World Health Organization. Global Tuberculosis Report. (2023).
Targeted tuberculin testing and treatment of latent tuberculosis infection. This official statement of the American Thoracic Society was adopted by the ATS Board of Directors, July 1999. This is a Joint Statement of the American Thoracic Society (ATS) and the Centers for Disease Control and Prevention (CDC). This statement was endorsed by the Council of the Infectious Diseases Society of America. (IDSA), September 1999, and the sections of this statement. Am. J. Respir. Crit. Care Med. 161, S221–S247 (2000).
Lillebaek, T. et al. Molecular evidence of endogenous reactivation of Mycobacterium tuberculosis after 33 years of latent infection. J. Infect. Dis. 185, 401–404 (2002).
Google Scholar
Mangtani, P. et al. Protection by BCG vaccine against tuberculosis: a systematic review of randomized controlled trials. Clin. Infect. Dis. 58, 470–480 (2014).
Google Scholar
Lai, R., Ogunsola, A. F., Rakib, T. & Behar, S. M. Key advances in vaccine development for tuberculosis-success and challenges. NPJ Vaccines 8, 158 (2023).
Google Scholar
Bhatt, K., Verma, S., Ellner, J. J. & Salgame, P. Quest for correlates of protection against tuberculosis. Clin. Vaccin. Immunol. 22, 258–266 (2015).
Google Scholar
Britto, C. & Alter, G. The next frontier in vaccine design: blending immune correlates of protection into rational vaccine design. Curr. Opin. Immunol. 78, 102234 (2022).
Google Scholar
Escudero-Perez, B., Lawrence, P. & Castillo-Olivares, J. Immune correlates of protection for SARS-CoV-2, Ebola and Nipah virus infection. Front. Immunol. 14, 1156758 (2023).
Google Scholar
Chandra, P., Grigsby, S. J. & Philips, J. A. Immune evasion and provocation by Mycobacterium tuberculosis. Nat. Rev. Microbiol 20, 750–766 (2022).
Google Scholar
Yang, J., Zhang, L., Qiao, W. & Luo, Y. Mycobacterium tuberculosis: pathogenesis and therapeutic targets. MedComm (2020) 4, e353 (2023).
Google Scholar
Prezzemolo, T. et al. Functional signatures of human CD4 and CD8 T cell responses to Mycobacterium tuberculosis. Front. Immunol. 5, 180 (2014).
Google Scholar
Derrick, S. C., Yabe, I. M., Yang, A. & Morris, S. L. Vaccine-induced anti-tuberculosis protective immunity in mice correlates with the magnitude and quality of multifunctional CD4 T cells. Vaccine 29, 2902–2909 (2011).
Google Scholar
Sakai, S., Mayer-Barber, K. D. & Barber, D. L. Defining features of protective CD4 T cell responses to Mycobacterium tuberculosis. Curr. Opin. Immunol. 29, 137–142 (2014).
Google Scholar
Flynn, J. L. et al. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J. Exp. Med. 178, 2249–2254 (1993).
Google Scholar
Cooper, A. M. et al. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J. Exp. Med. 178, 2243–2247 (1993).
Google Scholar
Sakai, S. et al. CD4 T cell-derived IFN-gamma plays a minimal role in control of pulmonary Mycobacterium tuberculosis infection and must be actively repressed by PD-1 to prevent lethal disease. PLoS Pathog. 12, e1005667 (2016).
Google Scholar
McShane, H., Brookes, R., Gilbert, S. C. & Hill, A. V. Enhanced immunogenicity of CD4(+) t-cell responses and protective efficacy of a DNA-modified vaccinia virus Ankara prime-boost vaccination regimen for murine tuberculosis. Infect. Immun. 69, 681–686 (2001).
Google Scholar
Williams, A. et al. Boosting with poxviruses enhances Mycobacterium bovis BCG efficacy against tuberculosis in guinea pigs. Infect. Immun. 73, 3814–3816 (2005).
Google Scholar
Vordermeier, H. M. et al. Cellular immune responses induced in cattle by heterologous prime-boost vaccination using recombinant viruses and bacille Calmette-Guerin. Immunology 112, 461–470 (2004).
Google Scholar
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).
Google Scholar
McShane, H. et al. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat. Med. 10, 1240–1244 (2004).
Google Scholar
Scriba, T. J. et al. Modified vaccinia Ankara-expressing Ag85A, a novel tuberculosis vaccine, is safe in adolescents and children, and induces polyfunctional CD4+ T cells. Eur. J. Immunol. 40, 279–290 (2010).
Google Scholar
Tameris, M. D. 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 381, 1021–1028 (2013).
Google Scholar
Khader, S. A. 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. 8, 369–377 (2007).
Google Scholar
Okamoto Yoshida, Y. et al. Essential role of IL-17A in the formation of a mycobacterial infection-induced granuloma in the lung. J. Immunol. 184, 4414–4422 (2010).
Google Scholar
Shanmugasundaram, U. et al. Pulmonary Mycobacterium tuberculosis control associates with CXCR3- and CCR6-expressing antigen-specific Th1 and Th17 cell recruitment. JCI Insight 5, e137858 (2020).
Google Scholar
Ottenhoff, T. H. New pathways of protective and pathological host defense to mycobacteria. Trends Microbiol. 20, 419–428 (2012).
Google Scholar
Miranda, N. & Hoyer, K. K. Coccidioidomycosis granulomas informed by other diseases: advancements, gaps, and challenges. J Fungi (Basel) 9, 650 (2023).
Google Scholar
Ogongo, P. et al. Tissue-resident-like CD4+ T cells secreting IL-17 control Mycobacterium tuberculosis in the human lung. J. Clin. Invest. 131, e142014 (2021).
Google Scholar
Freches, D. et al. Mice genetically inactivated in interleukin-17A receptor are defective in long-term control of Mycobacterium tuberculosis infection. Immunology 140, 220–231 (2013).
Google Scholar
Dijkman, K. et al. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep. Med. 2, 100187 (2021).
Google Scholar
Jong, R. M. et al. Mucosal vaccination with cyclic dinucleotide adjuvants induces effective T cell homing and IL-17-dependent protection against Mycobacterium tuberculosis infection. J. Immunol. 208, 407–419 (2022).
Google Scholar
Aguilo, N. et al. Pulmonary but not subcutaneous delivery of BCG vaccine confers protection to tuberculosis-susceptible mice by an interleukin 17-dependent mechanism. J. Infect. Dis. 213, 831–839 (2016).
Google Scholar
Counoupas, C. et al. Mucosal delivery of a multistage subunit vaccine promotes development of lung-resident memory T cells and affords interleukin-17-dependent protection against pulmonary tuberculosis. NPJ Vaccines 5, 105 (2020).
Google Scholar
Butcher, D. L. & Deng, H. W. Hypothetical SisterKiller. Nature 369, 26 (1994).
Google Scholar
Wu, J. et al. Incorporation of immunostimulatory motifs in the transcribed region of a plasmid DNA vaccine enhances Th1 immune responses and therapeutic effect against Mycobacterium tuberculosis in mice. Vaccine 29, 7624–7630 (2011).
Google Scholar
Desvignes, L. & Ernst, J. D. Interferon-gamma-responsive nonhematopoietic cells regulate the immune response to Mycobacterium tuberculosis. Immunity 31, 974–985 (2009).
Google Scholar
Blanco, F. C. et al. Increased IL-17 expression is associated with pathology in a bovine model of tuberculosis. Tuberculosis 91, 57–63 (2011).
Google Scholar
Jurado, J. O. et al. IL-17 and IFN-gamma expression in lymphocytes from patients with active tuberculosis correlates with the severity of the disease. J. Leukoc. Biol. 91, 991–1002 (2012).
Google Scholar
Cruz, A. et al. Pathological role of interleukin 17 in mice subjected to repeated BCG vaccination after infection with Mycobacterium tuberculosis. J. Exp. Med. 207, 1609–1616 (2010).
Google Scholar
Kaufmann, S. H. Immunity to intracellular bacteria. Annu. Rev. Immunol. 11, 129–163 (1993).
Google Scholar
Ali, A. et al. Recent advancement, immune responses, and mechanism of action of various vaccines against intracellular bacterial infections. Life Sci. 314, 121332 (2023).
Google Scholar
van Pinxteren, L. A., Cassidy, J. P., Smedegaard, B. H., Agger, E. M. & Andersen, P. Control of latent Mycobacterium tuberculosis infection is dependent on CD8 T cells. Eur. J. Immunol. 30, 3689–3698 (2000).
Google Scholar
Sousa, A. O. et al. Relative contributions of distinct MHC class I-dependent cell populations in protection to tuberculosis infection in mice. Proc. Natl Acad. Sci. USA 97, 4204–4208 (2000).
Google Scholar
Villarreal-Ramos, B. et al. Investigation of the role of CD8+ T cells in bovine tuberculosis in vivo. Infect. Immun. 71, 4297–4303 (2003).
Google Scholar
Chen, C. Y. et al. A critical role for CD8 T cells in a nonhuman primate model of tuberculosis. PLoS Pathog. 5, e1000392 (2009).
Google Scholar
Wang, J., Santosuosso, M., Ngai, P., Zganiacz, A. & Xing, Z. Activation of CD8 T cells by mycobacterial vaccination protects against pulmonary tuberculosis in the absence of CD4 T cells. J. Immunol. 173, 4590–4597 (2004).
Google Scholar
Wu, Y., Woodworth, J. S., Shin, D. S., Morris, S. & Behar, S. M. Vaccine-elicited 10-kilodalton culture filtrate protein-specific CD8+ T cells are sufficient to mediate protection against Mycobacterium tuberculosis infection. Infect. Immun. 76, 2249–2255 (2008).
Google Scholar
Hu, Z. et al. Sendai virus mucosal vaccination establishes lung-resident memory CD8 T cell immunity and boosts BCG-primed protection against TB in mice. Mol. Ther. 25, 1222–1233 (2017).
Google Scholar
Moliva, J. I. et al. Exposure to human alveolar lining fluid enhances Mycobacterium bovis BCG vaccine efficacy against Mycobacterium tuberculosis infection in a CD8(+) T-cell-dependent manner. Mucosal Immunol. 11, 968–978 (2018).
Google Scholar
Vasilyev, K. et al. Enhancement of the local CD8(+) T-cellular immune response to Mycobacterium tuberculosis in BCG-primed mice after intranasal administration of influenza vector vaccine carrying TB10.4 and HspX antigens. Vaccines (Basel) 9, 1273 (2021).
Google Scholar
Baldwin, S. L. et al. Protection against tuberculosis with homologous or heterologous protein/vector vaccine approaches is not dependent on CD8+ T cells. J. Immunol. 191, 2514–2525 (2013).
Google Scholar
Lindenstrom, T., Aagaard, C., Christensen, D., Agger, E. M. & Andersen, P. High-frequency vaccine-induced CD8(+) T cells specific for an epitope naturally processed during infection with Mycobacterium tuberculosis do not confer protection. Eur. J. Immunol. 44, 1699–1709 (2014).
Google Scholar
Hu, Z. et al. A multistage Sendai virus vaccine incorporating latency-associated antigens induces protection against acute and latent tuberculosis. Emerg. Microbes Infect. 13, 2300463 (2024).
Google Scholar
Ashhurst, A. S. et al. PLGA particulate subunit tuberculosis vaccines promote humoral and Th17 responses but do not enhance control of Mycobacterium tuberculosis infection. PLoS ONE 13, e0194620 (2018).
Google Scholar
Lu, L. L. et al. IFN-gamma-independent immune markers of Mycobacterium tuberculosis exposure. Nat. Med. 25, 977–987 (2019).
Google Scholar
Davies, L. R. L. et al. Age and sex influence antibody profiles associated with tuberculosis progression. Nat. Microbiol. 9, 1513–1525 (2024).
Google Scholar
Ishida, E. et al. Mucosal and systemic antigen-specific antibody responses correlate with protection against active tuberculosis in nonhuman primates. EBioMedicine 99, 104897 (2024).
Google Scholar
Chen, T. et al. Capsular glycan recognition provides antibody-mediated immunity against tuberculosis. J. Clin. Invest. 130, 1808–1822 (2020).
Google Scholar
Lu, L. L. et al. A functional role for antibodies in tuberculosis. Cell 167, 433–443 e414 (2016).
Google Scholar
du Plessis, W. J. et al. The functional response of B cells to antigenic stimulation: a preliminary report of latent tuberculosis. PLoS ONE 11, e0152710 (2016).
Google Scholar
Dubois Cauwelaert, N. et al. Antigen presentation by B cells guides programing of memory CD4(+) T-cell responses to a TLR4-agonist containing vaccine in mice. Eur. J. Immunol. 46, 2719–2729 (2016).
Google Scholar
Phuah, J. Y., Mattila, J. T., Lin, P. L. & Flynn, J. L. Activated B cells in the granulomas of nonhuman primates infected with Mycobacterium tuberculosis. Am. J. Pathol. 181, 508–514 (2012).
Google Scholar
Hunter, L., Hingley-Wilson, S., Stewart, G. R., Sharpe, S. A. & Salguero, F. J. Dynamics of macrophage, T and B cell infiltration within pulmonary granulomas induced by Mycobacterium tuberculosis in two non-human primate models of aerosol infection. Front. Immunol. 12, 776913 (2021).
Google Scholar
Carpenter, S. M. & Lu, L. L. Leveraging antibody, B cell and Fc receptor interactions to understand heterogeneous immune responses in tuberculosis. Front Immunol. 13, 830482 (2022).
Google Scholar
Stewart, P. et al. Role of B cells in Mycobacterium tuberculosis infection. Vaccines (Basel) 11, 955 (2023).
Google Scholar
Rijnink, W. F., Ottenhoff, T. H. M. & Joosten, S. A. B-cells and antibodies as contributors to effector immune responses in tuberculosis. Front. Immunol. 12, 640168 (2021).
Google Scholar
Achkar, J. M., Chan, J. & Casadevall, A. B cells and antibodies in the defense against Mycobacterium tuberculosis infection. Immunol. Rev. 264, 167–181 (2015).
Google Scholar
Schenkel, J. M. & Masopust, D. Tissue-resident memory T cells. Immunity 41, 886–897 (2014).
Google Scholar
Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune defence. Nat. Rev. Immunol. 16, 79–89 (2016).
Google Scholar
Beverley, P. C., Sridhar, S., Lalvani, A. & Tchilian, E. Z. Harnessing local and systemic immunity for vaccines against tuberculosis. Mucosal Immunol. 7, 20–26 (2014).
Google Scholar
Urdahl, K. B. Understanding and overcoming the barriers to T cell-mediated immunity against tuberculosis. Semin. Immunol. 26, 578–587 (2014).
Google Scholar
Hu, Z., Lu, S. H., Lowrie, D. B. & Fan, X. Y. Research advances for virus-vectored tuberculosis vaccines and latest findings on tuberculosis vaccine development. Front. Immunol. 13, 895020 (2022).
Google Scholar
Sakai, S. et al. Cutting edge: control of Mycobacterium tuberculosis infection by a subset of lung parenchyma-homing CD4 T cells. J. Immunol. 192, 2965–2969 (2014).
Google Scholar
Torrado, E. et al. Interleukin 27R regulates CD4+ T cell phenotype and impacts protective immunity during Mycobacterium tuberculosis infection. J. Exp. Med 212, 1449–1463 (2015).
Google Scholar
Hu, Z. et al. The role of KLRG1 in human CD4+ T-cell immunity against tuberculosis. J. Infect. Dis. 217, 1491–1503 (2018).
Google Scholar
Perdomo, C. et al. Mucosal BCG vaccination induces protective lung-resident memory T cell populations against tuberculosis. mBio 7, e01686-16 (2016).
Google Scholar
Woodworth, J. S. et al. Subunit vaccine H56/CAF01 induces a population of circulating CD4 T cells that traffic into the Mycobacterium tuberculosis-infected lung. Mucosal Immunol. 10, 555–564 (2017).
Google Scholar
Carpenter, S. M., Yang, J. D., Lee, J., Barreira-Silva, P. & Behar, S. M. Vaccine-elicited memory CD4+ T cell expansion is impaired in the lungs during tuberculosis. PLoS Pathog. 13, e1006704 (2017).
Google Scholar
Jeyanathan, M. et al. CXCR3 signaling is required for restricted homing of parenteral tuberculosis vaccine-induced T cells to both the lung parenchyma and airway. J. Immunol. 199, 2555–2569 (2017).
Google Scholar
Lindenstrom, T. et al. T cells primed by live mycobacteria versus a tuberculosis subunit vaccine exhibit distinct functional properties. EBioMedicine 27, 27–39 (2018).
Google Scholar
Darrah, P. A. et al. Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature 577, 95–102 (2020).
Google Scholar
Netea, M. G., Quintin, J. & van der Meer, J. W. Trained immunity: a memory for innate host defense. Cell Host Microbe 9, 355–361 (2011).
Google Scholar
Netea, M. G. & van der Meer, J. W. Trained immunity: an ancient way of remembering. Cell Host Microbe 21, 297–300 (2017).
Google Scholar
Hu, Z., Lu, S. H., Lowrie, D. B. & Fan, X. Y. Trained immunity: a Yin-Yang balance. MedComm (2020) 3, e121 (2022).
Google Scholar
Cheng, S. H. et al. Demonstration of increased anti-mycobacterial activity in peripheral blood monocytes after BCG vaccination in British school children. Clin. Exp. Immunol. 74, 20–25 (1988).
Google Scholar
Cheng, S. H. et al. Monocyte antimycobacterial activity before and after Mycobacterium bovis BCG vaccination in Chingleput, India, and London, United Kingdom. Infect. Immun. 61, 4501–4503 (1993).
Google Scholar
Kleinnijenhuis, J. et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl Acad. Sci. USA 109, 17537–17542 (2012).
Google Scholar
Kleinnijenhuis, J. et al. Long-lasting effects of BCG vaccination on both heterologous Th1/Th17 responses and innate trained immunity. J. Innate Immun. 6, 152–158 (2014).
Google Scholar
Arts, R. J. W. et al. Immunometabolic pathways in BCG-induced trained immunity. Cell Rep. 17, 2562–2571 (2016).
Google Scholar
Kaufmann, E. et al. BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell 172, 176–190.e119 (2018).
Google Scholar
Giamarellos-Bourboulis, E. J. et al. Activate: randomized clinical trial of BCG vaccination against infection in the elderly. Cell 183, 315–323.e319 (2020).
Google Scholar
Xu, J. C. et al. Multi-omics analysis reveals that linoleic acid metabolism is associated with variations of trained immunity induced by distinct BCG strains. Sci. Adv. 10, eadk8093 (2024).
Google Scholar
Cohen, S. B. et al. Alveolar macrophages provide an early mycobacterium tuberculosis niche and initiate dissemination. Cell Host Microbe 24, 439–446 e434 (2018).
Google Scholar
Rothchild, A. C. et al. Alveolar macrophages generate a noncanonical NRF2-driven transcriptional response to Mycobacterium tuberculosis in vivo. Sci. Immunol. 4, eaaw6693 (2019).
Rajaram, M. V., Ni, B., Dodd, C. E. & Schlesinger, L. S. Macrophage immunoregulatory pathways in tuberculosis. Semin. Immunol. 26, 471–485 (2014).
Google Scholar
Lovey, A. et al. Early alveolar macrophage response and IL-1R-dependent T cell priming determine transmissibility of Mycobacterium tuberculosis strains. Nat. Commun. 13, 884 (2022).
Google Scholar
Mai, D. et al. Exposure to Mycobacterium remodels alveolar macrophages and the early innate response to Mycobacterium tuberculosis infection. PLoS Pathog. 20, e1011871 (2024).
Google Scholar
Yao, Y. et al. Induction of autonomous memory alveolar macrophages requires T cell help and is critical to trained immunity. Cell 175, 1634–1650.e1617 (2018).
Google Scholar
D’Agostino, M. R. et al. Airway macrophages mediate mucosal vaccine-induced trained innate immunity against Mycobacterium tuberculosis in early stages of infection. J. Immunol. 205, 2750–2762 (2020).
Google Scholar
Mata, E. et al. Pulmonary BCG induces lung-resident macrophage activation and confers long-term protection against tuberculosis. Sci. Immunol. 6, eabc2934 (2021).
Google Scholar
Jeyanathan, M. et al. Parenteral BCG vaccine induces lung-resident memory macrophages and trained immunity via the gut-lung axis. Nat. Immunol. 23, 1687–1702 (2022).
Google Scholar
Ravesloot-Chavez, M. M., Van Dis, E. & Stanley, S. A. The innate immune response to Mycobacterium tuberculosis infection. Annu Rev. Immunol. 39, 611–637 (2021).
Google Scholar
Larsen, S. E., Williams, B. D., Rais, M., Coler, R. N. & Baldwin, S. L. It takes a village: the multifaceted immune response to Mycobacterium tuberculosis infection and vaccine-induced immunity. Front. Immunol. 13, 840225 (2022).
Google Scholar
Kim, H., Choi, H. G. & Shin, S. J. Bridging the gaps to overcome major hurdles in the development of next-generation tuberculosis vaccines. Front. Immunol. 14, 1193058 (2023).
Google Scholar
Morrison, H. & McShane, H. Local pulmonary immunological biomarkers in tuberculosis. Front. Immunol. 12, 640916 (2021).
Google Scholar
Huang, X., Lowrie, D. B., Fan, X. Y. & Hu, Z. Natural products in anti-tuberculosis host-directed therapy. Biomed. Pharmacother. 171, 116087 (2024).
Google Scholar
Tanner, R., O’Shea, M. K., Fletcher, H. A. & McShane, H. In vitro mycobacterial growth inhibition assays: a tool for the assessment of protective immunity and evaluation of tuberculosis vaccine efficacy. Vaccine 34, 4656–4665 (2016).
Google Scholar
Tanner, R. et al. The in vitro direct mycobacterial growth inhibition assay (MGIA) for the early evaluation of TB vaccine candidates and assessment of protective immunity: a protocol for non-human primate cells. F1000Res 10, 257 (2021).
Google Scholar
Tanner, R. et al. A non-human primate in vitro functional assay for the early evaluation of TB vaccine candidates. NPJ Vaccines 6, 3 (2021).
Google Scholar
Painter, H. et al. Demonstrating the utility of the ex vivo murine mycobacterial growth inhibition assay (MGIA) for high-throughput screening of tuberculosis vaccine candidates against multiple Mycobacterium tuberculosis complex strains. Tuberculosis 146, 102494 (2024).
Google Scholar
Hoft, S. G. et al. Imprinting of gut-homing receptors on Mtb-specific Th1* cells is associated with reduced lung homing after gavage BCG vaccination of Rhesus macaques. mBio 14, e0022023 (2023).
Google Scholar
Nemeth, J. et al. Contained Mycobacterium tuberculosis infection induces concomitant and heterologous protection. PLoS Pathog. 16, e1008655 (2020).
Google Scholar
Hamasur, B. et al. Rapid diagnosis of tuberculosis by detection of mycobacterial lipoarabinomannan in urine. J. Microbiol. Methods 45, 41–52 (2001).
Google Scholar
Flores, J., Cancino, J. C. & Chavez-Galan, L. Lipoarabinomannan as a point-of-care assay for diagnosis of tuberculosis: how far are we to use it? Front. Microbiol. 12, 638047 (2021).
Google Scholar
Gu, W. F. et al. Effectiveness of Histopathological Examination of Ultrasound-guided Puncture Biopsy Samples for Diagnosis of Extrapulmonary Tuberculosis. Biomed. Environ. Sci. 37, 170–177 (2024).
Google Scholar
Bohrer, A. C. et al. Eosinophils are part of the granulocyte response in tuberculosis and promote host resistance in mice. J. Exp. Med. 218, e20210469 (2021).
Hu, Z. et al. Pathomorphological characteristics of tuberculous placenta and its clinical implication. Diagn. Pathol. 18, 128 (2023).
Google Scholar
Saktiawati, A. M. I., Putera, D. D., Setyawan, A., Mahendradhata, Y. & van der Werf, T. S. Diagnosis of tuberculosis through breath test: a systematic review. EBioMedicine 46, 202–214 (2019).
Google Scholar
Phillips, M. et al. Point-of-care breath test for biomarkers of active pulmonary tuberculosis. Tuberculosis 92, 314–320 (2012).
Google Scholar
Coussens, A. K. et al. Classification of early tuberculosis states to guide research for improved care and prevention: an international Delphi consensus exercise. Lancet Respir. Med. 12, 484–498 (2024).
Google Scholar
Gordon, S. B. et al. A framework for Controlled Human Infection Model (CHIM) studies in Malawi: report of a Wellcome Trust workshop on CHIM in Low Income Countries held in Blantyre, Malawi. Wellcome Open Res. 2, 70 (2017).
Google Scholar
Minassian, A. M. et al. A human challenge model for Mycobacterium tuberculosis using Mycobacterium bovis bacille Calmette-Guerin. J. Infect. Dis. 205, 1035–1042 (2012).
Google Scholar
Harris, S. A. 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. 209, 1259–1268 (2014).
Google Scholar
Blazevic, A. et al. Phase 1 open-label dose escalation trial for the development of a human bacillus Calmette-Guerin challenge model for assessment of tuberculosis immunity in vivo. J. Infect. Dis. 229, 1498–1508 (2024).
Google Scholar
Satti, I. et al. Safety of a controlled human infection model of tuberculosis with aerosolised, live-attenuated Mycobacterium bovis BCG versus intradermal BCG in BCG-naive adults in the UK: a dose-escalation, randomised, controlled, phase 1 trial. Lancet Infect. Dis. 24, 909–921 (2024).
Davids, M. et al. A human lung challenge model to evaluate the safety and immunogenicity of PPD and live bacillus Calmette-Guerin. Am. J. Respir. Crit. Care Med. 201, 1277–1291 (2020).
Google Scholar
Basu Roy, R. et al. An auto-luminescent fluorescent BCG whole blood assay to enable evaluation of paediatric mycobacterial responses using minimal blood volumes. Front. Pediatr. 7, 151 (2019).
Google Scholar
Bekeredjian-Ding, I. et al. Human challenge trial workshop: focus on quality requirements for challenge agents, Langen, Germany, October 22, 2019. Biologicals 66, 53–61 (2020).
Google Scholar
Wang, X. et al. Development of an engineered Mycobacterium tuberculosis strain for a safe and effective tuberculosis human challenge model. bioRxiv https://doi.org/10.1101/2023.11.19.567569 (2023).
Balasingam, S. et al. Review of the current TB human infection studies for use in accelerating TB vaccine development: a meeting report. J. Infect. Dis. 230, e457–e464 (2024).
Donald, P. R. et al. Droplets, dust and guinea pigs: an historical review of tuberculosis transmission research, 1878-1940. Int. J. Tuberc. Lung Dis. 22, 972–982 (2018).
Google Scholar
Plumlee, C. R. et al. Ultra-low dose aerosol infection of mice with Mycobacterium tuberculosis more closely models human tuberculosis. Cell Host Microbe 29, 68–82 e65 (2021).
Google Scholar
Vidal, S. J. et al. Attenuated Mycobacterium tuberculosis vaccine protection in a low-dose murine challenge model. iScience 26, 106963 (2023).
Google Scholar
Plumlee, C. R. et al. Assessing vaccine-mediated protection in an ultra-low dose Mycobacterium tuberculosis murine model. PLoS Pathog. 19, e1011825 (2023).
Google Scholar
Kwon, K. W. et al. Immunogenicity and protective efficacy of RipA, a peptidoglycan hydrolase, against Mycobacterium tuberculosis Beijing outbreak strains. Vaccine 42, 1941–1952 (2024).
Google Scholar
Smith, C. M. et al. Host-pathogen genetic interactions underlie tuberculosis susceptibility in genetically diverse mice. Elife 11, e74419 (2022).
Lai, R. et al. Host genetic background is a barrier to broadly effective vaccine-mediated protection against tuberculosis. J. Clin. Invest. 133, e167762 (2023).
Ahmed, M. et al. Immune correlates of tuberculosis disease and risk translate across species. Sci. Transl. Med. 12, eaay0233 (2020).
Kurtz, S. L. et al. The diversity outbred mouse population is an improved animal model of vaccination against tuberculosis that reflects heterogeneity of protection. mSphere 5, https://doi.org/10.1128/msphere.00097-20 (2020).
Kurtz, S. L. et al. Multiple genetic loci influence vaccine-induced protection against Mycobacterium tuberculosis in genetically diverse mice. PLoS Pathog. 20, e1012069 (2024).
Google Scholar
Leroux-Roels, I. et al. Improved CD4(+) T cell responses to Mycobacterium tuberculosis in PPD-negative adults by M72/AS01 as compared to the M72/AS02 and Mtb72F/AS02 tuberculosis candidate vaccine formulations: a randomized trial. Vaccine 31, 2196–2206 (2013).
Google Scholar
Tait, D. R. et al. Final analysis of a trial of M72/AS01(E) vaccine to prevent tuberculosis. N. Engl. J. Med. 381, 2429–2439 (2019).
Google Scholar
Nemes, E. et al. The quest for vaccine-induced immune correlates of protection against tuberculosis. Vaccin. Insights 1, 165–181 (2022).
Google Scholar
Hansen, S. G. et al. Prevention of tuberculosis in rhesus macaques by a cytomegalovirus-based vaccine. Nat. Med. 24, 130–143 (2018).
Google Scholar
Larson, E. C. et al. Intravenous Bacille Calmette-Guerin vaccination protects simian immunodeficiency virus-infected macaques from tuberculosis. Nat. Microbiol. 8, 2080–2092 (2023).
Google Scholar
Darrah, P. A. et al. Airway T cells are a correlate of i.v. Bacille Calmette-Guerin-mediated protection against tuberculosis in rhesus macaques. Cell Host Microbe 31, 962–977 e968 (2023).
Google Scholar
Peters, J. M. et al. Protective intravenous BCG vaccination induces enhanced immune signaling in the airways. bioRxiv https://doi.org/10.1101/2023.07.16.549208 (2023).
Irvine, E. B. et al. Humoral correlates of protection against Mycobacterium tuberculosis following intravenous Bacille Calmette-Guerin vaccination in Rhesus macaques. bioRxiv https://doi.org/10.1101/2023.07.31.551245 (2023).
Simonson, A. W. et al. CD4 T cells and CD8alpha+ lymphocytes are necessary for intravenous BCG-induced protection against tuberculosis in macaques. bioRxiv https://doi.org/10.1101/2024.05.14.594183 (2024).
Liu, Y. E. et al. Blood transcriptional correlates of BCG-induced protection against tuberculosis in Rhesus macaques. Cell Rep. Med. 4, 101096 (2023).
Google Scholar
Flores-Valdez, M. A., Kupz, A. & Subbian, S. Recent developments in mycobacteria-based live attenuated vaccine candidates for tuberculosis. Biomedicines 10, 2749 (2022).
Google Scholar
Shah, M. & Dorman, S. E. Latent tuberculosis infection. N. Engl. J. Med. 385, 2271–2280 (2021).
Google Scholar
Zhao, H. M. et al. Differential T cell responses against DosR-associated antigen Rv2028c in BCG-vaccinated populations with tuberculosis infection. J. Infect. 78, 275–280 (2019).
Google Scholar
Jenum, S. et al. A phase I/II randomized trial of H56:IC31 vaccination and adjunctive cyclooxygenase-2-inhibitor treatment in tuberculosis patients. Nat. Commun. 12, 6774 (2021).
Google Scholar
Day, T. A. et al. Safety and immunogenicity of the adjunct therapeutic vaccine ID93 + GLA-SE in adults who have completed treatment for tuberculosis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet Respir. Med. 9, 373–386 (2021).
Google Scholar
Sagawa, Z. K. et al. Safety and immunogenicity of a thermostable ID93 + GLA-SE tuberculosis vaccine candidate in healthy adults. Nat. Commun. 14, 1138 (2023).
Google Scholar
Chen, Z. et al. A multistage protein subunit vaccine as BCG-booster confers protection against Mycobacterium tuberculosis infection in murine models. Int. Immunopharmacol. 139, 112811 (2024).
Google Scholar
Simmons, J. D. et al. Immunological mechanisms of human resistance to persistent Mycobacterium tuberculosis infection. Nat. Rev. Immunol. 18, 575–589 (2018).
Google Scholar
Chen, Z. Y. et al. Decreased expression of CD69 on T cells in tuberculosis infection resisters. Front. Microbiol. 11, 1901 (2020).
Google Scholar
Jalbert, E. et al. Comparative immune responses to Mycobacterium tuberculosis in people with latent infection or sterilizing protection. iScience 26, 107425 (2023).
Google Scholar
Davies, L. R. L. et al. IFN-gamma independent markers of Mycobacterium tuberculosis exposure among male South African gold miners. EBioMedicine 93, 104678 (2023).
Google Scholar
Krishnananthasivam, S. et al. An anti-LpqH human monoclonal antibody from an asymptomatic individual mediates protection against Mycobacterium tuberculosis. NPJ Vaccines 8, 127 (2023).
Google Scholar
Cross, D. L. et al. MR1-restricted T cell clonotypes are associated with “resistance” to Mycobacterium tuberculosis infection. JCI Insight 9, e166505 (2024).
Google Scholar
Simmons, J. D. et al. Monocyte transcriptional responses to Mycobacterium tuberculosis associate with resistance to tuberculin skin test and interferon gamma release assay conversion. mSphere 7, e0015922 (2022).
Google Scholar
Simmons, J. D. et al. Monocyte metabolic transcriptional programs associate with resistance to tuberculin skin test/interferon-gamma release assay conversion. J. Clin. Invest. 131, e140073 (2021).
Google Scholar
Dill-McFarland, K. A. et al. Epigenetic programming of host lipid metabolism associates with resistance to TST/IGRA conversion after exposure to Mycobacterium tuberculosis. bioRxiv https://doi.org/10.1101/2024.02.27.582348 (2024).
Zhang, F. et al. HDAC6 contributes to human resistance against Mycobacterium tuberculosis infection via mediating innate immune responses. FASEB J. 35, e22009 (2021).
Google Scholar
Weiner, J. et al. Changes in transcript, metabolite, and antibody reactivity during the early protective immune response in humans to Mycobacterium tuberculosis infection. Clin. Infect. Dis. 71, 30–40 (2020).
Google Scholar
Ran, F. et al. Whole-transcriptome sequencing of phagocytes reveals a ceRNA network contributing to natural resistance to tuberculosis infection. Micro Pathog. 192, 106681 (2024).
Google Scholar
Nam, Y. et al. Harnessing artificial intelligence in multimodal omics data integration: paving the path for the next frontier in precision medicine. Annu. Rev. Biomed. Data Sci. 7, 225–250 (2024).
Google Scholar