of IB with 25 mM Tris pH 7.5 + 50 mM NaCl, W3 –wash of IB with 25 mM Tris pH 7.5 + 50 mM NaCl + 25%EtOH, U– 8M urea wash fractions, T—tail water elution fraction, P—peak water elution fraction, E—elution fractions. Both recombinant fusions were checked by western blotting (D) with rabbit antisera (for rAg85A) or antigen specific monoclonal antibodies (rESAT6 and rCFP10)
Fig 2. Immunogenicity of GamTBvac components in…
Fig 2. Immunogenicity of GamTBvac components in GamTBvac-prime/boost and BCG-prime/GamTBvac-boost regimens.
The popliteal lymph node (A, C) and spleen (B, D) cells (0.5–8×105/well) of the vaccinated mice were stimulated with DBD-Ag85A (10 μg/ml; A, B), DBD-ESAT6-CFP10 (10 μg/ml; C, D) separately in vitro and number of IFNγ secreting cells was assessed by ELISPOT. To estimate the lymphoproliferative potential of vaccine components, the popliteal lymph node cells of the vaccinated mice (2–4×105 cells/well) were stimulated with DBD-Ag85A (10 μg/ml), DBD-ESAT6-CFP10 (10 μg/ml) or PBS separately in vitro (E) five weeks after the final immunization with specific antigen composition. Total IFNγ secretion in splenocytes upon different vaccination regimens was estimated by ELISA (F). To evaluate the antigen-specific antibody titer, sera were collected from each mouse one week after last immunization. Endpoint IgG titers specific to ESAT6, CFP10 and Ag85a were detected by ELISA. Sera from mice treated with PBS or adjuvant (without antigen) were used as a negative control (G). Results were presented as group means ± 95%CIs, n = 4, * P<0.05, ** P<0.01. In graphs F and C asterisk indicate significant differences compare to both PBS and BCG groups.
Fig 3. Protective efficacy of GamTBvac against…
Fig 3. Protective efficacy of GamTBvac against M. tuberculosis H37Rv infection in murine and guinea pig models.
The mice were immunized subcutaneously with GamTBvac and BCG (5×106 CFU) in different regimens. BCG was administered once at the 1st week and GamTBvac twice (three weeks apart). The control group was injected with PBS. For the acute murine model, the mice were retro-orbitally infected with MTB H37Rv two weeks after the last immunization. The results presented are given as mean Log10 CFU per organ ± 95%CI from groups of seven mice (n = 7), * P<0.05, relative to PBS; ** P<0.05, relative to both PBS and BCG. Thirty days after infection, protective efficacy was measured (n = 6) in lungs (A) and spleens (B). For the aerosol murine model, the mice were infected in aerosol infection chamber to get 103 CFU per animal sixty days after the last immunization. Four weeks later, bacterial load (n = 6) was determined in lungs (C) and spleens (D). Aerosol infection (n = 15) was monitored for more than 400 days until all mice died (E).

References

    1. WHO, Global Tuberculosis Report 2016. 21th edition France: World Health Organisation; 2016;214
    1. Andersen P, Urdahl KB. TB vaccines; promoting rapid and durable protection in the lung. Curr Opin Immunol. Elsevier Ltd; 2015;35: 55–62.
    1. Glaziou P, Sismanidis C, Floyd K, Raviglione M. Global Epidemiology of Tuberculosis. Cold Spring harb Perspec Med. 2015; 1–18.
    1. Karp CL, Wilson CB, Stuart LM. Tuberculosis vaccines: Barriers and prospects on the quest for a transformative tool. Immunol Rev. 2015;264: 363–381. 10.1111/imr.12270
    1. Andersen P, Doherty TM. The success and failure of BCG—implications for a novel tuberculosis vaccine. Nature reviews Microbiology. 2005; 3(8):656–62. 10.1038/nrmicro1211
    1. Pang Y, Zhao A, Cohen C, Kang W, Lu J, Wang G, et al. Current status of new tuberculosis vaccine in children. Hum Vaccin Immunother. 2016;5515: 1–11.
    1. Fine PE Variation in protection by BCG: implications of and for heterologous immunity. Lancet 1995;346: 1339–1345. S0140-6736(95)92348-9.
    1. Nguipdop-Djomo P., Heldal E., Rodrigues L. C., Abubakar I., & Mangtani P. Duration of BCG protection against tuberculosis and change in effectiveness with time since vaccination in Norway: A retrospective population-based cohort study. The Lancet Infectious Diseases 2016;16(2): 219–226. 10.1016/S1473-3099(15)00400-4
    1. Skeiky YAW, Dietrich J, Lasco TM, Stagliano K, Dheenadhayalan V, Goetz MA, et al. Non-clinical efficacy and safety of HyVac4:IC31 vaccine administered in a BCG prime-boost regimen. Vaccine. 2010;28: 1084–1093. 10.1016/j.vaccine.2009.10.114
    1. Rodrigues LC, et al. Effect of BCG revaccination on incidence of tuberculosis in school-aged children in Brazil: the BCG-REVAC cluster-randomised trial. Lancet 2005;366(9493):1290–5. 10.1016/S0140-6736(05)67145-0
    1. Frick M. The Tuberculosis Vaccines Pipeline: A New Path to the Same Destination? HIV, HCV & TB Pipeline Report (Treatment Action Group). HIV& TB (2015). July 2015. . Cited 25 August 2016.
    1. Agger EM. Novel adjuvant formulations for delivery of anti-tuberculosis vaccine candidates. Adv Drug Deliv Rev. Elsevier B.V.; 2015;
    1. Carpenter C, Sidney J, Kolla R, Nayak K, Tomiyama H, Tomiyama C, et al. A side-by-side comparison of T cell reactivity to fifty-nine Mycobacterium tuberculosis antigens in diverse populations from five continents. Tuberculosis. Elsevier Ltd; 2015;95: 713–721.
    1. Xin Q, Niu H, Li Z, Zhang G, Hu L, Wang B, et al. Subunit Vaccine Consisting of Multi-Stage Antigens Has High Protective Efficacy against Mycobacterium tuberculosis Infection in Mice. PLoS One. 2013;8: 1–12.
    1. Niu H, Peng J, Bai C, Liu X, Hu L, Luo Y, et al. Multi-stage tuberculosis subunit vaccine candidate LT69 provides high protection against Mycobacterium tuberculosis infection in mice. PLoS One. 2015;10: 1–11.
    1. Baldwin SL, Bertholet S, Reese VA, Ching LK, Reed SG, Coler RN. The importance of adjuvant formulation in the development of a TB vaccine. J Immunol. 2012;188: 2189–2197. 10.4049/jimmunol.1102696
    1. Tukhvatulin AI, Dzharullaeva AS, Tukhvatulina NM, Shcheblyakov D V., Shmarov MM, Dolzhikova I V., et al. Powerful Complex Immunoadjuvant Based on Synergistic Effect of Combined TLR4 and NOD2 Activation Significantly Enhances Magnitude of Humoral and Cellular Adaptive Immune Responses. PLoS One. 2016;11: e0155650 10.1371/journal.pone.0155650
    1. Knudsen NPH, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, et al. Different human vaccine adjuvants promote distinct antigen- independent immunological signatures tailored to different pathogens. 2016; 1–13.
    1. Cordeiro AS, Alonso MJ, de la Fuente M. Nanoengineering of vaccines using natural polysaccharides. Biotechnol Adv. Elsevier B.V.; 2015;33: 1279–1293.
    1. Cox J.C., Coulter A.R. Adjuvant—a classification and rewiew of their modes of action. Vaccine. 1997;15 (3) 248–256.
    1. Ershova AS, Gra OA, Lyaschuk AM, Grunina TM, Tkachuk AP, Bartov MS, et al. Recombinant domains III of Tick-Borne Encephalitis Virus envelope protein in combination with dextran and CpGs induce immune response and partial protectiveness against TBE virus infection in mice. BMC Infect Dis. 2016;16: 544 10.1186/s12879-016-1884-5
    1. Tkachuk A., Lunin V., Karyagina A., Gintsburg A. Problems and prospects of development of the subunit TB vaccine. J Acquir Immune Defic Syndr 2014; 65(Suppl 2): 43.
    1. Cumming G, Fidler F, Vaux DL. Error bars in experimental biology. J Cell Biol. 2007;177: 7–11. 10.1083/jcb.200611141
    1. Olsen AW, Pinxteren L, Okkels LM, Rasmussen PB, Andersen P. Protection of Mice with a Tuberculosis Subunit Vaccine Based on a Fusion Protein of Antigen 85B and ESAT-6. Microbiology. 2001;69: 2773–2778.
    1. Agger EM, Rosenkrands I, Olsen AW, Hatch G, Williams A, Kritsch C, et al. Protective immunity to tuberculosis with Ag85B-ESAT-6 in a synthetic cationic adjuvant system IC31. Vaccine. 2006;24: 5452–5460. 10.1016/j.vaccine.2006.03.072
    1. Reither K, Katsoulis L, Beattie T, Gardiner N, Lenz N, Said K, et al. Safety and immunogenicity of h1/ic31h, an adjuvanted tb subunit vaccine, in hiv-infected adults with cd4+ lymphocyte counts greater than 350 cells/mm3: A phase ii, multi-centre, double-blind, randomized, placebo-controlled trial e114602. PLoS One. 2014;9: 1–19.
    1. Geldenhuys H, Mearns H, Miles DJC, Tameris M, Hokey D, Shi Z, et al. The tuberculosis vaccine H4: IC31 is safe and induces a persistent polyfunctional CD4 T cell response in South African adults: A randomized controlled trial. Vaccine. Elsevier Ltd; 2015;33: 3592–3599.
    1. Aagaard C, Hoang T, Dietrich J, Cardona P-J, Izzo A, Dolganov G, et al. A multistage tuberculosis vaccine that confers efficient protection before and after exposure. Nat Med. Nature Publishing Group; 2011;17: 189–194.
    1. Billeskov R, Elvang TT, Andersen PL, Dietrich J. The HyVac4 subunit vaccine efficiently boosts BCG-primed anti-mycobacterial protective immunity. PLoS One. 2012;7.
    1. Woodworth JS, Cohen SB, Moguche AO, Plumlee CR, Agger EM, Urdahl KB, et al. Subunit vaccine H56/CAF01 induces a population of circulating CD4 T cells that traffic into the Mycobacterium tuberculosis-infected lung. Mucosal Immunol. Nature Publishing Group; 2016;
    1. Li W, Li M, Deng G, Zhao L, Liu X, Wang Y. Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice. Mol Med Rep. 2015;12: 3073–3080. 10.3892/mmr.2015.3770
    1. Chen YY, Lin CW, Huang WF, Chang JR, Su IJ, Hsu CH, et al. Recombinant bacille Calmette-Guerin coexpressing Ag85b, CFP10, and interleukin-12 elicits effective protection against Mycobacterium tuberculosis. J Microbiol Immunol Infect. Elsevier Taiwan LLC; 2014; 1–7.
    1. Hoft DF, Blazevic A, Selimovic A, Turan A, Tennant J, Abate G, et al. Safety and Immunogenicity of the Recombinant BCG Vaccine AERAS-422 in Healthy BCG-naïve Adults: A Randomized, Active-controlled, First-in-human Phase 1 Trial. EBioMedicine. The Authors; 2016;7: 278–286.
    1. Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PEM, et al. Protection by BCG vaccine against tuberculosis: A systematic review of randomized controlled trials. Clin Infect Dis. 2014;58: 470–480. 10.1093/cid/cit790
    1. Abubakar I, Pimpin L, Ariti C, Beynon R, Mangtani P, Sterne J, et al. Systematic review and meta-analysis of the current evidence on the duration of protection by bacillus Calmette-Gue rin vaccination against tuberculosis. Health Technol Assess (Rockv). 2013;17: 1–4.
    1. Aronson NE, Santosham Mathuram, Comstock GW, Howard RS, Moulton LH, Everett R. Rhoades, et al. Long-term Efficacy of BCG Vaccine. 2004;291: 2086–2091.
    1. Hoft DF, Blazevic A, Selimovic A, Turan A, Tennant J, Abate G, et al. Safety and Immunogenicity of the Recombinant BCG Vaccine AERAS-422 in Healthy BCG-naïve Adults: A Randomized, Active-controlled, First-in-human Phase 1 Trial. EBioMedicine. The Authors; 2016;7: 278–286.
    1. Mohamed W, Domann E, Chakraborty T, Mannala G, Lips KS, Heiss C, et al. TLR9 mediates S. aureus killing inside osteoblasts via induction of oxidative stress. BMC Microbiol. BMC Microbiology; 2016; 1–8.
    1. Pustylnikov S, Sagar D, Jain P, Khan ZK. Targeting the C-type Lectins-Mediated Host-Pathogen Interactions with Dextran. 2014;17: 371–392.
    1. Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, Shea JE, McClain JB, Hussey GD, Hanekom WA et al.: Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomised, placebo-controlled phase 2b trial. Lancet 2013;381:1021–1028. 10.1016/S0140-6736(13)60177-4
    1. Beverley P: TB vaccine failure was predictable. Nature. 2013;503:p469;
    1. van Dissel JT, Arend SM, Prins C, Bang P, Tingskov PN, Lingnau K, et al. Ag85B-ESAT-6 adjuvanted with IC31 promotes strong and long-lived Mycobacterium tuberculosis specific T cell responses in naive human volunteers. Vaccine. 2010;28: 3571–3581. 10.1016/j.vaccine.2010.02.094
    1. van Dissel JT, Soonawala D, Joosten SA, Prins C, Arend SM, Bang P, et al. Ag85B-ESAT-6 adjuvanted with IC31 promotes strong and long-lived Mycobacterium tuberculosis specific T cell responses in volunteers with previous BCG vaccination or tuberculosis infection. Vaccine. Elsevier Ltd; 2011;29: 2100–2109.
    1. Van Zyl-Smit RN, Pai M, Peprah K, Meldau R, Kieck J, et al. Within-subject variability and boosting of T-cell interferon-gamma responses after tuberculin skin testing. Am J Respir Crit Care Med 2009;180: 49–58. 10.1164/rccm.200811-1704OC
    1. Frick B. M. The Tuberculosis Prevention Pipeline. 2016:143–162.
    1. Millington KA, Fortune SM, Low J, Garces A, Hingley-Wilson SM, et al. Rv3615c is a highly immunodominant RD1 (Region of Difference 1)-dependent secreted antigen specific for Mycobacterium tuberculosis infection. Proc Natl Acad Sci U S A 2011;108: 5730–5735. 10.1073/pnas.1015153108

Source: PubMed

3
Tilaa