Reactogenicity to major tuberculosis antigens absent in BCG is linked to improved protection against Mycobacterium tuberculosis

Nacho Aguilo, Jesus Gonzalo-Asensio, Samuel Alvarez-Arguedas, Dessislava Marinova, Ana Belen Gomez, Santiago Uranga, Ralf Spallek, Mahavir Singh, Regine Audran, François Spertini, Carlos Martin, Nacho Aguilo, Jesus Gonzalo-Asensio, Samuel Alvarez-Arguedas, Dessislava Marinova, Ana Belen Gomez, Santiago Uranga, Ralf Spallek, Mahavir Singh, Regine Audran, François Spertini, Carlos Martin

Abstract

MTBVAC is a live-attenuated Mycobacterium tuberculosis vaccine, currently under clinical development, that contains the major antigens ESAT6 and CFP10. These antigens are absent from the current tuberculosis vaccine, BCG. Here we compare the protection induced by BCG and MTBVAC in several mouse strains that naturally express different MHC haplotypes differentially recognizing ESAT6 and CFP10. MTBVAC induces improved protection in C3H mice, the only of the three tested strains reactive to both ESAT6 and CFP10. Deletion of both antigens in MTBVAC reduces its efficacy to BCG levels, supporting a link between greater efficacy and CFP10- and ESAT6-specific reactogenicity. In addition, MTBVAC (but not BCG) triggers a specific response in human vaccinees against ESAT6 and CFP10. Our results warrant further exploration of this response as potential biomarker of protection in MTBVAC clinical trials.

Conflict of interest statement

C.M. and J.G.-A. are co-inventors in a patent application entitled “Tuberculosis vaccine” filled by the University of Zaragoza (application number: PCT/ES 2007/070051). The remaining authors declare no competing financial interests.

Figures

Figure 1. Expression and secretion of ESAT6…
Figure 1. Expression and secretion of ESAT6 and CFP10 by MTBVAC.
(a) Normalized expression (using sigA as housekeeping gene) of fbpB, esat6 and cfp10 genes in log-phase broth-cultured BCG and MTBVAC. Data in the graph are represented as the relative quantity (RQ) using MTBVAC as comparator. Results are the average from triplicate experiments. Data are mean±s.d. (b,c) Immunoblot analysis of GroEL2, Ag85A, ESAT6 and CFP10 in BCG, MTBVAC and M. tuberculosis protein extracts. (b) Intracellular (left) and supernatant (right) fractions of BCG and MTBVAC were assessed. (c) Comparison between M. tuberculosis and MTBVAC-secreted fractions. A BCG cell lysate sample was used as positive control for GroEL2 detection. Four independent protein extractions of each strain were included in the analysis. Full blots are shown in Supplementary Fig. 7a–c.
Figure 2. Improved protection of MTBVAC compared…
Figure 2. Improved protection of MTBVAC compared to BCG is dependent on the host genetics.
(a,b,c) Antigen-specific IFNγ production following stimulation with PPD (5 μg ml−1), ESAT6 (2 μg ml−1), CFP10 (2 μg ml−1) and Ag85B (2 μg ml−1) during 48 h of splenocytes from mock-, BCG- and MTBVAC-vaccinated C57BL/6, BALB/c and C3H/HeNRj mice. (di) Lung (d,e,f) and spleen (g,h,i) bacterial load 4 weeks post low-dose H37Rv intranasal challenge. C57BL/6 (d,g), BALB/c (e,h) and C3H/HeNRj (f,i) were vaccinated with BCG, MTBVAC or unvaccinated eight weeks before challenge. (a,b,c) Data are representative from one of two independent experiments (n=5 mice per group per experiment). (di) Data in the graphs represent a pool of two independent experiments (n=12 mice per group). All data are mean±s.e.m. (a,b,c) NS, non-significant; ***P<0.001 by unpaired t-student test. (di) NS, non-significant; *P<0.05; **P<0.01; ***P<0.001 by one-way ANOVA and Bonferroni post-test.
Figure 3. MTBVAC-induced immune response specific to…
Figure 3. MTBVAC-induced immune response specific to ESAT6 and CFP10 is protective.
(a) Immunoblot analysis of GroEL2, ESAT6 and CFP10 in MTBVAC and MTBVACΔE6C10 lysate samples. Full blot is shown in Supplementary Fig. 7d. (b,c) Antigen-specific IFNγ production following stimulation with PPD (5 μg ml−1), ESAT6 (2 μg ml−1), CFP10 (2 μg ml−1) and Ag85B (2 μg ml−1) during 48 h of splenocytes from mock-, MTBVAC- and MTBVACΔE6C10-vaccinated C57BL/6 (left) and C3H/HeNRj (right) mice. (di) Lung (d,e,f) and spleen (g,h,i) bacterial load 4 weeks post low-dose H37Rv intranasal challenge. C57BL/6 (d,g), BALB/c (e,h) and C3H/HeNRj (f,i) were vaccinated with BCG, MTBVAC, MTBVACΔE6C10 or unvaccinated 8 weeks before challenge. (b,c) Data are representative from one of two independent experiments (n=5 mice per group per experiment). (d,e,g,h) Data are derived from n=6. (f,i) Data represent a pool of two independent experiments (n=12 mice per group). All data are mean±s.e.m. (b,c) NS, non-significant; **P<0.01; ***P<0.001 by unpaired t-student test. (di) NS, non-significant; *P<0.05; **P<0.01; ***P<0.001 by one-way ANOVA and Bonferroni post-test.
Figure 4. Ag85B-specific immunogenicity induced by MTBVAC…
Figure 4. Ag85B-specific immunogenicity induced by MTBVAC is not protective.
(a,b) Antigen-specific IFNγ production following stimulation with PPD (5 μg ml), ESAT6 (2 μg ml−1), CFP10 (2 μg ml−1) and Ag85B (2 μg ml−1) during 48 h of splenocytes from mock-, MTBVAC- and MTBVACΔAg85B-vaccinated C57BL/6 (left) and C3H/HeNRj (right) mice. (ce) Lung bacterial load 4 weeks post low-dose H37Rv intranasal challenge. C57BL/6 (c), BALB/c (d) and C3H/HeNRj (e) were vaccinated with BCG, MTBVAC, MTBVACΔAg85B or unvaccinated 8 weeks before challenge. (a,b) Data are representative from one of two independent experiments (n=5 mice per group per experiment). (ce) Data are derived from n=6. All data are mean±s.e.m. (a,b) NS, non-significant; **P<0.01; ***P<0.001 by unpaired t-student test. (ce) *P<0.05; **P<0.01; ***P<0.001 by one-way ANOVA and Bonferroni post-test.
Figure 5. Enriched in vivo expression of…
Figure 5. Enriched in vivo expression of genes from ESX-1 secretion system.
(a,b) Expression of esat6, cfp10, espA, espC, fbpA and fbpB genes (normalized with 16s gene expression) from H37Rv isolated from lungs from C3H/HeNRj (a) and C57BL/6 (b) 4 weeks after high-dose (103 CFU) intranasal challenge, in comparison to expression obtained under 7H9-culture in vitro conditions. Normalized results are represented for each gene and experimental condition as the fold-change induction in comparison to fbpB expression level. In vivo data are derived from n=4 mice (a) and n=6 mice (b). In vitro data represent a pool of four independent RNA extractions. All data are mean±s.e.m. *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001 by unpaired t-student test.
Figure 6. Vaccination with MTBVAC induces a…
Figure 6. Vaccination with MTBVAC induces a CFP10-specific immune response in humans.
Specific IFNγ ELISPOT results from the first-in-human MTBVAC clinical trial are shown for CFP10 (a,b) and ESAT6 (c,d), comparing for each volunteer the number of spots pre- and post-BCG (b,d) and MTBVAC (a,c) vaccination with 5 × 105 CFU. Wilcoxon matched-pairs signed rank test was used to compare pre- and post-vaccination status for each vaccine and antigen. P value is indicated for MTBVAC vaccinees.

References

    1. Marinova D., Gonzalo-Asensio J., Aguilo N. & Martin C. Recent developments in tuberculosis vaccines. Expert Rev. Vaccines 12, 1431–1448 (2013).
    1. WHO. Global tuberculosis report 2016. (2016).
    1. Fine P. E. Variation in protection by BCG: implications of and for heterologous immunity. Lancet 346, 1339–1345 (1995).
    1. Brosch R. et al.. Genome plasticity of BCG and impact on vaccine efficacy. Proc. Natl Acad. Sci. USA 104, 5596–5601 (2007).
    1. Pym A. S., Brodin P., Brosch R., Huerre M. & Cole S. T. Loss of RD1 contributed to the attenuation of the live tuberculosis vaccines Mycobacterium bovis BCG and Mycobacterium microti. Mol. Microbiol. 46, 709–717 (2002).
    1. Aagaard C. et al.. A multistage tuberculosis vaccine that confers efficient protection before and after exposure. Nat. Med. 17, 189–194 (2011).
    1. Groschel M. I. et al.. Recombinant BCG expressing ESX-1 of Mycobacterium marinum combines low virulence with cytosolic immune signaling and improved TB protection. Cell Rep. 18, 2752–2765 (2017).
    1. Pym A. S. et al.. Recombinant BCG exporting ESAT-6 confers enhanced protection against tuberculosis. Nat. Med. 9, 533–539 (2003).
    1. Arbues A. et al.. Construction, characterization and preclinical evaluation of MTBVAC, the first live-attenuated M. tuberculosis-based vaccine to enter clinical trials. Vaccine 31, 4867–4873 (2013).
    1. Stucki D. et al.. Mycobacterium tuberculosis lineage 4 comprises globally distributed and geographically restricted sublineages. Nat. Genet. 48, 1535–1543 (2016).
    1. Kamath A. T. et al.. New live mycobacterial vaccines: the Geneva consensus on essential steps towards clinical development. Vaccine 23, 3753–3761 (2005).
    1. Frigui W. et al.. Control of M. tuberculosis ESAT-6 secretion and specific T cell recognition by PhoP. PLoS Pathog. 4, e33 (2008).
    1. Camacho L. R., Ensergueix D., Perez E., Gicquel B. & Guilhot C. Identification of a virulence gene cluster of Mycobacterium tuberculosis by signature-tagged transposon mutagenesis. Mol. Microbiol. 34, 257–267 (1999).
    1. Martin C. et al.. The live Mycobacterium tuberculosis phoP mutant strain is more attenuated than BCG and confers protective immunity against tuberculosis in mice and guinea pigs. Vaccine 24, 3408–3419 (2006).
    1. Verreck F. A. et al.. MVA.85A boosting of BCG and an attenuated, phoP deficient M. tuberculosis vaccine both show protective efficacy against tuberculosis in rhesus macaques. PLoS ONE 4, e5264 (2009).
    1. Aguilo N. et al.. MTBVAC vaccine is safe, immunogenic and confers protective efficacy against Mycobacterium tuberculosis in newborn mice. Tuberculosis 96, 71–74 (2016).
    1. Spertini F. et al.. Safety of human immunisation with a live-attenuated Mycobacterium tuberculosis vaccine: a randomised, double-blind, controlled phase I trial. Lancet Respir. Med. 3, 953–962 (2015).
    1. Renshaw P. S. et al.. Conclusive evidence that the major T-cell antigens of the Mycobacterium tuberculosis complex ESAT-6 and CFP-10 form a tight, 1:1 complex and characterization of the structural properties of ESAT-6, CFP-10, and the ESAT-6*CFP-10 complex. Implications for pathogenesis and virulence. J. Biol. Chem. 277, 21598–21603 (2002).
    1. Kamath A. B. et al.. Cytolytic CD8+ T cells recognizing CFP10 are recruited to the lung after Mycobacterium tuberculosis infection. J. Exp. Med. 200, 1479–1489 (2004).
    1. Copin R., Coscolla M., Efstathiadis E., Gagneux S. & Ernst J. D. Impact of in vitro evolution on antigenic diversity of Mycobacterium bovis bacillus Calmette-Guerin (BCG). Vaccine 32, 5998–6004 (2014).
    1. Bold T. D., Banaei N., Wolf A. J. & Ernst J. D. Suboptimal activation of antigen-specific CD4+ effector cells enables persistence of M. tuberculosis in vivo. PLoS Pathog. 7, e1002063 (2011).
    1. Fortune S. M. et al.. Mutually dependent secretion of proteins required for mycobacterial virulence. Proc. Natl Acad. Sci. USA 102, 10676–10681 (2005).
    1. Lou Y., Rybniker J., Sala C. & Cole S. T. EspC forms a filamentous structure in the cell envelope of Mycobacterium tuberculosis and impacts ESX-1 secretion. Mol. Microbiol. 103, 26–38 (2017).
    1. MacGurn J. A., Raghavan S., Stanley S. A. & Cox J. S. A non-RD1 gene cluster is required for Snm secretion in Mycobacterium tuberculosis. Mol. Microbiol. 57, 1653–1663 (2005).
    1. Rogerson B. J. et al.. Expression levels of Mycobacterium tuberculosis antigen-encoding genes versus production levels of antigen-specific T cells during stationary level lung infection in mice. Immunology 118, 195–201 (2006).
    1. Mehra N. K. & Kaur G. MHC-based vaccination approaches: progress and perspectives. Expert Rev. Mol. Med. 5, 1–17 (2003).
    1. Zhang W. et al.. Genome sequencing and analysis of BCG vaccine strains. PLoS ONE 8, e71243 (2013).
    1. Simeone R. et al.. Phagosomal rupture by Mycobacterium tuberculosis results in toxicity and host cell death. PLoS Pathog. 8, e1002507 (2012).
    1. Horwitz M. A. & Harth G. A new vaccine against tuberculosis affords greater survival after challenge than the current vaccine in the guinea pig model of pulmonary tuberculosis. Infect. Immun. 71, 1672–1679 (2003).
    1. Lindestam Arlehamn C. S. et al.. A quantitative analysis of complexity of human pathogen-specific CD4 T cell responses in healthy M. tuberculosis infected South Africans. PLoS Pathog. 12, e1005760 (2016).
    1. Gonzalo-Asensio J. et al.. Evolutionary history of tuberculosis shaped by conserved mutations in the PhoPR virulence regulator. Proc. Natl Acad. Sci. USA 111, 11491–11496 (2014).
    1. Supply P. et al.. Genomic analysis of smooth tubercle bacilli provides insights into ancestry and pathoadaptation of Mycobacterium tuberculosis. Nat. Genet. 45, 172–179 (2013).
    1. Solans L. et al.. Hyper-attenuated MTBVAC erp mutant protects against tuberculosis in mice. Vaccine 32, 5192–5197 (2014).
    1. Grotzke J. E., Siler A. C., Lewinsohn D. A. & Lewinsohn D. M. Secreted immunodominant Mycobacterium tuberculosis antigens are processed by the cytosolic pathway. J. Immunol. 185, 4336–4343 (2010).
    1. Gao L. Y. et al.. A mycobacterial virulence gene cluster extending RD1 is required for cytolysis, bacterial spreading and ESAT-6 secretion. Mol. Microbiol. 53, 1677–1693 (2004).
    1. Pang X. et al.. MprAB regulates the espA operon in Mycobacterium tuberculosis and modulates ESX-1 function and host cytokine response. J. Bacteriol. 195, 66–75 (2013).
    1. Solans L. et al.. A specific polymorphism in Mycobacterium tuberculosis H37Rv causes differential ESAT-6 expression and identifies WhiB6 as a novel ESX-1 component. Infect. Immun. 82, 3446–3456 (2014).
    1. Solans L. et al.. The PhoP-dependent ncRNA Mcr7 modulates the TAT secretion system in Mycobacterium tuberculosis. PLoS Pathog. 10, e1004183 (2014).
    1. Smith C. M. et al.. Tuberculosis susceptibility and vaccine protection are independently controlled by host genotype. MBio 7,, e01516-16 (2016).
    1. Millington K. A. et al.. Rv3615c is a highly immunodominant RD1 (region of difference 1)-dependent secreted antigen specific for Mycobacterium tuberculosis infection. Proc. Natl Acad. Sci. USA 108, 5730–5735 (2011).
    1. Andrews J. R. et al.. Serial QuantiFERON testing and tuberculosis disease risk among young children: an observational cohort study. Lancet Respir. Med. 5, 282–290 (2017).
    1. Kaufmann S. H., Weiner J. & von Reyn C. F. Novel approaches to tuberculosis vaccine development. Int. J. Infect. Dis. 56, 263–267 (2017).
    1. Andrews J. R. et al.. Risk of progression to active tuberculosis following reinfection with Mycobacterium tuberculosis. Clin. Infect. Dis. 54, 784–791 (2012).
    1. Brosch R. et al.. Use of a Mycobacterium tuberculosis H37Rv bacterial artificial chromosome library for genome mapping, sequencing, and comparative genomics. Infect. Immun. 66, 2221–2229 (1998).
    1. Datsenko K. A. & Wanner B. L. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc. Natl Acad. Sci. USA 97, 6640–6645 (2000).
    1. van Kessel J. C. & Hatfull G. F. Recombineering in Mycobacterium tuberculosis. Nat. Methods 4, 147–152 (2007).
    1. Zhao M. et al.. Systems infection biology: a compartmentalized immune network of pig spleen challenged with Haemophilus parasuis. BMC Genomics 14, 46 (2013).

Source: PubMed

3
Subscribe