Licensed Bacille Calmette-Guérin (BCG) formulations differ markedly in bacterial viability, RNA content and innate immune activation

Asimenia Angelidou, Maria-Giulia Conti, Joann Diray-Arce, Christine S Benn, Frank Shann, Mihai G Netea, Mark Liu, Lakshmi Prasad Potluri, Guzman Sanchez-Schmitz, Robert Husson, Al Ozonoff, Beate Kampmann, Simon Daniël van Haren, Ofer Levy, Asimenia Angelidou, Maria-Giulia Conti, Joann Diray-Arce, Christine S Benn, Frank Shann, Mihai G Netea, Mark Liu, Lakshmi Prasad Potluri, Guzman Sanchez-Schmitz, Robert Husson, Al Ozonoff, Beate Kampmann, Simon Daniël van Haren, Ofer Levy

Abstract

Background: Bacille Calmette-Guérin (BCG), the live attenuated tuberculosis vaccine, is manufactured under different conditions across the globe generating formulations that may differ in clinical efficacy. Innate immune recognition of live BCG contributes to immunogenicity suggesting that differences in BCG viability may contribute to divergent activity of licensed formulations.

Methods: We compared BCG-Denmark (DEN), -Japan (JPN), -India (IND), -Bulgaria (BUL) and -USA in vitro with respect to a) viability as measured by colony-forming units (CFU), mycobacterial membrane integrity, and RNA content, and b) cytokine/chemokine production in newborn cord and adult peripheral blood.

Results: Upon culture, relative growth was BCG-USA > JPN ≫ DEN > BUL = IND. BCG-IND and -BUL demonstrated >1000-fold lower growth than BCG-JPN in 7H9 medium and >10-fold lower growth in commercial Middlebrook 7H11 medium. BCG-IND demonstrated significantly decreased membrane integrity, lower RNA content, and weaker IFN-γ inducing activity in whole blood compared to other BCGs. BCG-induced whole blood cytokines differed significantly by age, vaccine formulation and concentration. BCG-induced cytokine production correlated with CFU, suggesting that mycobacterial viability may contribute to BCG-induced immune responses.

Conclusions: Licensed BCG vaccines differ markedly in their content of viable mycobacteria possibly contributing to formulation-dependent activation of innate and adaptive immunity and distinct protective effects.

Keywords: BCG vaccine formulation; Chemokine; Colony forming units; Cord blood; Cytokine; Viability.

Conflict of interest statement

Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: OL is a named inventor on several vaccine adjuvant formulation patent applications. The other authors do not have a commercial or other association that might pose a conflict of interest.

Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.

Figures

Fig. 1.
Fig. 1.
BCG formulations exhibit differences in mycobacterial cell membrane integrity and RNA content as assessed by flow cytometry. (A) Representative dot-plot and (B) average percentage of live cells as a proportion out of all events captured. N = 5 experiments testing at least 2 different lots of each BCG formulation. (C) Histogram showing an overlay of BCG cytometric analysis using SYTO® RNA SelectTM Green Fluorescent Cell Stain. (D) Bar graph showing the % of RNA + cells per equal number of CFU. Unstained BCG USA was used as control. N = 4. Data are presented as mean +/ SD. Statistical significance denoted by one-way non-repeated measures ANOVA with Fisher’s LSD test. **p < 0.01, ***p < 0.01, ****p < 0.001.
Fig. 2.
Fig. 2.
Licensed BCG formulations differ markedly from one another in viability as measured by colony forming units (CFU) in diverse culture media. Six serial 10-fold dilutions of each BCG formulation were plated in (A) solid enriched 7H9 medium (e7H9) and (B) commercial Middlebrook 7H11 medium (M7H11) in triplicate, and colonies counted weekly for 6 weeks. In both media types, BCG-IND and -BUL demonstrated significantly lower viability (CFU) compared to the other BCG formulations. Moreover, BCG-BUL viability (CFU) was >100-fold lower than anticipated per label. (C) Growth of BCG-IND and -BUL was significantly lower (>100-fold) in e7H9 compared to M7H11 medium. Mean CFU per ampule of each candidate was calculated from the average colonies counted in independent experiments (N = 2–6) and also plotted against the anticipated mycobacterial growth based on the vaccine label. Data are presented as mean +/ SD. Statistical comparisons with ANOVA. *p < 0.05, **p < 0.01, ***p < 0.01.
Fig. 3.
Fig. 3.
(A) BCG-induced whole blood cytokine/chemokine pattern at 18 h differs significantly by age. Heat map depicts cytokines/chemokines after 18 h stimulation with equal CFU concentrations of BCG, calculated from the vaccine inserts. Repeated-measures one-way ANOVA with Sidak’s post hoc test. N = 4–9 for newborn and 8–13 for adult whole blood. (B) In NB, BCG-IND induced the weakest IFN-c responses compared to the other formulations. Box plots display medians and min-max values. Grey stars indicate comparisons against RPMI control (Kruskal-Wallis test). (C) BCG-induced hematopoietic factors and cytokines at concentrations reflecting human equivalent doses differ significantly by BCG formulation. Radar plots representing the BCG effect as a log-fold change over RPMI control. N = 6–10 for newborn and N = 7–11 for adult whole blood. NB, newborn; AD, adult. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Fig. 4.
Fig. 4.
(A) BCG formulations tested at equal CFU concentrations differ in induction of cytokines/chemokines. Radar plots represent the BCG effect as a log-fold change over RPMI control. (B) BCG-DEN induced significantly higher log-fold changes in CCL7, G-CSF, PDGF AB/BB, IL-1α and IL-1β compared to equal calculated CFU of the other BCG formulations. ANOVA with Tukey correction. N = 6–9 for NB; 10–11 for AD. NB, newborn; AD, adult.
Fig. 5.
Fig. 5.
BCG cytokine-inducing activity significantly correlates with BCG viability. (A) Spearman correlations between CFU–cytokines produced in NB after 18 h BCG stimulation with concentrations reflecting human equivalent doses. (B) CFU-cytokine correlations for all cytokines/chemokines that were significantly changed over RPMI. NB = newborn; AD = adult.

References

    1. UNICEF. BCG vaccine: current supply & demand outlook; 2015.
    1. Trunz BB, Fine P, Dye C. Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 2006;367:1173–80.
    1. Basu Roy R, Whittaker E, Seddon JA, Kampmann B. Tuberculosis susceptibility and protection in children. Lancet Infect Dis 2019;19(3):e96–e108. 10.1016/S1473-3099(18)30157-9.
    1. Higgins JP, Soares-Weiser K, Lopez-Lopez JA, Kakourou A, Chaplin K, Christensen H, et al. Association of BCG, DTP, and measles containing vaccines with childhood mortality: systematic review. BMJ 2016;355:i5170.
    1. Biering-Sorensen S, Aaby P, Lund N, Monteiro I, Jensen KJ, Eriksen HB, et al. Early BCG-Denmark and neonatal mortality among infants weighing <2500 g: a randomized controlled trial. Clin Infect Dis 2017;65:1183–90.
    1. Frankel H, Byberg S, Bjerregaard-Andersen M, Martins CL, Aaby P, Benn CS, et al. Different effects of BCG strains – A natural experiment evaluating the impact of the Danish and the Russian BCG strains on morbidity and scar formation in Guinea-Bissau. Vaccine 2016;34:4586–93.
    1. Funch KM, Thysen SM, Rodrigues A, Martins CL, Aaby P, Benn CS, et al. Determinants of BCG scarification among children in rural Guinea-Bissau: A prospective cohort study. Hum Vaccin Immunother 2018;14:2434–42.
    1. Kagina BM, Abel B, Scriba TJ, Hughes EJ, Keyser A, Soares A, et al. Specific T cell frequency and cytokine expression profile do not correlate with protection against tuberculosis after bacillus Calmette-Guerin vaccination of newborns. Am J Respir Crit Care Med 2010;182:1073–9.
    1. Fletcher HA, Snowden MA, Landry B, Rida W, Satti I, Harris SA, et al. T-cell activation is an immune correlate of risk in BCG vaccinated infants. Nat Commun 2016;7:11290.
    1. Anderson EJ, Webb EL, Mawa PA, Kizza M, Lyadda N, Nampijja M, et al. The influence of BCG vaccine strain on mycobacteria-specific and non-specific immune responses in a prospective cohort of infants in Uganda. Vaccine 2012;30:2083–9.
    1. Storgaard L, Rodrigues A, Martins C, Nielsen BU, Ravn H, Benn CS, et al. Development of BCG Scar and Subsequent Morbidity and Mortality in Rural Guinea-Bissau. Clin Infect Dis 2015;61:950–9.
    1. Biering-Sorensen S, Jensen KJ, Aamand SH, Blok B, Andersen A, Monteiro I, et al. Variation of growth in the production of the BCG vaccine and the association with the immune response. An observational study within a randomised trial. Vaccine 2015;33:2056–65.
    1. Cernuschi T, Malvolti S, Nickels E, Friede M. Bacillus Calmette-Guerin (BCG) vaccine: a global assessment of demand and supply balance. Vaccine 2018;36:498–506.
    1. Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PE, et al. Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin Infect Dis 2014;58:470–80.
    1. Curtis N. BCG vaccination and all-cause neonatal mortality. Pediatr Infect Dis J 2019;38:195–7.
    1. Ponte C, Hacker M, Moraes M, Castello-Branco L, Silva F, Antas P. The patterns of in vitro cell-death and inflammatory cytokines induced by distinct BCG vaccine strains are differentially induced in human mononuclear cells. Hum Vaccin Immunother 2018;14:28–35.
    1. Behr MA, Small PM. A historical and molecular phylogeny of BCG strains. Vaccine 1999;17:915–22.
    1. Kroger L, Brander E, Korppi M, Wasz-Hockert O, Backman A, Kroger H, et al. Osteitis after newborn vaccination with three different Bacillus Calmette-Guerin vaccines: twenty-nine years of experience. Pediatr Infect Dis J 1994;13:113–6.
    1. Davids V, Hanekom WA, Mansoor N, Gamieldien H, Gelderbloem SJ, Hawkridge A, et al. The effect of bacille Calmette-Guerin vaccine strain and route of administration on induced immune responses in vaccinated infants. J Infect Dis 2006;193:531–6.
    1. Gheorghiu M, Lagrange PH. Viability, heat stability and immunogenicity of four BCG vaccines prepared from four different BCG strains. Ann Immunol (Paris) 1983;134C:125–47.
    1. Shann F. Editorial commentary: different strains of bacillus calmette-guerin vaccine have very different effects on tuberculosis and on unrelated infections. Clin Infect Dis 2015;61:960–2.
    1. Ritz N, Dutta B, Donath S, Casalaz D, Connell TG, Tebruegge M, et al. The influence of bacille Calmette-Guerin vaccine strain on the immune response against tuberculosis: a randomized trial. Am J Respir Crit Care Med 2012;185:213–22.
    1. Ugolini M, Gerhard J, Burkert S, Jensen KJ, Georg P, Ebner F, et al. Recognition of microbial viability via TLR8 drives TFH cell differentiation and vaccine responses. Nat Immunol 2018;19:386–96.
    1. Dai FY, Wang JF, Gong XL, Bao L. Immunogenicity and protective efficacy of recombinant Bacille Calmette-Guerin strains expressing mycobacterium antigens Ag85A, CFP10, ESAT-6, GM-CSF and IL-12p70. Hum Vaccin Immunother 2017;13:1–8.
    1. Scheid A, Borriello F, Pietrasanta C, Christou H, Diray-Arce J, Pettengill MA, et al. Adjuvant Effect of Bacille Calmette-Guerin on Hepatitis B Vaccine Immunogenicity in the Preterm and Term Newborn. Front Immunol 2018;9:29.
    1. McAuliffe L, Ellis RJ, Miles K, Ayling RD, Nicholas RA. Biofilm formation by mycoplasma species and its role in environmental persistence and survival. Microbiology 2006;152:913–22.
    1. Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park JH, Franchi L, et al. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 2006;440:233–6.
    1. Sander LE, Davis MJ, Boekschoten MV, Amsen D, Dascher CC, Ryffel B, et al. Detection of prokaryotic mRNA signifies microbial viability and promotes immunity. Nature 2011;474:385–9.
    1. Dagg B, Hockley J, Rigsby P, Ho MM. The establishment of sub-strain specific WHO Reference Reagents for BCG vaccine. Vaccine 2014;32:6390–5.
    1. Osborn TW. Changes in BCG strains. Tubercle 1983;64:1–13.
    1. Corner LA, Gormley E, Pfeiffer DU. Primary isolation of Mycobacterium bovis from bovine tissues: conditions for maximising the number of positive cultures. Vet Microbiol 2012;156:162–71.
    1. Messina NL, Germano S, Bonnici R, Lee LY, Daley AJ, Bustamante A, et al. Can colony-forming unit testing be used to extend the shelf life of BCG vaccines?. Tuberculosis (Edinb) 2018;111:188–92.
    1. Gheorghiu M, Lagranderie M, Balazuc AM. Stabilisation of BCG vaccines. Dev Biol Stand 1996;87:251–61.
    1. Favorov M, Ali M, Tursunbayeva A, Aitmagambetova I, Kilgore P, Ismailov S, et al. Comparative tuberculosis (TB) prevention effectiveness in children of Bacillus Calmette-Guerin (BCG) vaccines from different sources, Kazakhstan. PloS one 2012;7:e32567.
    1. Jayaraman K, Adhisivam B, Nallasivan S, Krishnan RG, Kamalarathnam C, Bharathi M, et al. Two randomized trials of the effect of BCG-Russia alone or with oral polio vaccine on neonatal mortality in infants weighing <2000 G in India. Pediatr Infect Dis J 2018;38(2):198–202. 10.1097/INF.0000000000002198.
    1. Yamamoto S, Yamamoto T. Historical review of BCG vaccine in Japan. Jpn J Infect Dis 2007;60:331–6.
    1. Means TK, Wang S, Lien E, Yoshimura A, Golenbock DT, Fenton MJ. Human toll-like receptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol 1999;163:3920–7.
    1. Chambers MA, Marshall BG, Wangoo A, Bune A, Cook HT, Shaw RJ, et al. Differential responses to challenge with live and dead Mycobacterium bovis Bacillus Calmette-Guerin. J Immunol 1997;158:1742–8.
    1. Turner J, Dockrell HM. Stimulation of human peripheral blood mononuclear cells with live Mycobacterium bovis BCG activates cytolytic CD8+ T cells in vitro. Immunology 1996;87:339–42.
    1. Hoffman RJE, Silberstein LE, Heslop H, Weitz J, Anastasi J. Hematology: Diagnosis and Treatment. 6 ed. Elsevier; 2013. p. 311.
    1. White GP, Watt PM, Holt BJ, Holt PG. Differential patterns of methylation of the IFN-gamma promoter at CpG and non-CpG sites underlie differences in IFN-gamma gene expression between human neonatal and adult CD45RO- T cells. J Immunol 2002;168:2820–7.
    1. van den Biggelaar AH, Prescott SL, Roponen M, Nadal-Sims MA, Devitt CJ, Phuanukoonnon S, et al. Neonatal innate cytokine responses to BCG controlling T-cell development vary between populations. J Allergy Clin Immunol 2009;124:544–50. 50 e1–2.
    1. Sanchez-Schmitz G, Stevens CR, Bettencourt IA, Flynn PJ, Schmitz-Abe K, Metser G, et al. Microphysiologic Human Tissue Constructs Reproduce Autologous Age-Specific BCG and HBV Primary Immunization in vitro. Front Immunol 2018;9:2634.
    1. Jensen KJ, Larsen N, Biering-Sorensen S, Andersen A, Eriksen HB, Monteiro I, et al. Heterologous immunological effects of early BCG vaccination in low-birth-weight infants in Guinea-Bissau: a randomized-controlled trial. J Infect Dis 2015;211:956–67.
    1. Wu B, Huang C, Garcia L, Ponce de Leon A, Osornio JS, Bobadilla-del-Valle M, et al. Unique gene expression profiles in infants vaccinated with different strains of Mycobacterium bovis bacille Calmette-Guerin. Infect Immun 2007;75:3658–64.
    1. Moorlag S, Roring RJ, Joosten LAB, Netea MG. The role of the interleukin-1 family in trained immunity. Immunol Rev 2018;281:28–39.
    1. de Castro MJ, Pardo-Seco J, Martinon-Torres F. Nonspecific (Heterologous) Protection of Neonatal BCG Vaccination Against Hospitalization Due to Respiratory Infection and Sepsis. Clin Infect Dis 2015;60:1611–9.
    1. Vouret-Craviari V, Cenzuales S, Poli G, Mantovani A. Expression of monocyte chemotactic protein-3 in human monocytes exposed to the mycobacterial cell wall component lipoarabinomannan. Cytokine 1997;9:992–8.
    1. Benmerzoug S, Marinho FV, Rose S, Mackowiak C, Gosset D, Sedda D, et al. GM-CSF targeted immunomodulation affects host response to M. tuberculosis infection. Sci Rep 2018;8:8652.
    1. Denis M, Ghadirian E. Granulocyte-macrophage colony-stimulating factor restricts growth of tubercle bacilli in human macrophages. Immunol Lett 1990;24:203–6.
    1. Dowling DJ, Scott EA, Scheid A, Bergelson I, Joshi S, Pietrasanta C, et al. Toll-like receptor 8 agonist nanoparticles mimic immunomodulating effects of the live BCG vaccine and enhance neonatal innate and adaptive immune responses. J Allergy Clin Immunol 2017;140:1339–50.
    1. Berendsen MLT, Øland CB, Bles P, Jensen AKG, Kofoed PE, Whittle H, et al. Maternal Priming: Bacillus Calmette-Guérin (BCG) Vaccine Scarring in Mothers Enhances the Survival of Their Child With a BCG Vaccine Scar. J Pediatric Infect Dis Soc 2019. 10.1093/jpids/piy142 [Epub ahead of print].
    1. Schaltz-Buchholzer F, Bjerregaard-Andersen M, Bjerregård Øland C, Golding C, Brenno Stjernholm E, Monteiro I, et al. Early vaccination with BCG-Denmark or BCG-Japan versus BCG-Russia to healthy newborns in Guinea-Bissau: A randomized controlled trial. Clin Infect Dis 2019. 10.1093/cid/ciz1080. In press.

Source: PubMed

3
Se inscrever