The influence of BCG vaccine strain on mycobacteria-specific and non-specific immune responses in a prospective cohort of infants in Uganda

Elizabeth J Anderson, Emily L Webb, Patrice A Mawa, Moses Kizza, Nancy Lyadda, Margaret Nampijja, Alison M Elliott, Elizabeth J Anderson, Emily L Webb, Patrice A Mawa, Moses Kizza, Nancy Lyadda, Margaret Nampijja, Alison M Elliott

Abstract

Background: Globally, BCG vaccination varies in efficacy and has some non-specific protective effects. Previous studies comparing BCG strains have been small-scale, with few or no immunological outcomes and have compared TB-specific responses only. We aimed to evaluate both specific and non-specific immune responses to different strains of BCG within a large infant cohort and to evaluate further the relationship between BCG strain, scarring and cytokine responses.

Methods: Infants from the Entebbe Mother and Baby Study (ISRCTN32849447) who received BCG-Russia, BCG-Bulgaria or BCG-Denmark at birth, were analysed by BCG strain group. At one year, interferon-gamma (IFN-γ), interleukin (IL)-5, IL-13 and IL-10 responses to mycobacteria-specific antigens (crude culture filtrate proteins and antigen 85) and non-mycobacterial stimuli (tetanus toxoid and phytohaemagglutinin) were measured using ELISA. Cytokine responses, scar frequency, BCG associated adverse event frequency and mortality rates were compared across groups, with adjustments for potential confounders.

Results: Both specific and non-specific IFN-γ, IL-13 and IL-10 responses in 1341 infants differed between BCG strain groups including in response to stimulation with tetanus toxoid. BCG-Denmark immunised infants showed the highest cytokine responses. The proportion of infants who scarred differed significantly, with BCG scars occurring in 52.2%, 64.1% and 92.6% of infants immunised with BCG Russia, BCG-Bulgaria and BCG-Denmark, respectively (p<0.001). Scarred infants had higher IFN-γ and IL-13 responses to mycobacterial antigens only than infants without a scar. The BCG-Denmark group had the highest frequency of adverse events (p=0.025). Mortality differences were not significant.

Conclusions: Both specific and non-specific immune responses to the BCG vaccine differ by strain. Scarring after BCG vaccination is also strain-dependent and is associated with higher IFN-γ and IL-13 responses to mycobacterial antigens. The choice of BCG strain may be an important factor and should be evaluated when testing novel vaccine strategies that employ BCG in prime-boost sequences, or as a vector for other vaccine antigens.

Copyright © 2012 Elsevier Ltd. All rights reserved.

References

    1. Calmette A., Guérin C., Boquet A., Négre L. Sur la vaccination préventive des enfants nouveaux-nés contre la tuberculose par le BCG. Ann Inst Pasteur. 1927;41:201–231.
    1. WHO-UNICEF Estimates of BCG Coverage. Data Source: WHO Vaccine-preventable Diseases: Monitoring System 2010 Global Summary. Available at: [accessed 16.04.2011].
    1. Behr M.A., Small M.A. A historical and molecular phylogeny of BCG strains. Vaccine. 1999;17:915–922.
    1. Brosch R., Gordon S.V., Garnier T., Eiglmeier K., Frigui W., Valenti P. Genome plasticity of BCG and impact on vaccine efficacy. PNAS. 2007;104(13):5596–5601.
    1. Ritz N., Curtis N. Mapping the global use of different BCG vaccine strains. Tuberculosis. 2009;89:248–251.
    1. Brewer T.F. Preventing tuberculosis with Bacillus Calmette–Guérin vaccine: a meta-analysis of the literature. CID. 2000;31(3):S64–S67.
    1. Colditz G.A., Berkey C.S., Mosteller F., Brewer T.F., Wilson M.E., Burdick E. The efficacy of bacillus Calmette–Guerin vaccination of newborns and infants in the prevention of tuberculosis: meta-analyses of the published literature. Pediatrics. 1995;96(1):29–35.
    1. Fine P.E. Variation in protection by BCG: implications of and for heterologous immunity. Lancet. 1995;346(8986):1339–1345.
    1. Wilson M.E., Fineberg H.V., Colditz G.A. Geographic latitude and the efficacy of Bacillus Calmette–Guerin vaccine. CID. 1995;20:982–991.
    1. Elliott A.M., Mawaa P.A., Webb E.L., Nampijjaa M. Effects of maternal and infant co-infections, and of maternal immunisation, on the infant response to BCG and tetanus immunization. Vaccine. 2011;29:247–255.
    1. Ritz N., Hanekom W.A., Robins-Browne R., Britton W.J., Curtis N. Influence of BCG vaccine strain on the immune response and protection against tuberculosis. FEMS Microbiol Rev. 2008;32:821–841.
    1. Davids V., Hanekom W.A., Mansoor N., Gamieldien H., Gelderbloem S.J., Hawkridge A. The effect of Bacille Calmette–Guerin vaccine strain and route of administration on induced immune responses in vaccinated infants. JID. 2006;193:531–536.
    1. Hussey G.D., Watkins M.L.V., Goddard E.A., Gottschalk S., Hughes E.J., Iloni K. Neonatal mycobacterial specific cytotoxic T-lymphocyte and cytokine profiles in response to distinct BCG vaccination strategies. Immunology. 2002;105:314–324.
    1. Roth A., Sodemann M., Jensen H., Poulsen A., Gustafson P., Weise C. Tuberculin reaction, BCG scar, and lower female mortality. Epidemiology. 2006;17:562–568.
    1. Guerin N., Teulieres L., Noba A., Schlumberger M., Bregère P., Chauvin P. Comparison of the safety and immunogenicity of the lyophilized Merieux seed and the World Health Organization working reference BCG vaccines in school-aged children in Senegal. Vaccine. 2009;17:105–109.
    1. Wu B., Huang C., Garcia L., Ponce de Leon A., Sifuentes Osornio J., Bobadilla-del-Valle M. Unique gene expression profiles in infants vaccinated with different strains of Mycobacterium bovis Bacille Calmette–Guerin. Infect Immun. 2007;75:3658–3664.
    1. Gorak-Stolinska P., Weir R.E., Floyd S., Lalor M.K., Stenson S., Branson K. Immunogenicity of Danish-SSI 1331 BCG vaccine in the UK: comparison with Glaxo-Evans 1077 BCG vaccine. Vaccine. 2006;24:5726–5733.
    1. Hoft D.F., Kemp E.B., Marinaro M., Cruz O., Kiyono H., McGhee J.R. A double-blind, placebo-controlled study of Mycobacterium-specific human immune responses induced by intradermal Bacille Calmette–Guerin vaccination. J Lab Clin Med. 1999;134:244–252.
    1. Aronson N.E., Santosham M., Comstock G.W., Howard R.S., Moulton L.H., Rhoades E.R. Long-term efficacy of BCG vaccine in American Indians and Alaska Natives: a 60-year follow-up study. JAMA. 2004;291:2086–2091.
    1. Walzl G., Ronacher K., Hanekom W., Scriba T.J., Zumla A. Immunological biomarkers of tuberculosis. Nat Rev Immunol. 2011;11:343–354.
    1. Dietrich J., Doherty M. Interaction of Mycobacterium tuberculosis with the host: consequences for vaccine development. APMIS. 2009;117:440–457.
    1. Soares A.P., Scriba T.J., Joseph S., Harbacheuski R., Murray R.A., Gelderbloem S.J. Bacillus Calmette–Guérin vaccination of human newborns induces T cells with complex cytokine and phenotypic profiles. J Immunol. 2008;180:3569–3577.
    1. Mittrucker H.-W., Steinhoff U., Kohler K., Krause M., Lazar D., Mex P. Poor correlation between BCG vaccination-induced T cell responses and protection against tuberculosis. PNAS. 2007;104(30):12434–12439.
    1. Kagina B.M., Abel B., Scriba T.J., Hughes E.J., Keyser A., Soares A. Specific T cell frequency and cytokine expression profile do not correlate with protection against tuberculosis after bacillus Calmette–Guérin vaccination of newborns. Am J Respir Crit Care Med. 2010;182(8):1073–1079.
    1. Kristensen I., Aaby P., Jensen H. Routine vaccinations and child survival: follow up study in Guinea-Bissau, West Africa. BMJ. 2000;321:1435–1438.
    1. Stensballe L.G., Nantea E., Jensen I.P., Kofoed P.-E., Poulsen A., Jensen H. Acute lower respiratory tract infections and respiratory syncytial virus in infants in Guinea-Bissau: a beneficial effect of BCG vaccination for girls: community based case–control study. Vaccine. 2005;23:1251–1257.
    1. Roth A., Gustafson P., Nhaga A., Djana Q., Poulsen A., Garly M.L. BCG vaccination scar associated with better childhood survival in Guinea-Bissau. Int J Epidemiol. 2005;34:540–547.
    1. Djuardi Y., Sartono E., Wibowo H., Supali T., Yazdanbakhsh M. A longitudinal study of BCG vaccination in early childhood: the development of innate and adaptive immune responses. PLoS One. 2010;5(11):e14066.
    1. Ota M.O.C., Vekemans J., Schlegel-Haueter S.E., Fielding K. Influence of Mycobacterium bovis Bacillus Calmette–Guérin on antibody and cytokine responses to human neonatal vaccination. J Immunol. 2002;168:919–925.
    1. Webb E.L., Mawa P.A., Ndibazza J., Kizito D., Namatovu A., Kyosiimire-Lugemwa J. Effect of single-dose anthelmintic treatment during pregnancy on an infant's response to immunisation and on susceptibility to infectious diseases in infancy: a randomised, double-blind, placebo-controlled trial. Lancet. 2011;377:52–62.
    1. Ndibazza J., Muhangi L., Akishule D., Kiggundu M., Ameke C1 Oweka J. Effects of deworming during pregnancy on maternal and perinatal outcomes in Entebbe, Uganda: a randomized controlled trial. CID. 2010;50:531–540.
    1. Elliott A.M., Kizza M., Quigley M.A., Ndibazza J., Nampijja M., Muhangi L. The impact of helminths on the response to immunization and on the incidence of infection and disease in childhood in Uganda: design of a randomized, double-blind, placebo-controlled, factorial trial of deworming interventions delivered in pregnancy and early childhood. Clin Trials. 2007;4:42–57.
    1. McGuinness D., Bennett S., Riley E. Statistical analysis of highly skewed immune response data. J Immunol Methods. 1997;201(1):99–114.
    1. Aaby P., Jensen H., Rodrigues A., Garly M.-L., Stabell Benn C., Maria Lisse I. Divergent female–male mortality ratios associated with different routine vaccinations among female–male twin pairs. Int J Epidemiol. 2004;33:367–373.
    1. Flanagan K.L., Klein S.L., Skakkebaek N.E., Marriott I., Marchant A., Selin L. Sex differences in the vaccine-specific and non-targeted effects of vaccines. Vaccine. 2011;29:2349–2354.
    1. Elliott A.M., Hurst T.J., Balyeku M.N., Quigley M.A., Kaleebu P., French N. The immune response to Mycobacterium tuberculosis in HIV-infected and uninfected adults in Uganda: application of a whole blood cytokine assay in an epidemiological study. Int J Tuberc Lung Dis. 1999;3(3):239–247.
    1. Keller P.M., Boettger E.C., Sander P. Tuberculosis vaccine strain Mycobacterium bovis BCG Russia is a natural recA mutant. BMC Microbiol. 2008;8:120.
    1. Stefanova T., Chouchkova M., Hinds J., Butcher P.D., Inwald J., Dale J. Genetic composition of Mycobacterium bovis BCG substrain Sofia. J Clin Microbiol. 2003;41(11):5349.
    1. Redford P.S., Murray P.J., O’Garra A. The role of IL-10 in immune regulation during M. tuberculosis infection. Mucosal Immunol. 2011;4(3):261–270.
    1. Hong Y.P., Kim S.J., Lew W.J., Lee E.K., Han Y.C. The seventh nationwide tuberculosis prevalence survey in Korea, 1995. Int J Tuberc Lung Dis. 1998;2(1):27–36.

Source: PubMed

3
Tilaa