38
Views
4
CrossRef citations to date
0
Altmetric
Review

Tuberculosis vaccines: present and future

&
Pages 721-738 | Published online: 09 Jan 2014

References

  • Global Tuberculosis Control – Surveillance, Planning, Financing. WHO Report (2008).
  • Matteelli A, Migliori GB, Cirillo D, Centis R, Girard E, Raviglion M. Multidrug-resistant and extensively drug-resistant Mycobacterium tuberculosis: epidemiology and control. Expert Rev. Anti. Infect. Ther.5(5), 857–871 (2007).
  • Dye C, Watt CJ, Bleed DM, Hosseini SM, Raviglione MC. Evolution of tuberculosis control and prospects for reducing tuberculosis incidence, prevalence, and deaths globally. JAMA293(22), 2767–2775 (2005).
  • Corbett EL, Watt CJ, Walker N et al. The growing burden of tuberculosis: global trends and interactions with the HIV epidemic. Arch. Intern. Med.163(9), 1009–1021 (2003).
  • Girardi E, Raviglione MC, Antonucci G, Godfrey-Faussett P, Ippolito G. Impact of the HIV epidemic on the spread of other diseases: the case of tuberculosis. AIDS14(Suppl. 3), S47–S56 (2000).
  • Stop TB Partnership. The global plan to stop TB, 2006–2015. Actions for life: towards a world free of tuberculosis. Int. J. Tuberc. Lung Dis.10(3), 240–241 (2006).
  • 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. Lancet367(9517), 1173–1180 (2006).
  • Colditz GA, Brewer TF, Berkey CS et al. Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. JAMA271(9), 698–702 (1994).
  • Ritz N, Hanekom WA, Robins-Browne R, Britton WJ, Curtis N. Influence of BCG vaccine strain on the immune response and protection against tuberculosis. FEMS Microbiol. Rev.32(5), 821–841 (2008).
  • Brandt L, Feino Cunha J, Weinreich Olsen A et al. Failure of the Mycobacterium bovis BCG vaccine: some species of environmental mycobacteria block multiplication of BCG and induction of protective immunity to tuberculosis. Infect. Immun.70(2), 672–678 (2002).
  • Black GF, Dockrell HM, Crampin AC et al. Patterns and implications of naturally acquired immune responses to environmental and tuberculous mycobacterial antigens in Northern Malawi. J. Infect. Dis.184(3), 322–329 (2001).
  • Palmer CE, Long MW. Effects of infection with atypical mycobacteria on BCG vaccination and tuberculosis. Am. Rev. Respir. Dis.94(4), 553–568 (1966).
  • Weir RE, Black GF, Nazareth B et al. The influence of previous exposure to environmental mycobacteria on the interferon-γ response to bacille Calmette–Guerin vaccination in Southern England and Northern Malawi. Clin. Exp. Immunol.146(3), 390–399 (2006).
  • Elias D, Britton S, Aseffa A, Engers H, Akuffo H. Poor immunogenicity of BCG in helminth infected population is associated with increased in vitro TGF-β production. Vaccine26(31), 3897–3902 (2008).
  • Behr MA, Wilson MA, Gill WP et al. Comparative genomics of BCG vaccines by whole-genome DNA microarray. Science284(5419), 1520–1523 (1999).
  • Brosch R, Gordon SV, Garnier T et al. Genome plasticity of BCG and impact on vaccine efficacy. Proc. Natl Acad. Sci. USA104(13), 5596–5601 (2007).
  • Davids V, Hanekom WA, Mansoor N et al. The effect of bacille Calmette–Guerin vaccine strain and route of administration on induced immune responses in vaccinated infants. J. Infect. Dis.193(4), 531–536 (2006).
  • Hesseling AC, Rabie H, Marais BJ et al. Bacille Calmette–Guerin vaccine-induced disease in HIV-infected and HIV-uninfected children. Clin. Infect. Dis.42(4), 548–558 (2006).
  • Knezevic I, Corbel MJ. WHO discussion on the improvement of the quality control of BCG vaccines. Vaccine24(18), 3874–3877 (2006).
  • Qin L, Gilbert PB, Corey L, McElrath MJ, Self SG. A framework for assessing immunological correlates of protection in vaccine trials. J. Infect. Dis.196(9), 1304–1312 (2007).
  • Beveridge NE, Fletcher HA, Hughes J et al. A comparison of IFNγ detection methods used in tuberculosis vaccine trials. 88(6), 631–640 (2008).
  • Ladel CH, Daugelat S, Kaufmann SH. Immune response to Mycobacterium bovis bacille Calmette–Guerin infection in major histocompatibility complex class I- and II-deficient knock-out mice: contribution of CD4 and CD8 T cells to acquired resistance. Eur. J. Immunol.25(2), 377–384 (1995).
  • Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, Flynn JL. Mice deficient in CD4 T cells have only transiently diminished levels of IFN-γ, yet succumb to tuberculosis. J. Immunol.162(9), 5407–5416 (1999).
  • Saunders BM, Frank AA, Orme IM, Cooper AM. CD4 is required for the development of a protective granulomatous response to pulmonary tuberculosis. Cell. Immunol.216(1–2), 65–72 (2002).
  • Scanga CA, Mohan VP, Yu K et al. Depletion of CD4(+) T cells causes reactivation of murine persistent tuberculosis despite continued expression of interferon gamma and nitric oxide synthase 2. J. Exp. Med.192(3), 347–358 (2000).
  • Cowley SC, Elkins KL. CD4+ T cells mediate IFN-γ-independent control of Mycobacterium tuberculosis infection both in vitro and in vivo. J. Immunol.171(9), 4689–4699 (2003).
  • Feng CG, Britton WJ. CD4+ and CD8+ T cells mediate adoptive immunity to aerosol infection of Mycobacterium bovis bacillus Calmette–Guerin. J. Infect. Dis.181(5), 1846–1849 (2000).
  • Wangoo A, Sparer T, Brown IN et al. Contribution of Th1 and Th2 cells to protection and pathology in experimental models of granulomatous lung disease. J. Immunol.166(5), 3432–3439 (2001).
  • Andersen P, Smedegaard B. CD4(+) T-cell subsets that mediate immunological memory to Mycobacterium tuberculosis infection in mice. Infect. Immun.68(2), 621–629 (2000).
  • Aaron L, Saadoun D, Calatroni I et al. Tuberculosis in HIV-infected patients: a comprehensive review. Clin. Microbiol. Infect.10(5), 388–398 (2004).
  • Jones BE, Young SM, Antoniskis D, Davidson PT, Kramer F, Barnes PF. Relationship of the manifestations of tuberculosis to CD4 cell counts in patients with human immunodeficiency virus infection. Am. Rev. Respir. Dis.148(5), 1292–1297 (1993).
  • Steinman L. A brief history of T(h)17, the first major revision in the T(h)1/T(h)2 hypothesis of T cell-mediated tissue damage. Nat. Med.13(2), 139–145 (2007).
  • Wozniak TM, Ryan AA, Britton WJ. Interleukin-23 restores immunity to Mycobacterium tuberculosis infection in IL-12p40-deficient mice and is not required for the development of IL-17-secreting T cell responses. J. Immunol.177(12), 8684–8692 (2006).
  • Khader SA, Bell GK, Pearl JE 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(4), 369–377 (2007).
  • Khader SA, Cooper AM. IL-23 and IL-17 in tuberculosis. Cytokine41(2), 79–83 (2008).
  • Lockhart E, Green AM, Flynn JL. IL-17 production is dominated by γδ T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J. Immunol.177(7), 4662–4669 (2006).
  • Umemura M, Yahagi A, Hamada S et al. IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette–Guerin infection. J. Immunol.178(6), 3786–3796 (2007).
  • Peng MY, Wang ZH, Yao CY et al. Interleukin 17-producing γ δ T cells increased in patients with active pulmonary tuberculosis. Cell. Mol. Immunol.5(3), 203–208 (2008).
  • Happel KI, Lockhart EA, Mason CM et al. Pulmonary interleukin-23 gene delivery increases local T-cell immunity and controls growth of Mycobacterium tuberculosis in the lungs. Infect. Immun.73(9), 5782–5788 (2005).
  • Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme IM. Disseminated tuberculosis in interferon γ gene-disrupted mice. J. Exp. Med.178(6), 2243–2247 (1993).
  • Flynn JL, Goldstein MM, Triebold KJ, Koller B, Bloom BR. Major histocompatibility complex class I-restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc. Natl Acad. Sci. USA89(24), 12013–12017 (1992).
  • Derrick SC, Repique C, Snoy P, Yang AL, Morris S. Immunization with a DNA vaccine cocktail protects mice lacking CD4 cells against an aerogenic infection with Mycobacterium tuberculosis. Infect. Immun.72(3), 1685–1692 (2004).
  • van Pinxteren LA, Cassidy JP, Smedegaard BH, Agger EM, Andersen P. Control of latent Mycobacterium tuberculosis infection is dependent on CD8 T cells. Eur. J. Immunol.30(12), 3689–3698 (2000).
  • Stenger S, Hanson DA, Teitelbaum R et al. An antimicrobial activity of cytolytic T cells mediated by granulysin. Science282(5386), 121–125 (1998).
  • Boom WH. New TB vaccines: is there a requirement for CD8 T cells? J. Clin. Invest.117(8), 2092–2094 (2007).
  • Hinchey J, Lee S, Jeon BY et al. Enhanced priming of adaptive immunity by a proapoptotic mutant of Mycobacterium tuberculosis. J. Clin. Invest.117(8), 2279–2288 (2007).
  • Hoft DF. Tuberculosis vaccine development: goals, immunological design, and evaluation. Lancet372(9633), 164–175 (2008).
  • Flynn JL, Chan J. Immunology of tuberculosis. Annu. Rev. Immunol.19, 93–129 (2001).
  • Lewinsohn DM, Lewinsohn DA, Grotzke JE. TB vaccines at the turn of the century: insights into immunity to M. tuberculosis and modern approaches for prevention of an ancient disease. Semin. Respir. Infect.18(4), 320–338 (2003).
  • Mehra V, Gong JH, Iyer D et al. Immune response to recombinant mycobacterial proteins in patients with tuberculosis infection and disease. J. Infect. Dis.174(2), 431–434 (1996).
  • Sanchez FO, Rodriguez JI, Agudelo G, Garcia LF. Immune responsiveness and lymphokine production in patients with tuberculosis and healthy controls. Infect. Immun.62(12), 5673–5678 (1994).
  • Newport MJ, Huxley CM, Huston S et al. A mutation in the interferon-γ-receptor gene and susceptibility to mycobacterial infection. N. Engl. J. Med.335(26), 1941–1949 (1996).
  • Dorman SE, Picard C, Lammas D et al. Clinical features of dominant and recessive interferon γ receptor 1 deficiencies. Lancet364(9451), 2113–2121 (2004).
  • Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. An essential role for interferon γ in resistance to Mycobacterium tuberculosis infection. J. Exp. Med.178(6), 2249–2254 (1993).
  • Jouanguy E, Altare F, Lamhamedi S et al. Interferon-γ-receptor deficiency in an infant with fatal bacille Calmette–Guerin infection. N. Engl. J. Med.335(26), 1956–1961 (1996).
  • Bonecini-Almeida MG, Chitale S, Boutsikakis I et al. Induction of in vitro human macrophage anti-Mycobacterium tuberculosis activity: requirement for IFN-γ and primed lymphocytes. J. Immunol.160(9), 4490–4499 (1998).
  • Condos R, Rom WN, Schluger NW. Treatment of multidrug-resistant pulmonary tuberculosis with interferon-γ via aerosol. Lancet349(9064), 1513–1515 (1997).
  • Silver RF, Li Q, Boom WH, Ellner JJ. Lymphocyte-dependent inhibition of growth of virulent Mycobacterium tuberculosis H37Rv within human monocytes: requirement for CD4+ T cells in purified protein derivative-positive, but not in purified protein derivative-negative subjects. J. Immunol.160(5), 2408–2417 (1998).
  • Hope JC, Thom ML, Villarreal-Ramos B, Vordermeier HM, Hewinson RG, Howard CJ. Vaccination of neonatal calves with Mycobacterium bovis BCG induces protection against intranasal challenge with virulent M. bovis. Clin. Exp. Immunol.139(1), 48–56 (2005).
  • Buddle BM, Denis M, Aldwell FE, Martin Vordermeier H, Glyn Hewinson R, Neil Wedlock D. Vaccination of cattle with Mycobacterium bovis BCG by a combination of systemic and oral routes. Tuberculosis (Edinb.)88(6), 595–600 (2008).
  • Soares AP, Scriba TJ, Joseph S et al. Bacillus Calmette–Guerin vaccination of human newborns induces T cells with complex cytokine and phenotypic profiles. J. Immunol.180(5), 3569–3577 (2008).
  • Gardam MA, Keystone EC, Menzies R et al. Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. Lancet Infect. Dis.3(3), 148–155 (2003).
  • Jacobs M, Togbe D, Fremond C et al. Tumor necrosis factor is critical to control tuberculosis infection. Microbes Infect.9(5), 623–628 (2007).
  • Allie N, Alexopoulou L, Quesniaux VJ et al. Protective role of membrane tumour necrosis factor in the host’s resistance to mycobacterial infection. Immunology125(4), 522–534 (2008).
  • Campbell JJ, Murphy KE, Kunkel EJ et al. CCR7 expression and memory T cell diversity in humans. J. Immunol.166(2), 877–884 (2001).
  • Geginat J, Sallusto F, Lanzavecchia A. Cytokine-driven proliferation and differentiation of human naive, central memory, and effector memory CD4+ T cells. J. Exp. Med.194(12), 1711–1719 (2001).
  • Chambers MA, Marshall BG, Wangoo A et al. Differential responses to challenge with live and dead Mycobacterium bovis Bacillus Calmette–Guerin. J. Immunol.158(4), 1742–1748 (1997).
  • Whelan AO, Wright DC, Chambers MA, Singh M, Hewinson RG, Vordermeier HM. Evidence for enhanced central memory priming by live Mycobacterium bovis BCG vaccine in comparison with killed BCG formulations. Vaccine26(2), 166–173 (2008).
  • Buentke E, Mathiot A, Tolaini M, Di Santo J, Zamoyska R, Seddon B. Do CD8 effector cells need IL-7R expression to become resting memory cells? Blood108(6), 1949–1956 (2006).
  • Onoda T, Rahman M, Nara H et al. Human CD4+ central and effector memory T cells produce IL-21: effect on cytokine-driven proliferation of CD4+ T cell subsets. Int. Immunol.19(10), 1191–1199 (2007).
  • Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival. Nat. Rev. Immunol.3(4), 269–279 (2003).
  • Ribeiro-Rodrigues R, Resende Co T, Rojas R et al. A role for CD4+CD25+ T cells in regulation of the immune response during human tuberculosis. Clin. Exp. Immunol.144(1), 25–34 (2006).
  • Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A. Regulatory T cells are expanded in blood and disease sites in patients with tuberculosis. Am. J. Respir. Crit. Care Med.173(7), 803–810 (2006).
  • Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science299(5609), 1057–1061 (2003).
  • Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat. Immunol.4(4), 337–342 (2003).
  • Leng Q, Bentwich Z, Borkow G. Increased TGF-β, Cbl-b and CTLA-4 levels and immunosuppression in association with chronic immune activation. Int. Immunol.18(5), 637–644 (2006).
  • Mahanty S, Mollis SN, Ravichandran M et al. High levels of spontaneous and parasite antigen-driven interleukin-10 production are associated with antigen-specific hyporesponsiveness in human lymphatic filariasis. J. Infect. Dis.173(3), 769–773 (1996).
  • Doetze A, Satoguina J, Burchard G et al. Antigen-specific cellular hyporesponsiveness in a chronic human helminth infection is mediated by T(h)3/T(r)1-type cytokines IL-10 and transforming growth factor-β but not by a T(h)1 to T(h)2 shift. Int. Immunol.12(5), 623–630 (2000).
  • Huygen K, Content J, Denis O et al. Immunogenicity and protective efficacy of a tuberculosis DNA vaccine. Nat. Med.2(8), 893–898 (1996).
  • Skjot RL, Oettinger T, Rosenkrands I et al. Comparative evaluation of low-molecular-mass proteins from Mycobacterium tuberculosis identifies members of the ESAT-6 family as immunodominant T-cell antigens. Infect. Immun.68(1), 214–220 (2000).
  • Dillon DC, Alderson MR, Day CH et al. Molecular characterization and human T-cell responses to a member of a novel Mycobacterium tuberculosis mtb39 gene family. Infect. Immun.67(6), 2941–2950 (1999).
  • Alderson MR, Bement T, Day CH et al. Expression cloning of an immunodominant family of Mycobacterium tuberculosis antigens using human CD4(+) T cells. J. Exp. Med.191(3), 551–560 (2000).
  • Skeiky YA, Ovendale PJ, Jen S et al. T cell expression cloning of a Mycobacterium tuberculosis gene encoding a protective antigen associated with the early control of infection. J. Immunol.165(12), 7140–7149 (2000).
  • Bastian M, Braun T, Bruns H, Rollinghoff M, Stenger S. Mycobacterial lipopeptides elicit CD4+ CTLs in Mycobacterium tuberculosis-infected humans. J. Immunol.180(5), 3436–3446 (2008).
  • Lin MY, Ottenhoff TH. Not to wake a sleeping giant: new insights into host–pathogen interactions identify new targets for vaccination against latent Mycobacterium tuberculosis infection. Biol. Chem.389(5), 497–511 (2008).
  • Locht C, Hougardy JM, Rouanet C, Place S, Mascart F. Heparin-binding hemagglutinin, from an extrapulmonary dissemination factor to a powerful diagnostic and protective antigen against tuberculosis. Tuberculosis (Edinb.)86(3–4), 303–309 (2006).
  • Hougardy JM, Schepers K, Place S et al. Heparin-binding-hemagglutinin-induced IFN-γ release as a diagnostic tool for latent tuberculosis. PLoS ONE2(10), E926 (2007).
  • TB Vaccines for the World 2008 Conference, Atlanta, GA, USA, 9–11 April 2008.
  • Lewin A, Baus D, Kamal E et al. The mycobacterial DNA-binding protein 1 (MDP1) from Mycobacterium bovis BCG influences various growth characteristics. BMC Microbiol.8, 91 (2008).
  • Kana BD, Gordhan BG, Downing KJ et al. The resuscitation-promoting factors of Mycobacterium tuberculosis are required for virulence and resuscitation from dormancy but are collectively dispensable for growth in vitro. Mol. Microbiol.67(3), 672–684 (2008).
  • Russell-Goldman E, Xu J, Wang X, Chan J, Tufariello JM. A Mycobacterium tuberculosis Rpf double-knockout strain exhibits profound defects in reactivation from chronic tuberculosis and innate immunity phenotypes. Infect. Immun.76(9), 4269–4281 (2008).
  • Lin MY, Geluk A, Smith SG et al. Lack of immune responses to Mycobacterium tuberculosis DosR regulon proteins following Mycobacterium bovis BCG vaccination. Infect. Immun.75(7), 3523–3530 (2007).
  • Andersen P. Vaccine strategies against latent tuberculosis infection. Trends Microbiol.15(1), 7–13 (2007).
  • Rustad TR, Harrell MI, Liao R, Sherman DR. The enduring hypoxic response of Mycobacterium tuberculosis. PLoS ONE3(1), E1502 (2008).
  • Weir RE, Gorak-Stolinska P, Floyd S et al. Persistence of the immune response induced by BCG vaccination. BMC Infect. Dis.8, 9 (2008).
  • Goonetilleke NP, McShane H, Hannan CM, Anderson RJ, Brookes RH, Hill AV. Enhanced immunogenicity and protective efficacy against Mycobacterium tuberculosis of bacille Calmette–Guerin vaccine using mucosal administration and boosting with a recombinant modified vaccinia virus Ankara. J. Immunol.171(3), 1602–1609 (2003).
  • McShane H, Pathan AA, Sander CR et al. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat. Med.10(11), 1240–1244 (2004).
  • Karonga Prevention Trial Group. Randomised controlled trial of single BCG, repeated BCG, or combined BCG and killed Mycobacterium leprae vaccine for prevention of leprosy and tuberculosis in Malawi. Lancet348(9019), 17–24 (1996).
  • Rodrigues LC, Pereira SM, Cunha SS et al. Effect of BCG revaccination on incidence of tuberculosis in school-aged children in Brazil: the BCG-REVAC cluster-randomised trial. Lancet366(9493), 1290–1295 (2005).
  • Brandt L, Skeiky YA, Alderson MR et al. The protective effect of the Mycobacterium bovis BCG vaccine is increased by coadministration with the Mycobacterium tuberculosis 72-kilodalton fusion polyprotein Mtb72F in M. tuberculosis-infected guinea pigs. Infect. Immun.72(11), 6622–6632 (2004).
  • Skeiky YA, Alderson MR, Ovendale PJ et al. Differential immune responses and protective efficacy induced by components of a tuberculosis polyprotein vaccine, Mtb72F, delivered as naked DNA or recombinant protein. J. Immunol.172(12), 7618–7628 (2004).
  • Tsenova L, Harbacheuski R, Moreira AL et al. Evaluation of the Mtb72F polyprotein vaccine in a rabbit model of tuberculous meningitis. Infect. Immun.74(4), 2392–2401 (2006).
  • Langermans JA, Doherty TM, Vervenne RA et al. Protection of macaques against Mycobacterium tuberculosis infection by a subunit vaccine based on a fusion protein of antigen 85B and ESAT-6. Vaccine23(21), 2740–2750 (2005).
  • Olsen AW, Williams A, Okkels LM, Hatch G, Andersen P. Protective effect of a tuberculosis subunit vaccine based on a fusion of antigen 85B and ESAT-6 in the aerosol guinea pig model. Infect. Immun.72(10), 6148–6150 (2004).
  • Dietrich J, Aagaard C, Leah R et al. Exchanging ESAT6 with TB10.4 in an Ag85B fusion molecule-based tuberculosis subunit vaccine: efficient protection and ESAT6-based sensitive monitoring of vaccine efficacy. J. Immunol.174(10), 6332–6339 (2005).
  • Agger EM, Rosenkrands I, Olsen AW et al. Protective immunity to tuberculosis with Ag85B-ESAT-6 in a synthetic cationic adjuvant system IC31. Vaccine24(26), 5452–5460 (2006).
  • Christensen D, Foged C, Rosenkrands I, Nielsen HM, Andersen P, Agger EM. Trehalose preserves DDA/TDB liposomes and their adjuvant effect during freeze-drying. Biochim. Biophys. Acta1768(9), 2120–2129 (2007).
  • Williams A, Goonetilleke NP, McShane H et al. Boosting with poxviruses enhances Mycobacterium bovis BCG efficacy against tuberculosis in guinea pigs. Infect. Immun.73(6), 3814–3816 (2005).
  • Radosevic K, Wieland CW, Rodriguez A et al. Protective immune responses to a recombinant adenovirus type 35 tuberculosis vaccine in two mouse strains: CD4 and CD8 T-cell epitope mapping and role of γ interferon. Infect. Immun.75(8), 4105–4115 (2007).
  • Sambandamurthy VK, Wang X, Chen B et al. A pantothenate auxotroph of Mycobacterium tuberculosis is highly attenuated and protects mice against tuberculosis. Nat. Med.8(10), 1171–1174 (2002).
  • Sambandamurthy VK, Derrick SC, Jalapathy KV et al. Long-term protection against tuberculosis following vaccination with a severely attenuated double lysine and pantothenate auxotroph of Mycobacterium tuberculosis. Infect. Immun.73(2), 1196–1203 (2005).
  • Sambandamurthy VK, Derrick SC, Hsu T et al.Mycobacterium tuberculosis DRD1 ΔpanCD: a safe and limited replicating mutant strain that protects immunocompetent and immunocompromised mice against experimental tuberculosis. Vaccine24(37–39), 6309–6320 (2006).
  • Sampson SL, Dascher CC, Sambandamurthy VK et al. Protection elicited by a double leucine and pantothenate auxotroph of Mycobacterium tuberculosis in guinea pigs. Infect. Immun.72(5), 3031–3037 (2004).
  • Martin C, Williams A, Hernandez-Pando R 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. Vaccine24(17), 3408–3419 (2006).
  • Williams A, Hatch GJ, Clark SO et al. Evaluation of vaccines in the EU TB Vaccine cluster using a guinea pig aerosol infection model of tuberculosis. Tuberculosis (Edinb.)85(1–2), 29–38 (2005).
  • Aguilar D, Infante E, Martin C, Gormley E, Gicquel B, Hernandez Pando R. Immunological responses and protective immunity against tuberculosis conferred by vaccination of BALB/C mice with the attenuated Mycobacterium tuberculosis (phoP) SO2 strain. Clin. Exp. Immunol.147(2), 330–338 (2007).
  • Grode L, Seiler P, Baumann S et al. Increased vaccine efficacy against tuberculosis of recombinant Mycobacterium bovis bacille Calmette–Guerin mutants that secrete listeriolysin. J. Clin. Invest.115(9), 2472–2479 (2005).
  • Horwitz MA, 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(4), 1672–1679 (2003).
  • Skeiky YA, Sadoff JC. Advances in tuberculosis vaccine strategies. Nat. Rev. Microbiol.4(6), 469–476 (2006).
  • Xu Y, Zhu B, Wang Q et al. Recombinant BCG coexpressing Ag85B, ESAT-6 and mouse-IFN-γ confers effective protection against Mycobacterium tuberculosis in C57BL/6 mice. FEMS Immunol. Med. Microbiol.51(3), 480–487 (2007).
  • Tang C, Yamada H, Shibata K et al. Efficacy of recombinant bacille Calmette–Guerin vaccine secreting interleukin-15/antigen 85B fusion protein in providing protection against Mycobacterium tuberculosis. J. Infect. Dis.197(9), 1263–1274 (2008).
  • Wang JL, Qie YQ, Zhu BD et al. Evaluation of a recombinant BCG expressing antigen Ag85B and PPE protein Rv3425 from DNA segment RD11 of Mycobacterium tuberculosis in C57BL/6 mice. Med. Microbiol. Immunol. (2008).
  • Edwards KM, Cynamon MH, Voladri RK et al. Iron-cofactored superoxide dismutase inhibits host responses to Mycobacterium tuberculosis. Am. J. Respir. Crit. Care Med.164(12), 2213–2219 (2001).
  • Sheridan JP, Marsters SA, Pitti RM et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science277(5327), 818–821 (1997).
  • Chattergoon MA, Kim JJ, Yang JS et al. Targeted antigen delivery to antigen-presenting cells including dendritic cells by engineered Fas-mediated apoptosis. Nat. Biotechnol.18(9), 974–979 (2000).
  • Ziv E, Daley CL, Blower S. Potential public health impact of new tuberculosis vaccines. Emerg. Infect. Dis.10(9), 1529–1535 (2004).
  • Rubakova E, Petrovskaya S, Pichugin A et al. Specificity and efficacy of dendritic cell-based vaccination against tuberculosis with complex mycobacterial antigens in a mouse model. Tuberculosis (Edinb.)87(2), 134–144 (2007).
  • Lore K, Adams WC, Havenga MJ et al. Myeloid and plasmacytoid dendritic cells are susceptible to recombinant adenovirus vectors and stimulate polyfunctional memory T cell responses. J. Immunol.179(3), 1721–1729 (2007).
  • Bhatnagar S, Shinagawa K, Castellino FJ, Schorey JS. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood110(9), 3234–3244 (2007).
  • Giri PK, Schorey JS. Exosomes derived from M. Bovis BCG infected macrophages activate antigen-specific CD4+ and CD8+ T cells in vitro and in vivo. PLoS ONE3(6), E2461 (2008).
  • Okada M, Kita Y, Nakajima T et al. Evaluation of a novel vaccine (HVJ-liposome/HSP65 DNA+IL-12 DNA) against tuberculosis using the cynomolgus monkey model of TB. Vaccine25(16), 2990–2993 (2007).
  • Sadoff J. Aeras adeno 35 vectored and nucleocapsid vectored TB vaccines. Presented at: Viral Vector Vaccines Conference. Hinxton, Cambridge, UK 28–30 September 2008.
  • Waddell RD, Chintu C, Lein AD et al. Safety and immunogenicity of a five-dose series of inactivated Mycobacterium vaccae vaccination for the prevention of HIV-associated tuberculosis. Clin. Infect. Dis.30(Suppl. 3), S309–S315 (2000).
  • Cardona PJ, Amat I, Gordillo S et al. Immunotherapy with fragmented Mycobacterium tuberculosis cells increases the effectiveness of chemotherapy against a chronical infection in a murine model of tuberculosis. Vaccine23(11), 1393–1398 (2005).
  • Guirado E, Gil O, Caceres N, Singh M, Vilaplana C, Cardona PJ. Induction of a specific strong polyantigenic cellular immune response after short-term chemotherapy controls bacillary reactivation in murine and guinea pig experimental models of tuberculosis. Clin. Vaccine Immunol.15(8), 1229–1237 (2008).
  • Santosuosso M, Zhang X, McCormick S, Wang J, Hitt M, Xing Z. Mechanisms of mucosal and parenteral tuberculosis vaccinations: adenoviral-based mucosal immunization preferentially elicits sustained accumulation of immune protective CD4 and CD8 T cells within the airway lumen. J. Immunol.174(12), 7986–7994 (2005).
  • Santosuosso M, McCormick S, Roediger E et al. Mucosal luminal manipulation of T cell geography switches on protective efficacy by otherwise ineffective parenteral genetic immunization. J. Immunol.178(4), 2387–2395 (2007).
  • Garcia-Contreras L, Wong YL, Muttil P et al. Immunization by a bacterial aerosol. Proc. Natl Acad. Sci. USA105(12), 4656–4660 (2008).
  • Freidag BL, Melton GB, Collins F et al. CpG oligodeoxynucleotides and interleukin-12 improve the efficacy of Mycobacterium bovis BCG vaccination in mice challenged with M. tuberculosis. Infect. Immun.68(5), 2948–2953 (2000).
  • Palendira U, Bean AG, Feng CG, Britton WJ. Lymphocyte recruitment and protective efficacy against pulmonary mycobacterial infection are independent of the route of prior Mycobacterium bovis BCG immunization. Infect. Immun.70(3), 1410–1416 (2002).
  • Dietrich J, Andersen C, Rappuoli R, Doherty TM, Jensen CG, Andersen P. Mucosal administration of Ag85B-ESAT-6 protects against infection with Mycobacterium tuberculosis and boosts prior bacillus Calmette–Guerin immunity. J. Immunol.177(9), 6353–6360 (2006).
  • D’Souza S, Rosseels V, Denis O et al. Improved tuberculosis DNA vaccines by formulation in cationic lipids. Infect. Immun.70(7), 3681–3688 (2002).
  • Xing Z, Santosuosso M, McCormick S et al. Recent advances in the development of adenovirus- and poxvirus-vectored tuberculosis vaccines. Curr. Gene Ther.5(5), 485–492 (2005).
  • Casimiro DR, Chen L, Fu TM et al. Comparative immunogenicity in rhesus monkeys of DNA plasmid, recombinant vaccinia virus, and replication-defective adenovirus vectors expressing a human immunodeficiency virus type 1 gag gene. J. Virol.77(11), 6305–6313 (2003).
  • Wang J, Thorson L, Stokes RW et al. Single mucosal, but not parenteral, immunization with recombinant adenoviral-based vaccine provides potent protection from pulmonary tuberculosis. J. Immunol.173(10), 6357–6365 (2004).
  • Santosuosso M, McCormick S, Zhang X, Zganiacz A, Xing Z. Intranasal boosting with an adenovirus-vectored vaccine markedly enhances protection by parenteral Mycobacterium bovis BCG immunization against pulmonary tuberculosis. Infect. Immun.74(8), 4634–4643 (2006).
  • Forbes EK, Sander C, Ronan E et al. The presence of multifunctional, high-cytokine-producing Th1 cells in the lung, but not spleen, correlates with protection against Mycobacterium tuberculosis aerosol challenge in mice. J. Immunol.181(7), 4955–4964 (2008).
  • Vordermeier HM, Huygen K, Singh M, Hewinson RG, Xing Z. Immune responses induced in cattle by vaccination with a recombinant adenovirus expressing mycobacterial antigen 85A and Mycobacterium bovis BCG. Infect. Immun.74(2), 1416–1418 (2006).
  • Kohama H, Umemura M, Okamoto Y et al. Mucosal immunization with recombinant heparin-binding haemagglutinin adhesin suppresses extrapulmonary dissemination of Mycobacterium bovis bacillus Calmette–Guerin (BCG) in infected mice. Vaccine26(7), 924–932 (2008).
  • Fourie PB, Germishuizen WA, Wong YL, Edwards DA. Spray drying TB vaccines for pulmonary administration. Expert Opin. Biol. Ther.8(7), 857–863 (2008).
  • Hanekom WA. The immune response to BCG vaccination of newborns. Ann. NY Acad. Sci.1062, 69–78 (2005).
  • Worku S, Hoft DF. Differential effects of control and antigen-specific T cells on intracellular mycobacterial growth. Infect. Immun.71(4), 1763–1773 (2003).
  • Cheng SH, Walker KB, Lowrie DB et al. Monocyte antimycobacterial activity before and after Mycobacterium bovis BCG vaccination in Chingleput, India, and London, United Kingdom. Infect. Immun.61(10), 4501–4503 (1993).
  • Cheon SH, Kampmann B, Hise AG et al. Bactericidal activity in whole blood as a potential surrogate marker of immunity after vaccination against tuberculosis. Clin. Diagn. Lab. Immunol.9(4), 901–907 (2002).
  • Wallis RS, Palaci M, Vinhas S et al. A whole blood bactericidal assay for tuberculosis. J. Infect. Dis.183(8), 1300–1303 (2001).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.