Tools for Assessing the Protective Efficacy of TB Vaccines in Humans: in vitro Mycobacterial Growth Inhibition Predicts Outcome of in vivo Mycobacterial Infection

Rachel Tanner, Iman Satti, Stephanie A Harris, Matthew K O'Shea, Deniz Cizmeci, Daniel O'Connor, Agnieszka Chomka, Magali Matsumiya, Rachel Wittenberg, Angela M Minassian, Joel Meyer, Helen A Fletcher, Helen McShane, Rachel Tanner, Iman Satti, Stephanie A Harris, Matthew K O'Shea, Deniz Cizmeci, Daniel O'Connor, Agnieszka Chomka, Magali Matsumiya, Rachel Wittenberg, Angela M Minassian, Joel Meyer, Helen A Fletcher, Helen McShane

Abstract

Tuberculosis (TB) remains a leading global cause of morbidity and mortality and an effective new vaccine is urgently needed. A major barrier to the rational development of novel TB vaccines is the lack of a validated immune correlate or biomarker of protection. Mycobacterial Growth Inhibition Assays (MGIAs) provide an unbiased measure of ability to control mycobacterial growth in vitro, and may represent a functional correlate of protection. However, the biological relevance of any potential correlate can only be assessed by determining the association with in vivo protection from either a controlled mycobacterial infection or natural development of TB disease. Our data demonstrate that the direct MGIA using peripheral blood mononuclear cells (PBMC) is measuring a biologically relevant response that correlates with protection from in vivo human BCG infection across two independent cohorts. This is the first report of an MGIA correlating with in vivo protection in the species-of-interest, humans, and furthermore on a per-individual as well as per-group basis. Control of mycobacterial growth in the MGIA is associated with a range of immune parameters measured post-BCG infection in vivo including the IFN-γ ELISpot response, frequency of PPD-specific IFN-γ or TNF-α producing CD4+ T cells and frequency of specific sub-populations of polyfunctional CD4+ T cells. Distinct transcriptomic profiles are associated with good vs. poor mycobacterial control in the MGIA, with good controllers showing enrichment for gene sets associated with antigen processing/presentation and the IL-23 pathway, and poor controllers showing enrichment for hypoxia-related pathways. This study represents an important step toward biologically validating the direct PBMC MGIA for use in TB vaccine development and furthermore demonstrates the utility of this assay in determining relevant immune mechanisms and pathways of protection.

Trial registration: ClinicalTrials.gov NCT01194180.

Keywords: BCG; MGIA; Tuberculosis; mycobacterial growth inhibition assay; vaccine.

Copyright © 2020 Tanner, Satti, Harris, O'Shea, Cizmeci, O'Connor, Chomka, Matsumiya, Wittenberg, Minassian, Meyer, Fletcher and McShane.

Figures

Figure 1
Figure 1
Mycobacterial growth in the MGIA is inhibited in BCG-vaccinated compared with naïve volunteers and correlates with BCG recovered from biopsy following in vivo BCG infection (Study 1). Samples were taken from Study 1. n = 24 healthy human volunteers, half of whom were BCG-naïve and half of whom were historically BCG-vaccinated, were infected with intradermal BCG. The direct PBMC MGIA was conducted on cells and serum taken at the day of infection and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined (A). As previously reported, the BCG load was quantified from skin biopsy specimens at the site of challenge 2 weeks later using PCR (B). The association between mycobacterial growth in the MGIA and BCG recovered from biopsies in the BCG vaccinated group by both PCR (C) and culture CFU (D) was determined. Bars represent the median values with interquartile range (IQR). For (A,B) a Mann-Whitney U test was performed, where *p < 0.05 and ****p < 0.0001. For (C,D) a Spearman's correlation was performed. MGIA growth ratio = log10(CFU of sample/CFU of control).
Figure 2
Figure 2
Mycobacterial growth in the MGIA is inhibited in BCG-vaccinated compared with naïve volunteers and correlates with BCG recovered from biopsy following in vivo BCG infection (Study 2). Samples were taken from Study 2. n = 48 healthy human volunteers were assigned to groups A and B (BCG-naïve) or groups C and D (historically BCG vaccinated). Groups B and D received the candidate TB vaccine MVA85A. All volunteers were then infected with intradermal BCG. The direct PBMC MGIA was conducted on cells and plasma taken at the day of infection and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined (A). As previously reported, the BCG load was quantified from skin biopsy specimens at the site of challenge 2 weeks later using PCR (B). The association between mycobacterial growth in the MGIA and BCG recovered from biopsies was determined for all groups combined (C) and for the BCG-vaccinated group (group C) only (D). Bars represent the median values with IQR. For (A) a Kruskal Wallis test with Dunn's multiple comparisons was performed and for (B) a one-way ANOVA with Tukey's multiple comparisons test was performed, where *p < 0.05, **p < 0.005, and ***p < 0.0005. For (C,D) a Spearman's correlation was performed. MGIA growth ratio = log10(CFU of sample/CFU of control).
Figure 3
Figure 3
Frequency of cytokine-producing specific CD4+ T cells is associated with improved control of mycobacterial growth in the MGIA. Samples were taken from Study 2. n = 48 healthy human volunteers were assigned to groups A and B (BCG-naïve) or groups C and D (historically BCG vaccinated). Groups B and D received the candidate TB vaccine MVA85A. All volunteers were then infected with intradermal BCG, and frequency of PPD-specific IFN-γ producing CD4+ T cells were measured at 2 weeks post-infection using whole blood ICS (A). The direct PBMC MGIA was conducted on cells and serum taken at the day of challenge and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined. The association between mycobacterial growth in the MGIA and the frequency of PPD-specific IFN-γ producing CD4+ T cells was determined for all groups combined (B) and for the BCG-vaccinated group (group C) only (C). Bars represent the median values with IQR. For (A), a one-way ANOVA with Tukey's multiple comparisons was performed where *p < 0.05, ***p < 0.0005, and ****p < 0.0001. For (B,C) a Spearman's correlation was performed. MGIA growth ratio = log10(CFU of sample/CFU of control).
Figure 4
Figure 4
Frequency of sub-populations of cytokine-producing specific CD4+ T cells is associated with improved control of mycobacterial growth in the MGIA. Samples were taken from Study 2. n = 48 healthy human volunteers were assigned to groups A and B (BCG-naïve) or groups C and D (historically BCG vaccinated). Groups B and D received the candidate TB vaccine MVA85A. All volunteers were then infected with intradermal BCG, and the frequency of PPD-specific CD4+ T cells producing each of the possible expression profile permutations of the cytokines IFN-γ, TNF-α, IL-2, and IL-17 was determined by flow cytometry using whole blood samples taken at 2 weeks post-infection. The direct PBMC MGIA was conducted on cells and plasma taken at the day of challenge and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined. The association between mycobacterial growth in the MGIA at day of challenge and frequency of CD4+ T cells producing each profile was determined for all groups combined (A) or for the BCG-vaccinated group (group C) only (B) using Spearman's rank correlation, where light blue shading indicates a p < 0.05 and darker blue shading indicates a p < 0.01. Gray bars indicate the mean frequency of each sub-population of cells with the SEM.
Figure 5
Figure 5
Specific IgG ELISA responses are induced by BCG vaccination and are associated with improved control of mycobacterial growth in the MGIA. Samples were taken from Study 2. n = 48 healthy human volunteers were assigned to groups A and B (BCG-naïve) or groups C and D (historically BCG vaccinated). Groups B and D received the candidate TB vaccine MVA85A. All volunteers were then infected with intradermal BCG, and BCG-specific IgG responses (A) were measured at 2 weeks post-infection. The direct PBMC MGIA was conducted on cells and plasma taken at the day of challenge and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined. The association between mycobacterial growth in the MGIA and the BCG-specific IgG ELISA response was determined for all groups combined (B) and for the BCG-vaccinated group (group C) only (C). Bars represent the median values with IQR. For (A) a one-way ANOVA with Tukey's multiple comparisons was performed where **p < 0.005 and ***p < 0.0005. For (B,C) a Spearman's correlation was performed. MGIA growth ratio = log10(CFU of sample/CFU of control).
Figure 6
Figure 6
Distinct transcriptomic profiles are associated with good vs. poor mycobacterial control in the MGIA. Samples were taken from 10 volunteers assigned to group C (historically BCG vaccinated) in Study 2. Volunteers were infected with intradermal BCG and gene expression microarray analysis was conducted on PBMC taken at 2 weeks post-infection. The direct PBMC MGIA was conducted on cells and plasma taken at the day of challenge and the ratio of mycobacterial growth at the end of the 96 h culture relative to an inoculum control was determined. Volunteers were classified as ‘good' or “poor” controllers defined as having MGIA mycobacterial growth values below or above the group median, respectively. Gene expression was analyzed using a 2 × 2 factorial design with the interaction term (BCG stimulated − unstimulated PBMC for good MGIA controllers) vs. (BCG stimulated − unstimulated PBMC for poor MGIA controllers). Differentially expressed genes were defined as those with a p < 0.01 and log2 FC > 0.5 and are presented as a heatmap (A). GSEA analysis was conducted to identify biological pathways that were enriched in “good” vs. “poor” controllers and vice versa using an FDR cut-off of < 0.05. Representative top GSEA pathways for each category are shown from the top 50 gene sets ranked by normalized enrichment score (nES) (B).

References

    1. WHO Global Tuberculosis Report (2019).
    1. Young D, Dye C. The Development and impact of tuberculosis vaccines. Cell. (2006) 124:683–7. 10.1016/j.cell.2006.02.013
    1. Rodrigues LC, Diwan VK, Wheeler JG. Protective effect of BCG against tuberculous meningitis and miliary tuberculosis: a meta-analysis. Int J Epidemiol. (1993) 22: 1154–8. 10.1093/ije/22.6.1154
    1. Fine PE. Variation in protection by BCG: implications of and for heterologous immunity. Lancet. (1995) 346:1339–45. 10.1016/S0140-6736(95)92348-9
    1. Plotkin SA, Gilbert PB. Nomenclature for immune correlates of protection after vaccination. Clin Infect Dis. (2012) 54:1615–7. 10.1093/cid/cis238
    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-Guérin vaccination of newborns. Am J Respir Crit Care Med. (2010) 182:1073–9. 10.1164/rccm.201003-0334OC
    1. Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, 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) 381:1021–8. 10.1016/S0140-6736(13)60177-4
    1. Tanner R, O'Shea MK, Fletcher HA, McShane H. In vitro mycobacterial growth inhibition assays: A tool for the assessment of protective immunity and evaluation of tuberculosis vaccine efficacy. Vaccine. (2016) 34:4656–65. 10.1016/j.vaccine.2016.07.058
    1. Brennan MJ, Tanner R, Morris S, Scriba TJ, Achkar JM, Zelmer A, et al. . The cross-species mycobacterial growth inhibition assay (MGIA) project, 2010–2014. Clin Vaccine Immunol. (2017) 24:e00142–17. 10.1128/CVI.00142-17
    1. Tanner R, Smith SG, van Meijgaarden KE, Giannoni F, Wilkie M, Gabriele L, et al. . Optimisation, harmonisation and standardisation of the direct mycobacterial growth inhibition assay using cryopreserved human peripheral blood mononuclear cells. J Immunol Methods. (2019) 469:1–10. 10.1016/j.jim.2019.01.006
    1. Wallis RS, Palaci M, Vinhas S, Hise AG, Ribeiro FC, Landen K, et al. . A whole blood bactericidal assay for tuberculosis. J Infect Dis. (2001) 183:1300–3. 10.1086/319679
    1. Fletcher HA, Tanner R, Wallis RS, Meyer J, Manjaly ZR, Harris S, et al. Inhibition of mycobacterial growth in vitro following primary but not secondary vaccination with Mycobacterium bovis BCG. Clin Vaccine Immunol. (2013) 20:1683–9. 10.1128/CVI.00427-13
    1. Smith SG, Zelmer A, Blitz R, Fletcher HA, Dockrell HM. Polyfunctional CD4 T-cells correlate with in vitro mycobacterial growth inhibition following Mycobacterium bovis BCG-vaccination of infants. Vaccine. (2016) 34:5298–305. 10.1016/j.vaccine.2016.09.002
    1. Joosten SA, Meijgaarden KEV, Arend SM, Prins C, Oftung F, Korsvold GE, et al. . Mycobacterial growth inhibition is associated with trained innate immunity. J Clin Invest. (2018) 128: 1837–51. 10.1172/JCI97508
    1. Duncan CJ, Hill AV, Ellis RD. Can growth inhibition assays (GIA) predict blood-stage malaria vaccine efficacy? Hum Vaccin Immunother. (2012) 8:706–14. 10.4161/hv.19712
    1. Huang KY, Li CK, Clutterbuck E, Chui C, Wilkinson T, Gilbert A, et al. . Virus-specific antibody secreting cell, memory B-cell, and sero-antibody responses in the human influenza challenge model. J Infect Dis. (2014) 209:1354–61. 10.1093/infdis/jit650
    1. McArthur MA, Fresnay S, Magder LS, Darton TC, Jones C, Waddington CS, et al. . Activation of Salmonella Typhi-specific regulatory T cells in typhoid disease in a wild-type S. Typhi challenge model. PLoS Pathog. (2015) 11:e1004914. 10.1371/journal.ppat.1004914
    1. Dunachie S, Hill AV, Fletcher HA. Profiling the host response to malaria vaccination and malaria challenge. Vaccine. (2015) 33:5316–20. 10.1016/j.vaccine.2015.07.107
    1. Shirley DA, McArthur MA. The utility of human challenge studies in vaccine development: lessons learned from cholera. Vaccine. (2011) 2011:3–13. 10.2147/VDT.S23634
    1. Harris SA, Meyer J, Satti I, Marsay L, Poulton ID, Tanner R, et al. . Evaluation of a human BCG challenge model to assess antimycobacterial immunity induced by BCG and a candidate tuberculosis vaccine, MVA85A, alone and in combination. J Infect Dis. (2014) 209:1259–68. 10.1093/infdis/jit647
    1. Minassian AM, Satti I, Poulton ID, Meyer J, Hill AV, McShane H. A human challenge model for Mycobacterium tuberculosis using Mycobacterium bovis bacille Calmette-Guerin. J Infect Dis. (2012) 205:1035–42. 10.1093/infdis/jis012
    1. Minhinnick A, Harris S, Wilkie M, Peter J, Stockdale L, Manjaly-Thomas ZR, et al. . Optimization of a Human Bacille Calmette-Guérin Challenge Model: a Tool to Evaluate Antimycobacterial Immunity. J Infect Dis. (2016) 213:824–30. 10.1093/infdis/jiv482
    1. Matsumiya M, Satti I, Chomka A, Harris SA, Stockdale L, Meyer J, et al. . Gene expression and cytokine profile correlate with mycobacterial growth in a human BCG challenge model. J Infect Dis. (2015) 211:1499–509. 10.1093/infdis/jiu615
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. . Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. (2005) 102:15545–50. 10.1073/pnas.0506580102
    1. Marsay L, Matsumiya M, Tanner R, Poyntz H, Griffiths KL, Stylianou E, et al. . Mycobacterial growth inhibition in murine splenocytes as a surrogate for protection against Mycobacterium tuberculosis (M. tb). Tuberculosis. (2013) 93:551–7. 10.1016/j.tube.2013.04.007
    1. Jensen C, Lindebo Holm L, Svensson E, Aagaard C, Ruhwald M. Optimisation of a murine splenocyte mycobacterial growth inhibition assay using virulent Mycobacterium tuberculosis. Sci Rep. (2017) 7:2830. 10.1038/s41598-017-02116-1
    1. Parra M, Yang AL, Lim J, Kolibab K, Derrick S, Cadieux N, et al. . Development of a murine mycobacterial growth inhibition assay for evaluating vaccines against Mycobacterium tuberculosis. Clin Vaccine Immunol. (2009) 16:1025–32. 10.1128/CVI.00067-09
    1. McShane H, Williams A. A review of preclinical animal models utilised for TB vaccine evaluation in the context of recent human efficacy data. Tuberculosis. (2014) 94:105–10. 10.1016/j.tube.2013.11.003
    1. O'Shea MK, Tanner R, Müller J, Harris SA, Wright D, Stockdale L, et al. . Immunological correlates of mycobacterial growth inhibition describe a spectrum of tuberculosis infection. Sci Rep. (2018) 8:14480. 10.1038/s41598-018-32755-x
    1. Kolibab K, Parra M, Yang AL, Perera LP, Derrick SC, Morris SL. A practical in vitro growth inhibition assay for the evaluation of TB vaccines. Vaccine. (2009) 28:317–22. 10.1016/j.vaccine.2009.10.047
    1. Orme IM, Miller ES, Roberts AD, Furney SK, Griffin JP, Dobos KM, et al. . T lymphocytes mediating protection and cellular cytolysis during the course of Mycobacterium tuberculosis infection. Evidence for different kinetics and recognition of a wide spectrum of protein antigens. J Immunol. (1992) 148:189–96.
    1. Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med. (1993) 178:2249–54. 10.1084/jem.178.6.2249
    1. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme IM. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med. (1993) 178:2243–7. 10.1084/jem.178.6.2243
    1. Bustamante J, Boisson-Dupuis S, Abel L, Casanova JL. Mendelian susceptibility to mycobacterial disease: genetic, immunological, and clinical features of inborn errors of IFN-γ immunity. Semin Immunol. (2014) 26:454–70. 10.1016/j.smim.2014.09.008
    1. Hoft DF, Worku S, Kampmann B, Whalen CC, Ellner JJ, Hirsch CS, et al. . Investigation of the relationships between immune-mediated inhibition of mycobacterial growth and other potential surrogate markers of protective Mycobacterium tuberculosis immunity. J Infect Dis. (2002) 186:1448–57. 10.1086/344359
    1. Kampmann B, Tena GN, Mzazi S, Eley B, Young DB, Levin M. Novel human in vitro system for evaluating antimycobacterial vaccines. Infect Immun. (2004) 72:6401–7. 10.1128/IAI.72.11.6401-6407.2004
    1. Mittrücker HW, Steinhoff U, Köhler A, Krause M, Lazar D, Mex P, et al. . Poor correlation between BCG vaccination-induced T cell responses and protection against tuberculosis. Proc Natl Acad Sci U S A. (2007) 104:12434–9. 10.1073/pnas.0703510104
    1. Elias D, Akuffo H, Britton S. PPD induced in vitro interferon gamma production is not a reliable correlate of protection against Mycobacterium tuberculosis. Trans R Soc Trop Med Hyg. (2005) 99:363–8. 10.1016/j.trstmh.2004.08.006
    1. Soares AP, Scriba TJ, Joseph S, Harbacheuski R, Murray RA, Gelderbloem SJ, et al. . Bacillus Calmette-Guerin vaccination of human newborns induces T cells with complex cytokine and phenotypic profiles. i. (2008) 180:3569–77. 10.4049/jimmunol.180.5.3569
    1. Amyes E, McMichael AJ, Callan MFC. Human CD4+ T cells are predominantly distributed among six phenotypically and functionally distinct subsets. J Immunol. (2005) 175:5765–73. 10.4049/jimmunol.175.9.5765
    1. Lewinsohn DA, Lewinsohn DM, Scriba TJ. Polyfunctional CD4(+) T Cells As Targets for Tuberculosis Vaccination. Front immunol. (2017) 8:1262. 10.3389/fimmu.2017.01262
    1. Hawkridge T, Scriba TJ, Gelderbloem S, Smit E, Tameris M, Moyo S, et al. . Safety and immunogenicity of a new tuberculosis vaccine, MVA85A, in healthy adults in South Africa. J Infect Dis. (2008) 198:544–52. 10.1086/590185
    1. Beveridge NER, Fletcher HA, Hughes J, Pathan AA, Scriba TJ, Minassian A, et al. . A comparison of IFNγ detection methods used in tuberculosis vaccine trials. Tuberculosis. (2008) 88:631–40. 10.1016/j.tube.2008.06.005
    1. Harris SA, Satti I, Matsumiya M, Stockdale L, Chomka A, Tanner R, et al. . Process of assay selection and optimization for the study of case and control samples from a phase IIb efficacy trial of a candidate tuberculosis vaccine, MVA85A. Clin Vaccine Immunol. (2014) 21:1005–11. 10.1128/CVI.00128-14
    1. Scriba TJ, Kalsdorf B, Abrahams D-A, Isaacs F, Hofmeister J, Black G, et al. Distinct, Specific IL-17- and IL-22-Producing CD4 < sup>+ < /sup> T cell subsets contribute to the human anti-mycobacterial immune response. J. Immunol. (2008) 180:1962–70. 10.4049/jimmunol.180.3.1962
    1. McShane H, Pathan AA, Sander CR, Keating SM, Gilbert SC, Huygen K, et al. . Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat Med. (2004) 10:1240–4. 10.1038/nm1128
    1. Griffiths KL, Pathan AA, Minassian AM, Sander CR, Beveridge NER, Hill AVS, et al. . Th1/Th17 cell induction and corresponding reduction in ATP consumption following vaccination with the novel Mycobacterium tuberculosis vaccine MVA85A. PloS one. (2011) 6:e23463. 10.1371/journal.pone.0023463
    1. Lagercrantz R, Peterson JC, Lind J. Further studies of tuberculin hemagglutination in tuberculous infection, benign and virulent. Acta Paediatr. (1953) 42:113–25. 10.1111/j.1651-2227.1953.tb05573.x
    1. Parlett RC, Youmans GP. An evaluation of the specificty and sensitivity of a gel double-diffusion test for tuberculosis. Am Rev Respir Dis. (1959) 80:153–66.
    1. Turneer M, Van Vooren JP, Nyabenda J, Legros F, Lecomte A, Thiriaux J, et al. . The humoral immune response after BCG vaccination in humans: consequences for the serodiagnosis of tuberculosis. Eur Respir J. (1988) 1:589–93.
    1. Beyazova U, Rota S, Cevheroglu C, Karsligil T. Humoral immune response in infants after BCG vaccination. Tuber Lung Dis. (1995) 76:248–53. 10.1016/S0962-8479(05)80013-9
    1. Tanner R, Villarreal-Ramos B, Vordermeier HM, McShane H. The humoral immune response to BCG Vaccination. Front. Immunol. (2019) 10:1317. 10.3389/fimmu.2019.01317
    1. Jacobs AJ, Mongkolsapaya J, Screaton GR, McShane H, Wilkinson RJ. Antibodies and tuberculosis. Tuberculosis. (2016) 101:102–13. 10.1016/j.tube.2016.08.001
    1. Kawahara JY, Irvine EB, Alter G. A case for antibodies as mechanistic correlates of immunity in tuberculosis. Front. immunol. (2019) 10:996. 10.3389/fimmu.2019.00996
    1. Ma Y, Chen H-D, Wang Y, Wang Q, Li Y, Zhao Y, et al. . Interleukin 24 as a novel potential cytokine immunotherapy for the treatment of Mycobacterium tuberculosis infection. Micro. Infection. (2011) 13:1099–110. 10.1016/j.micinf.2011.06.012
    1. Peng R, Yue J, Han M, Zhao Y, Liu L, Liang L. The IL-17F sequence variant is associated with susceptibility to tuberculosis. Gene. (2013) 515:229–32. 10.1016/j.gene.2012.11.017
    1. Khader SA, Cooper AM. IL-23 and IL-17 in tuberculosis. Cytokine. (2008) 41:79–83. 10.1016/j.cyto.2007.11.022
    1. Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, Cilley GE, et al. . IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat Immunol. (2007) 8:369. 10.1038/ni1449
    1. Happel KI, Lockhart EA, Mason CM, Porretta E, Keoshkerian E, Odden AR, et al. . Pulmonary interleukin-23 gene delivery increases local T-cell immunity and controls growth of Mycobacterium tuberculosis in the lungs. Infect Immun. (2005) 73:5782–8. 10.1128/IAI.73.9.5782-5788.2005
    1. Sever JL, Youmans GP. The relation of oxygen tension to virulence of tubercle bacilli and to acquired resistance in tuberculosis. J Infect Dis. (1957):193–202. 10.1093/infdis/101.2.193
    1. Oehlers Stefan H. Revisiting hypoxia therapies for tuberculosis. Clin Sci. (2019) 133:1271–80. 10.1042/CS20190415
    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) 7:1129 10.1038/ncomms11633
    1. Fletcher HA, Filali-Mouhim A, Nemes E, Hawkridge A, Keyser A, Njikan S, et al. . Human newborn bacille Calmette-Guérin vaccination and risk of tuberculosis disease: a case-control study. BMC Med. (2016) 14:76. 10.1186/s12916-016-0617-3
    1. Muller J, Tanner R, Matsumiya M, Snowden MA, Landry B, Satti I, et al. Cytomegalovirus infection is a risk factor for TB disease in infants. JCI Insight. (2019) 4:e130090 10.1172/jci.insight.130090
    1. Westendorf AM, Skibbe K, Adamczyk A, Buer J, Geffers R, Hansen W, et al. . Hypoxia enhances immunosuppression by inhibiting CD4+ effector T cell function and promoting treg activity. Cell Physiol Biochem. (2017) 41:1271–84. 10.1159/000464429
    1. Thiel M, Caldwell CC, Kreth S, Kuboki S, Chen P, Smith P, et al. . Targeted deletion of HIF-1α gene in T cells prevents their inhibition in hypoxic inflamed tissues and improves septic mice survival. PLoS ONE. (2007) 2:e853. 10.1371/journal.pone.0000853
    1. Ryan EM, Whyte MKB, Walmsley SR. Hypoxia and reprogramming of host pathogen interactions. Curr Opin Physiol. (2019) 7:15–20. 10.1016/j.cophys.2018.11.004
    1. Tanner R, O'Shea MK, White AD, Müller J, Harrington-Kandt R, Matsumiya M, et al. . The influence of haemoglobin and iron on in vitro mycobacterial growth inhibition assays. Sci Rep. (2017) 7:43478. 10.1038/srep43478
    1. O'Shea MK, Fletcher TE, Muller J, Tanner R, Matsumiya M, Bailey JW, et al. . Human hookworm infection enhances mycobacterial growth inhibition and associates with reduced risk of tuberculosis infection. Front Immunol. (2018) 9:2893. 10.3389/fimmu.2018.02893

Source: PubMed

3
Subskrybuj