R. Coler, S. Baldwin, N. Shaverdian, S. Bertholet, S. Reed et al., A synthetic adjuvant to enhance and expand immune responses to influenza vaccines):e13677. Epub Mechanisms of T H 1 Induction by, PloS one, vol.5, issue.10, pp.10-1371, 2010.
DOI : 10.1371/journal.pone.0013677

URL : https://doi.org/10.1371/journal.pone.0013677

J. Kurche, C. Haluszczak, M. Ja, P. Sanchez, R. Kedl et al., Type I IFN-Dependent T Cell Activation Is Mediated by IFN-Dependent Dendritic Cell OX40 Ligand Expression and Is Independent of T Cell IFNR Expression, The Journal of Immunology, vol.188, issue.2, pp.585-9310, 2012.
DOI : 10.4049/jimmunol.1102550

J. J. Taylor, K. A. Pape, and M. K. Jenkins, A germinal center???independent pathway generates unswitched memory B cells early in the primary response, The Journal of Experimental Medicine, vol.89, issue.3, pp.597-606, 2012.
DOI : 10.1084/jem.176.1.47

U. H. Hacohen and . Von-andrian, B Cell Maintenance of Subcapsular Sinus Macrophages Protects against a Fatal Viral Infection Independent of Adaptive Immunity, Immunity, vol.36, pp.415-426, 2012.

T. Junt, E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang et al., Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells, Nature, vol.9, issue.7166, pp.110-114, 2007.
DOI : 10.1038/nature06287

S. Crotty, ), Annual Review of Immunology, vol.29, issue.1, pp.621-663, 2011.
DOI : 10.1146/annurev-immunol-031210-101400

S. G. Reed, M. T. Orr, and C. B. Fox, Key roles of adjuvants in modern vaccines, Nature Medicine, vol.491, issue.12, pp.1597-608, 2013.
DOI : 10.1074/jbc.M112.347179

M. Coler, S. G. Friede, T. S. Reed, and . Vedvick, Physicochemical characterization and biological activity of synthetic TLR4 agonist formulations, Colloids Surf. B. Biointerfaces, vol.75, pp.123-155, 2010.

S. G. Carter, M. S. Reed, and . Duthie, Squalene emulsion potentiates the adjuvant activity of the TLR4 agonist, GLA, via inflammatory caspases, IL-18, and IFN-?, Eur. J. Immunol, vol.45, pp.407-417, 2015.

S. G. Vedvick, R. N. Reed, and . Coler, Adjuvant formulation structure and composition are 90, 2013.

O. Donnell, A. L. Desbien, S. G. Reed, and R. N. Coler, MyD88 and TRIF synergistic interaction is required for TH1-cell polarization with a synthetic TLR4 agonist adjuvant, Eur. J. Immunol, vol.43, pp.2398-408, 2013.

T. The, . Agonist-vaccine-adjuvant, and . Gla-se, Requires Canonical and Atypical Mechanisms of Action for TH1 Induction, PLoS One, vol.11, p.146372

K. Pape, J. J. Taylor, R. W. Maul, P. J. Gearhart, and M. K. Jenkins, Different B Cell Populations Mediate Early and Late Memory During an Endogenous Immune Response, Science, vol.357, issue.19, pp.1203-1210, 2011.
DOI : 10.1056/NEJMoa066092

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3993090/pdf

K. Hayakawa, R. Ishii, K. Yamasaki, T. Kishimoto, and R. R. Hardy, Isolation of highaffinity memory B cells: phycoerythrin as a probe for antigen-binding cells, Proc. Natl. Acad. Sci, 1987.

S. G. Coler and . Reed, Protection of mice from Mycobacterium tuberculosis by, 2011.

W. Kastenmüller and P. Torabi-parizi, A Spatially-Organized Multicellular Innate Immune Response in Lymph Nodes Limits Systemic Pathogen Spread, Cell, vol.150, issue.6, pp.1235-1248, 2012.
DOI : 10.1016/j.cell.2012.07.021

S. Bagnoli and . Bertholet, Oil-in-Water Emulsion MF59 Increases Germinal Center B Cell Differentiation and Persistence in Response to Vaccination, J. Immunol, 2015.

G. Reed, Adjuvant solution for pandemic influenza vaccine production, Proc. Natl. Acad. Sci, p.91, 2012.

A. Pantel, C. Cheong, D. Dandamudi, E. Shrestha, S. Mehandru et al., A new synthetic TLR4 agonist, GLA, allows dendritic cells targeted with antigen to elicit Th1 T-cell immunity in vivo, European Journal of Immunology, vol.30, issue.1, pp.101-109, 2012.
DOI : 10.1016/j.it.2008.09.006

. Haberman, Germinal center B cell and T follicular helper cell development initiates in the interfollicular zone, Immunity, vol.34, pp.947-60, 2011.

. Garside, Th1 and Th2 CD4+ T cells provide help for B cell clonal expansion and antibody synthesis in a similar manner in vivo, J. Immunol, vol.165, pp.3136-3180, 2000.

J. J. Andrian, T. R. Moon, A. D. Mempel, and . Luster, CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation, Immunity, vol.37, pp.1091-103, 2012.

C. S. Ma, E. K. Deenick, M. Batten, and S. G. Tangye, The origins, function, and regulation of T follicular helper cells, The Journal of Experimental Medicine, vol.13, issue.7, pp.1241-1253, 2012.
DOI : 10.1084/jem.20091777

J. Hagar, D. Powell, Y. Aachoui, R. K. Ernst, and E. Miao, Cytoplasmic LPS Activates Caspase-11: Implications in TLR4-Independent Endotoxic Shock, Science, vol.31, issue.11, pp.1250-1253, 2013.
DOI : 10.1016/j.exphem.2003.07.005

J. Shi, Y. Zhao, Y. Wang, W. Gao, J. Ding et al., Inflammatory caspases are innate immune receptors for intracellular LPS, Nature, vol.24, issue.7521, 2014.
DOI : 10.1128/IAI.70.6.3264-3270.2002

O. Hagan, D. T. , G. S. Ott, E. D. Gregorio, and . Seubert, The mechanism of action of MF59 -an innately attractive adjuvant formulation, Vaccine, vol.30, pp.4341-4349, 2012.

D. Oro, M. M. Giuliani, M. Pallaoro, M. Pizza, D. T. O-'hagan et al., Adjuvanticity of the oil-in-water emulsion MF59 is independent of Nlrp3 inflammasome but requires the adaptor protein MyD88, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.11169-74, 2011.

F. D. Rappuoli, E. Virgilio, C. De-gregorio, A. Montecucco, and . Seubert, The adjuvant MF59 induces ATP release from muscle that potentiates response to vaccination, Proc. Natl. Acad. Sci. U, 2013.

C. B. Fox, S. L. Baldwin, M. S. Duthie, S. G. Reed, and T. S. Vedvick, Immunomodulatory and physical effects of oil composition in vaccine adjuvant emulsions, Vaccine, vol.29, issue.51, pp.9563-72, 2011.
DOI : 10.1016/j.vaccine.2011.08.089

C. B. Fox, S. L. Baldwin, T. S. Vedvick, E. Angov, and S. G. Reed, ABSTRACT, Clinical and Vaccine Immunology, vol.19, issue.10, pp.1633-1673, 2012.
DOI : 10.1128/CVI.00235-12

M. Roederer, J. L. Nozzi, and M. C. Nason, SPICE: exploration and analysis of postcytometric complex multivariate datasets, Cytometry. A, vol.79, pp.167-74, 2011.

R. N. Coler, S. L. Baldwin, N. Shaverdian, S. Bertholet, S. J. Reed et al., A Synthetic Adjuvant to Enhance and Expand Immune Responses to Influenza Vaccines, PLoS ONE, vol.20, issue.10, p.13677, 2010.
DOI : 10.1371/journal.pone.0013677.s004

URL : https://doi.org/10.1371/journal.pone.0013677

F. Sallusto, A. Lanzavecchia, K. Araki, and R. Ahmed, From Vaccines to Memory and Back, Immunity, vol.33, issue.4, pp.451-463, 2010.
DOI : 10.1016/j.immuni.2010.10.008

URL : https://doi.org/10.1016/j.immuni.2010.10.008

G. M. Urdahl, D. L. Winslow, and . Woodland, Distinct functions of antigen-specific CD4 T cells during murine Mycobacterium tuberculosis infection, Proceedings of the National Academy of Sciences of the United States of America, vol.107, pp.19408-19413, 2010.

H. D. Marshall, A. Chandele, Y. W. Jung, H. Meng, A. C. Poholek et al., Differential Expression of Ly6C and T-bet Distinguish Effector and Memory Th1 CD4+ Cell Properties during Viral Infection, Differential Expression of Ly6C and T-bet Distinguish Effector and Memory Th1 CD4 + Cell Properties during Viral Infection, pp.633-646, 2011.
DOI : 10.1016/j.immuni.2011.08.016

I. Misumi and J. K. Whitmire, B Cell Depletion Curtails CD4+ T Cell Memory and Reduces Protection against Disseminating Virus Infection, The Journal of Immunology, vol.192, issue.4, pp.1597-1608, 1950.
DOI : 10.4049/jimmunol.1302661

URL : http://www.jimmunol.org/content/jimmunol/192/4/1597.full.pdf

. Ahmed, Requirement of B cells for generating CD4+ T cell memory, J Immunol, vol.182, pp.1868-1876, 2009.

G. Matsuzaki, H. M. Vordermeier, A. Hashimoto, K. Nomoto, and J. Ivanyi, The Role of B Cells in the Establishment of T Cell Response in Mice Infected with an Intracellular Bacteria, Listeria monocytogenes, Cellular Immunology, vol.194, issue.2, pp.178-185, 1999.
DOI : 10.1006/cimm.1999.1503

K. L. Elkins, C. M. Bosio, T. R. Rhinehart, and -. , Importance of B cells, but not specific antibodies, in primary and secondary protective immunity to the intracellular bacterium Francisella tularensis live vaccine strain, Infect Immun, vol.67, pp.6002-6007, 1999.

H. M. Vordermeier, N. Venkataprasad, D. P. Harris, and J. Ivanyi, Increase of tuberculous infection in the organs of B cell-deficient mice, Clinical and Experimental Immunology, vol.106, issue.2, pp.312-316, 1996.
DOI : 10.1046/j.1365-2249.1996.d01-845.x

D. J. Dilillo, K. Yanaba, and T. F. Tedder, B Cells Are Required for Optimal CD4+ and CD8+ T Cell Tumor Immunity: Therapeutic B Cell Depletion Enhances B16 Melanoma Growth in Mice, The Journal of Immunology, vol.184, issue.7, pp.4006-4016, 2010.
DOI : 10.4049/jimmunol.0903009

A. Crawford, M. Macleod, T. Schumacher, L. Corlett, and D. Gray, Primary T Cell Expansion and Differentiation In Vivo Requires Antigen Presentation by B Cells, The Journal of Immunology, vol.176, issue.6, pp.3498-3506, 2006.
DOI : 10.4049/jimmunol.176.6.3498

B. Morales-aza, S. J. Glennie, T. P. Garcez, V. Davenport, S. L. Johnston et al., Impaired maintenance of naturally acquired T-cell memory to the meningococcus in patients with B-cell immunodeficiency, Blood, vol.113, issue.18, pp.4206-4212, 2009.
DOI : 10.1182/blood-2008-08-171587

R. Nayak, G. Lal, and M. S. Shaila, Perpetuation of immunological memory: role of serum antibodies and accessory cells, Microbes and Infection, vol.7, issue.11-12, pp.1276-1283, 2005.
DOI : 10.1016/j.micinf.2005.06.001

M. Anderton, A. Bar-or, S. Fillatreau, and D. Gray, B cell depletion therapy ameliorates autoimmune disease through ablation of IL-6-producing B cells, J Exp Med, vol.209, pp.1001-1010, 2012.

M. Mohrs, T. Mohrs, F. E. Randall, and . Lund, Cytokine-producing effector B cells regulate type 2 immunity to H. polygyrus, Immunity, vol.30, pp.421-433, 2009.

B. Leon, A. Ballesteros-tato, J. L. Browning, R. Dunn, T. D. Randall et al., Regulation of TH2 development by CXCR5+ dendritic cells and lymphotoxin-expressing B cells, Nature Immunology, vol.180, issue.7, pp.681-690, 2012.
DOI : 10.4049/jimmunol.180.7.4994

. Brombacher, IL-4Ralpha-associated antigen processing by B cells promotes immunity in Nippostrongylus brasiliensis infection, PLoS Pathog, vol.9, p.1003662, 2013.

T. A. Barr, S. Brown, P. Mastroeni, and D. Gray, TLR and B Cell Receptor Signals to B Cells Differentially Program Primary and Memory Th1 Responses to Salmonella enterica, The Journal of Immunology, vol.185, issue.5, pp.2783-2789, 2010.
DOI : 10.4049/jimmunol.1001431

URL : http://www.jimmunol.org/content/jimmunol/185/5/2783.full.pdf

J. O. Shaverdian, A. L. Donnell, S. G. Desbien, R. N. Reed, and . Coler, MyD88 and TRIF synergistic interaction is required for TH1-cell polarization with a synthetic TLR4 agonist adjuvant, European journal of immunology, vol.43, issue.120, pp.2398-2408, 2013.

C. B. Laurance, S. G. Fox, R. N. Reed, T. S. Coler, and . Vedvick, Elimination of the coldchain dependence of a nanoemulsion adjuvanted vaccine against tuberculosis by lyophilization, Journal of controlled release : official journal of the Controlled Release Society, vol.177, pp.20-26, 2013.

T. S. Orme, S. L. Vedvick, R. N. Baldwin, S. G. Coler, and . Reed, A defined tuberculosis vaccine candidate boosts BCG and protects against multidrug-resistant Mycobacterium tuberculosis, Science translational medicine, vol.2, pp.53-74, 2010.

S. G. Vedvick, R. N. Reed, and . Coler, Adjuvant formulation structure and composition are critical for the development of an effective vaccine against tuberculosis, Journal of controlled release : official journal of the Controlled Release Society, vol.172, pp.190-200, 2013.

S. L. Constant, B lymphocytes as antigen-presenting cells for CD4+ T cell priming in vivo, J Immunol, vol.162, pp.5695-5703, 1999.

M. M. Epstein, F. Di-rosa, D. Jankovic, A. Sher, and P. Matzinger, Successful T cell priming in B cell-deficient mice, Journal of Experimental Medicine, vol.182, issue.4, pp.915-922, 1995.
DOI : 10.1084/jem.182.4.915

G. S. Williams, A. Oxenius, H. Hengartner, C. Benoist, and D. Mathis, CD4+ T cell responses in mice lacking MHC class II molecules specifically on B cells, European Journal of Immunology, vol.133, issue.11, pp.3763-3772, 1998.
DOI : 10.1038/353622a0

S. Constant, N. Schweitzer, J. West, P. Ranney, and K. Bottomly, B lymphocytes can be competent antigen-presenting cells for priming CD4+ T cells to protein antigens in vivo, J Immunol, vol.155, pp.3734-3741, 1995.

S. B. Mollo, A. J. Zajac, and L. E. Harrington, Temporal Requirements for B Cells in the Establishment of CD4 T Cell Memory, The Journal of Immunology, vol.191, issue.12, pp.6052-6059, 1950.
DOI : 10.4049/jimmunol.1302033

R. Foa, Changes in the Treatment Landscape for Chronic Lymphoid Leukemia, New England Journal of Medicine, vol.371, issue.3, pp.273-274, 2014.
DOI : 10.1056/NEJMe1405766

R. Stasi, A. Pagano, E. Stipa, and S. Amadori, Rituximab chimeric anti-CD20 monoclonal antibody treatment for adults with chronic idiopathic thrombocytopenic purpura, Blood, vol.98, issue.4, pp.952-957, 2001.
DOI : 10.1182/blood.V98.4.952

S. L. Hauser, E. Waubant, D. L. Arnold, T. Vollmer, J. Antel et al., B-Cell Depletion with Rituximab in Relapsing???Remitting Multiple Sclerosis, New England Journal of Medicine, vol.358, issue.7, pp.676-688, 2008.
DOI : 10.1056/NEJMoa0706383

A. H. Song and . Cross, Rituximab add-on therapy for breakthrough relapsing multiple sclerosis: a 52-week phase II trial, Neurology, vol.74, pp.1860-1867, 2010.

J. A. Gomez-puerta, L. F. Quintana, J. H. Stone, M. Ramos-casals, and X. Bosch, B-cell depleting agents for ANCA vasculitides: A new therapeutic approach, Autoimmunity Reviews, vol.11, issue.9, pp.646-652, 2012.
DOI : 10.1016/j.autrev.2011.11.007

S. L. Goldberg, A. L. Pecora, R. S. Alter, M. S. Kroll, S. D. Rowley et al., Unusual viral infections (progressive multifocal leukoencephalopathy and cytomegalovirus disease) after high-dose chemotherapy with autologous blood stem cell rescue and peritransplantation rituximab, Blood, vol.99, issue.4, pp.1486-1488, 2002.
DOI : 10.1182/blood.V99.4.1486

URL : http://www.bloodjournal.org/content/bloodjournal/99/4/1486.full.pdf

F. Suzan, M. Ammor, and V. Ribrag, Fatal Reactivation of Cytomegalovirus Infection after Use of Rituximab for a Post-Transplantation Lymphoproliferative Disorder, New England Journal of Medicine, vol.345, issue.13, p.1000, 2001.
DOI : 10.1056/NEJM200109273451315

Y. Tsutsumi, Y. Yamamoto, S. Ito, H. Ohigashi, S. Shiratori et al., Hepatitis B virus reactivation with a rituximab-containing regimen, World Journal of Hepatology, vol.7, issue.21, pp.2344-2351, 2015.
DOI : 10.1056/NEJM200501203520324

E. M. Salvana, R. A. Salata-gea-banacloche, J. C. , and G. A. Weinberg, Infectious complications associated with monoclonal antibodies and related small molecules Table of Contents Monoclonal antibody therapeutics and risk for infection, Clin Microbiol Rev Pediatr Infect Dis J, vol.22, issue.26, pp.274-290, 2007.

S. N. Liossis and P. P. Sfikakis, Rituximab-induced B cell depletion in autoimmune diseases: Potential effects on T cells, Clinical Immunology, vol.127, issue.3, pp.280-285, 2008.
DOI : 10.1016/j.clim.2008.01.011

M. J. Leandro and I. De-la-torre, Translational Mini-Review Series on B Cell-Directed Therapies: The pathogenic role of B cells in autoantibody-associated autoimmune diseases - lessons from B cell-depletion therapy, Clinical & Experimental Immunology, vol.140, issue.14, pp.191-197, 2009.
DOI : 10.4049/jimmunol.174.7.4389

M. S. Asano and R. Ahmed, CD8 T cell memory in B cell-deficient mice, Journal of Experimental Medicine, vol.183, issue.5, pp.2165-2174, 1996.
DOI : 10.1084/jem.183.5.2165

M. T. Crowley, C. R. Reilly, and D. Lo, Influence of lymphocytes on the presence and organization of dendritic cell subsets in the spleen, J Immunol, vol.163, pp.4894-4900, 1999.

L. Teixeira, R. M. Bozzacco, M. P. Steinman, and . Longhi, A new synthetic TLR4 agonist, GLA, allows dendritic cells targeted with antigen to elicit Th1 T-cell immunity in vivo, European journal of immunology, vol.42, pp.101-109, 2012.

A. G. Nerlich, C. J. Haas, A. Zink, U. Szeimies, and H. G. Hagedorn, Molecular evidence for tuberculosis in an ancient Egyptian mummy, The Lancet, vol.350, issue.9088, p.1404, 1997.
DOI : 10.1016/S0140-6736(05)65185-9

T. M. Daniel, The history of tuberculosis, Respiratory Medicine, vol.100, issue.11, pp.1862-1870, 2006.
DOI : 10.1016/j.rmed.2006.08.006

L. J. Abu-raddad, L. Sabatelli, J. T. Achterberg, J. D. Sugimoto, I. M. Longini et al., Epidemiological benefits of more-effective tuberculosis vaccines, drugs, and diagnostics, Proceedings of the National Academy of Sciences, vol.121, issue.6, pp.13980-13985, 2009.
DOI : 10.2174/156652407780598575

R. N. Coler, S. Bertholet, M. Moutaftsi, J. Guderian, H. P. Windish et al., Development and Characterization of Synthetic Glucopyranosyl Lipid Adjuvant System as a Vaccine Adjuvant, PLoS ONE, vol.203, issue.1, pp.16333-16333, 2011.
DOI : 10.1371/journal.pone.0016333.s002

S. Bertholet, G. C. Ireton, D. J. Ordway, H. P. Windish, S. O. Pine et al., A Defined Tuberculosis Vaccine Candidate Boosts BCG and Protects Against Multidrug-Resistant Mycobacterium tuberculosis, Science Translational Medicine, vol.328, issue.16, pp.53-74, 2010.
DOI : 10.1056/NEJM199304223281601

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3110937/pdf

A. L. Desbien, S. J. Reed, H. R. Bailor, N. Dubois-cauwelaert, J. D. Laurance et al., Squalene emulsion potentiates the adjuvant activity of the TLR4 agonist, GLA, via inflammatory caspases, IL-18, and IFN-??, Squalene emulsion potentiates the adjuvant activity of the TLR4 agonist, GLA, via inflammatory caspases, IL-18, and IFN-gamma, pp.407-417, 2015.
DOI : 10.1038/nmeth.2089

A. S. Roguin and A. I. Zumla, the man behind the stethoscope Center for Disease Control and Prevention Reported Tuberculosis in the United States 2013. Department of Health and Human Services, CDC, Atlanta, GA: U.S. 16. European Centre for Disease Prevention and Control., and World Health Organization. Regional Office for Europe. 2014. Tuberculosis surveillance and monitoring in Europe 2014 : surveillance report. European Centre for Disease Prevention and Control (ECDC), Stockholm. 17, of HIV/AIDS., and World Health Organization, pp.1781-1826, 2006.

P. Diel, F. Ravn, P. J. Cobelens, B. Cardona, I. Kan et al., LTBI: latent tuberculosis infection or lasting immune responses to M. tuberculosis? A TBNET consensus statement Exogenous reinfection as a cause of recurrent tuberculosis after curative treatment, Eur Respir J N Engl J Med, vol.33, issue.341, pp.956-973, 1999.

E. L. Korenromp, F. Scano, B. G. Williams, C. Dye, and P. Nunn, Effects of Human Immunodeficiency Virus Infection on Recurrence of Tuberculosis after Rifampin???Based Treatment: An Analytical Review, Clinical Infectious Diseases, vol.6, issue.1, pp.101-112, 2003.
DOI : 10.1016/S0035-9203(00)90306-7

E. Vynnycky and P. E. Fine, The natural history of tuberculosis: the implications of age-dependent risks of disease and the role of reinfection, Epidemiology and Infection, vol.119, issue.2, pp.183-201, 1997.
DOI : 10.1017/S0950268897007917

P. Godfrey-faussett, W. Githui, B. Batchelor, R. Brindle, J. Paul et al., Recurrence of HIV-related tuberculosis in an endemic area may be due to relapse or reinfection, Tubercle and Lung Disease, vol.75, issue.3, pp.199-202, 1994.
DOI : 10.1016/0962-8479(94)90008-6

A. Seidler, A. Nienhaus, and R. Diel, The transmission of tuberculosis in the light of new molecular biological approaches, Occupational and Environmental Medicine, vol.61, issue.2, pp.96-102, 2004.
DOI : 10.1136/oem.2003.008573

R. E. Chaisson and G. J. Churchyard, Recurrent Tuberculosis: Relapse, Reinfection, and HIV, The Journal of Infectious Diseases, vol.201, issue.5, pp.653-655, 2010.
DOI : 10.1086/650531

URL : https://academic.oup.com/jid/article-pdf/201/5/653/2562483/201-5-653.pdf

E. L. Corbett, T. Bandason, Y. B. Cheung, B. Makamure, E. Dauya et al., Prevalent infectious tuberculosis in Harare, Zimbabwe: burden, risk factors and implications for control In search of a new paradigm for protective immunity to TB, Int J Tuberc Lung Dis Nat Rev Microbiol, vol.13, issue.12, pp.1231-1237, 2009.

M. Petrofsky, L. E. Bermudez, W. H. , D. H. Canaday, S. A. Fulton et al., Neutrophils from Mycobacterium avium-infected mice produce TNF-alpha, IL-12, and IL-1 beta and have a putative role in early host response Human immunity to M. tuberculosis: T cell subsets and antigen processing, Clin Immunol Tuberculosis (Edinb), vol.91, issue.83, pp.354-358, 1999.

J. L. Flynn and J. Chan, Tuberculosis: latency and reactivation Mycobacterium tuberculosis and the macrophage: maintaining a balance, Infect Immun Pieters, J. Cell Host Microbe, vol.69, issue.3, pp.4195-4201, 2001.

S. Sturgill-koszycki, P. H. Schlesinger, P. Chakraborty, P. L. Haddix, H. L. Collins et al., Lack of acidification in Mycobacterium phagosomes produced by exclusion of the vesicular proton-ATPase, Science, vol.263, issue.5147, pp.678-681, 1994.
DOI : 10.1126/science.8303277

B. H. Miller, R. A. Fratti, J. F. Poschet, G. S. Timmins, S. S. Master et al., Mycobacteria Inhibit Nitric Oxide Synthase Recruitment to Phagosomes during Macrophage Infection, Infection and Immunity, vol.72, issue.5, pp.2872-2878, 2004.
DOI : 10.1128/IAI.72.5.2872-2878.2004

URL : http://iai.asm.org/content/72/5/2872.full.pdf

K. H. Darwin, S. Ehrt, J. C. Gutierrez-ramos, N. Weich, and C. F. Nathan, The Proteasome of Mycobacterium tuberculosis Is Required for Resistance to Nitric Oxide, Science, vol.302, issue.5652, pp.1963-1966, 2003.
DOI : 10.1126/science.1091176

R. Vankayalapati and P. F. Barnes, Innate and adaptive immune responses to human Mycobacterium tuberculosis infection, Tuberculosis, vol.89, issue.1, pp.77-80, 2009.
DOI : 10.1016/S1472-9792(09)70018-6

G. M. Winslow, A. Cooper, W. Reiley, M. Chatterjee, and D. L. Woodland, Early T-cell responses in tuberculosis immunity, Immunological Reviews, vol.177, issue.1, pp.284-299, 2008.
DOI : 10.1111/j.1600-065X.2008.00693.x

A. Bru and P. J. Cardona, Mathematical modeling of tuberculosis bacillary counts and cellular populations in the organs of infected mice Understanding and overcoming the barriers to T cell-mediated immunity against tuberculosis, Semin Immunol, vol.26, pp.578-587, 2010.

A. M. Cooper, Cell-Mediated Immune Responses in Tuberculosis, Annual Review of Immunology, vol.27, issue.1, pp.393-422, 2009.
DOI : 10.1146/annurev.immunol.021908.132703

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4298253/pdf

K. B. Urdahl, S. Shafiani, and J. D. Ernst, Initiation and regulation of T-cell responses in tuberculosis, Mucosal Immunology, vol.167, issue.3, pp.288-293, 2011.
DOI : 10.1371/journal.pone.0008235

J. L. Flynn, J. Chan, and P. L. Lin, Macrophages and control of granulomatous inflammation in tuberculosis, Mucosal Immunology, vol.180, issue.3, pp.271-278, 2004.
DOI : 10.4049/jimmunol.180.5.3329

S. A. Khader, J. Rangel-moreno, J. J. Fountain, C. A. Martino, W. W. Reiley et al., In a Murine Tuberculosis Model, the Absence of Homeostatic Chemokines Delays Granuloma Formation and Protective Immunity, The Journal of Immunology, vol.183, issue.12, pp.8004-8014, 2009.
DOI : 10.4049/jimmunol.0901937

S. Miranda, M. , A. Breiman, S. Allain, F. Deknuydt et al., The tuberculous granuloma: an unsuccessful host defence mechanism providing a safety shelter for the bacteria? Clin Dev Immunol 2012: 139127. 44 Natural history of tuberculosis: duration and fatality of untreated pulmonary tuberculosis in HIV negative patients: a systematic review Drug-resistant tuberculosis--current dilemmas, unanswered questions, challenges, and priority needs, 46. WHO. 2010. Multidrug and extensively drug-resistant TB (M/XDR-TB): 2010 global report on surveillance and response, pp.228-240, 2011.

C. Aagaard, T. Hoang, J. Dietrich, P. J. Cardona, A. Izzo et al., A multistage tuberculosis vaccine that confers efficient protection before and after exposure, Nature Medicine, vol.182, issue.2, pp.189-194, 2011.
DOI : 10.4049/jimmunol.0801592

P. Andersen and T. M. Doherty, The success and failure of BCG ??? implications for a novel tuberculosis vaccine, Nature Reviews Microbiology, vol.171, issue.8, pp.656-662, 2005.
DOI : 10.4049/jimmunol.171.3.1602

L. C. Rodrigues, P. Mangtani, I. Abubakar, B. B. , P. Fine et al., How does the level of BCG vaccine protection against tuberculosis fall over time? Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of costeffectiveness, BMJ Lancet, vol.343, issue.367, pp.1173-1180, 2006.

G. A. Colditz, T. F. Brewer, C. S. Berkey, M. E. Wilson, E. Burdick et al., Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature, JAMA: The Journal of the American Medical Association, vol.271, issue.9, pp.698-702, 1994.
DOI : 10.1001/jama.271.9.698

C. Lienhardt and A. Zumla, BCG: the story continues, The Lancet, vol.366, issue.9495, pp.1414-1416, 2005.
DOI : 10.1016/S0140-6736(05)67535-6

L. C. Rodrigues, S. M. Pereira, S. S. Cunha, B. Genser, M. Y. Ichihara et al., Effect of BCG revaccination on incidence of tuberculosis in school-aged children in Brazil: the BCG-REVAC cluster-randomised trial, The Lancet, vol.366, issue.9493, pp.1290-1295, 2005.
DOI : 10.1016/S0140-6736(05)67145-0

P. R. Narayanan, Influence of sex, age & nontuberculous infection at intake on the efficacy of BCG: re-analysis of 15-year data from a double-blind randomized control trial in South India, Indian J Med Res, vol.123, pp.119-124, 2006.

P. Mangtani, I. Abubakar, C. Ariti, R. Beynon, L. Pimpin et al., Protection by BCG Vaccine Against Tuberculosis: A Systematic Review of Randomized Controlled Trials, Clinical Infectious Diseases, vol.375, issue.suppl, pp.470-480, 2010.
DOI : 10.1016/S0140-6736(13)60177-4

A. A. Alangari, F. Al-zamil, A. Al-mazrou, S. Al-muhsen, S. Boisson-dupuis et al., Treatment of disseminated mycobacterial infection with high-dose IFN-gamma in a patient with IL-12Rbeta1 deficiency, Clin Dev Immunol, p.691956, 2011.

C. Infante, Centers for Disease, and Prevention Vaccine-associated paralytic poliomyelitis and BCG-osis in an immigrant child with severe combined immunodeficiency syndrome -Texas, 2013, MMWR Morb Mortal Wkly Rep, vol.63, pp.721-724, 2014.

M. Sadeghi-shabestari, N. R. North, R. J. Shimokata, K. , H. Kishimoto et al., Disseminated bacille Calmette-Guerin in Iranian children with severe combined immunodeficiency Importance of thymus-derived lymphocytes in cell-mediated immunity to infection Determination of the T-cell subset producing gamma-interferon in tuberculous pleural effusion The relative importance of T cell subsets in immunity and immunopathology of airborne Mycobacterium tuberculosis infection in mice, Int J Infect Dis Cell Immunol Microbiol Immunol J Exp Med, vol.13, issue.193, pp.420-423, 1973.

C. Geldmacher, A. Zumla, and M. Hoelscher, Interaction between HIV and Mycobacterium tuberculosis, Current Opinion in HIV and AIDS, vol.7, pp.268-275, 2012.
DOI : 10.1097/COH.0b013e3283524e32

A. M. Cooper, D. K. Dalton, T. A. Stewart, J. P. Griffin, D. G. Russell et al., Disseminated tuberculosis in interferon gamma gene-disrupted mice, Journal of Experimental Medicine, vol.178, issue.6, pp.2243-2247, 1993.
DOI : 10.1084/jem.178.6.2243

E. Jouanguy, F. Altare, S. Lamhamedi, P. Revy, J. F. Emile et al., Interferon-?? ???Receptor Deficiency in an Infant with Fatal Bacille Calmette???Gu??rin Infection, New England Journal of Medicine, vol.335, issue.26, pp.1956-1961, 1996.
DOI : 10.1056/NEJM199612263352604

C. S. Hirsch, Z. Toossi, C. Othieno, J. L. Johnson, S. K. Schwander et al., Depressed T???Cell Interferon????? Responses in Pulmonary Tuberculosis: Analysis of Underlying Mechanisms and Modulation with Therapy, The Journal of Infectious Diseases, vol.180, issue.6, pp.2069-2073, 1999.
DOI : 10.1086/315114

F. Altare, A. Durandy, D. Lammas, J. F. Emile, S. Lamhamedi et al., Impairment of Mycobacterial Immunity in Human Interleukin-12 Receptor Deficiency, Science, vol.280, issue.5368, pp.1432-1435, 1998.
DOI : 10.1126/science.280.5368.1432

R. De-jong, F. Altare, I. A. Haagen, D. G. Elferink, T. Boer et al., Severe Mycobacterial and Salmonella Infections in Interleukin-12 Receptor-Deficient Patients, Science, vol.280, issue.5368, pp.1435-1438, 1998.
DOI : 10.1126/science.280.5368.1435

D. M. Frucht, S. M. Holland-okamura, H. , S. Kashiwamura, H. Tsutsui et al., Defective monocyte costimulation for IFN-gamma production in familial disseminated Mycobacterium avium complex infection: abnormal IL-12 regulation Regulation of interferon-gamma production by IL-12 and IL-18 Novel human immunodeficiencies reveal the essential role of type-I cytokines in immunity to intracellular bacteria, J Immunol Curr Opin Immunol Immunol Today, vol.157, issue.19, pp.411-416, 1996.

R. S. Wallis, P. Kim, S. Cole, D. Hanna, B. B. Andrade et al., Tuberculosis biomarkers discovery: developments, needs, and challenges, The Lancet Infectious Diseases, vol.13, issue.4, pp.362-372, 2013.
DOI : 10.1016/S1473-3099(13)70034-3

R. Diel, R. Loddenkemper, S. Niemann, K. Meywald-walter, and A. Nienhaus, Negative and Positive Predictive Value of a Whole-Blood Interferon-?? Release Assay for Developing Active Tuberculosis, American Journal of Respiratory and Critical Care Medicine, vol.183, issue.1, pp.88-95, 2011.
DOI : 10.1186/1471-2334-7-140

H. Mittrücker, U. Steinhoff, A. Köhler, M. Krause, D. Lazar et al., Poor correlation between BCG vaccination-induced T cell responses and protection against tuberculosis, Proceedings of the National Academy of Sciences, vol.24, issue.4, pp.12434-12439, 2007.
DOI : 10.1016/j.immuni.2006.04.003

H. J. Mollenkopf, K. Hahnke, S. H. Kaufmann-kagina, B. M. Abel, T. J. Scriba et al., Transcriptional responses in mouse lungs induced by vaccination with Mycobacterium bovis BCG and infection with Mycobacterium tuberculosis, Microbes and Infection, vol.8, issue.1, pp.136-144, 2006.
DOI : 10.1016/j.micinf.2005.06.015

A. M. Gallegos, J. W. Van-heijst, M. Samstein, X. Su, E. G. Pamer et al., A Gamma Interferon Independent Mechanism of CD4 T Cell Mediated Control of M. tuberculosis Infection in vivo, PLoS Pathogens, vol.51, issue.5, pp.1002052-1002052, 2011.
DOI : 10.1371/journal.ppat.1002052.s004

M. T. Orr, H. P. Windish, E. A. Beebe, D. Argilla, P. W. Huang et al., Interferon gamma and Tumor Necrosis Factor Are Not Essential Parameters of CD4+ T-Cell Responses for Vaccine Control of Tuberculosis, J Infect Dis, p.147, 2015.

K. Bhatt, S. Verma, J. J. Ellner, and P. Salgame, ABSTRACT, Clinical and Vaccine Immunology, vol.22, issue.3, pp.258-266, 2015.
DOI : 10.1128/CVI.00721-14

M. G. Duvall, M. L. Precopio, D. A. Ambrozak, A. Jaye, A. J. Mcmichael et al., Polyfunctional T cell responses are a hallmark of HIV-2 infection, European Journal of Immunology, vol.313, issue.2, pp.350-363, 2008.
DOI : 10.4049/jimmunol.169.11.6376

P. A. Darrah, D. T. Patel, P. M. De-luca, R. W. Lindsay, D. F. Davey et al., Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major, Nature Medicine, vol.313, issue.7, pp.843-850, 2007.
DOI : 10.4049/jimmunol.165.2.969

A. Harari, V. Rozot, F. Bellutti-enders, M. Perreau, J. M. Stalder et al., Dominant TNF-??+ Mycobacterium tuberculosis???specific CD4+ T cell responses discriminate between latent infection and active disease, Nature Medicine, vol.39, issue.3, pp.372-376, 2011.
DOI : 10.1016/S0001-2998(78)80014-2

T. Hawkridge, T. J. Scriba, S. Gelderbloem, E. Smit, M. Tameris et al., Safety and Immunogenicity of a New Tuberculosis Vaccine, MVA85A, in Healthy Adults in South Africa, The Journal of Infectious Diseases, vol.175, issue.4, pp.544-552, 2008.
DOI : 10.1038/nm881

H. Hatherill and . Mahomed, Lessons learnt from the first efficacy trial of a new infant tuberculosis vaccine since BCG, Tuberculosis, vol.93, pp.143-149, 2013.

W. W. Reiley, S. Shafiani, S. T. Wittmer, G. S. Tucker-heard, J. J. Moon et al., Distinct functions of antigen-specific CD4 T cells during murine Mycobacterium tuberculosis infection, Proceedings of the National Academy of Sciences of the United States of America, pp.19408-19413, 2010.
DOI : 10.1073/pnas.0801496105

T. Lindenstrom, N. P. Knudsen, E. M. Agger, and P. Andersen, Control of Chronic Mycobacterium tuberculosis Infection by CD4 KLRG1- IL-2-Secreting Central Memory Cells, The Journal of Immunology, vol.190, issue.12, pp.6311-6319, 2013.
DOI : 10.4049/jimmunol.1300248

J. S. Woodworth, C. S. Aagaard, P. R. Hansen, J. P. Cassidy, E. M. Agger et al., Protective CD4 T Cells Targeting Cryptic Epitopes of Mycobacterium tuberculosis Resist Infection-Driven Terminal Differentiation, The Journal of Immunology, vol.192, issue.7, pp.3247-3258, 1950.
DOI : 10.4049/jimmunol.1300283

S. Srivastava and J. D. Ernst, Cutting Edge: Direct Recognition of Infected Cells by CD4 T Cells Is Required for Control of Intracellular Mycobacterium tuberculosis In Vivo, The Journal of Immunology, vol.191, issue.3, pp.1016-1020, 1950.
DOI : 10.4049/jimmunol.1301236

S. Sakai, K. D. Kauffman, J. M. Schenkel, C. C. Mcberry, K. D. Mayer-barber et al., Cutting Edge: Control of Mycobacterium tuberculosis Infection by a Subset of Lung Parenchyma-Homing CD4 T Cells, The Journal of Immunology, vol.192, issue.7, pp.2965-2969, 2014.
DOI : 10.4049/jimmunol.1400019

A. O. Moguche, S. Shafiani, C. Clemons, R. P. Larson, C. Dinh et al., ICOS and Bcl6-dependent pathways maintain a CD4 T cell population with memory-like properties during tuberculosis, The Journal of Experimental Medicine, vol.212, issue.5, p.148, 2015.
DOI : 10.1016/j.immuni.2009.07.002

M. J. Ahsan, Recent advances in the development of vaccines for tuberculosis, Therapeutic Advances in Vaccines, vol.1, issue.3, pp.66-75, 2015.
DOI : 10.1371/journal.pone.0023826

X. Y. Yang, Q. F. Chen, Y. P. Li, S. M. Wu, J. Bourguignon et al., Mycobacterium vaccae as Adjuvant Therapy to Anti-Tuberculosis Chemotherapy in Never-Treated Tuberculosis Patients: A Meta-Analysis, PLoS ONE, vol.73, issue.6, 2011.
DOI : 10.1371/journal.pone.0023826.s006

W. A. Ofori-anyinam and . Hanekom, Induction and regulation of T-cell immunity by the novel tuberculosis vaccine M72/AS01 in South African adults, Am J Respir Crit Care Med, vol.188, pp.492-502, 2013.

G. Ramon, Sur la toxine et sur I'anatoxine diphtheriques, Ann. Inst. Pasteur, vol.38, pp.1-10, 1924.

S. Lee and M. T. Nguyen, Recent Advances of Vaccine Adjuvants for Infectious Diseases, Immune Network, vol.15, issue.2, pp.51-57, 2015.
DOI : 10.4110/in.2015.15.2.51

M. S. Duthie, H. P. Windish, C. B. Fox, and S. G. Reed, Use of defined TLR ligands as adjuvants within human vaccines, Immunological Reviews, vol.6, issue.1, pp.178-196, 2011.
DOI : 10.1172/JCI200522675

N. Jounai, K. Kobiyama, F. Takeshita, and K. J. Ishii, Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination, Frontiers in cellular and infection microbiology, p.168, 2012.
DOI : 10.3389/fcimb.2012.00168

B. K. Davis, H. Wen, and J. P. Ting, The Inflammasome NLRs in Immunity, Inflammation, and Associated Diseases, Annual Review of Immunology, vol.29, issue.1, pp.707-735, 2011.
DOI : 10.1146/annurev-immunol-031210-101405

E. Tritto, F. Mosca, and E. Gregorio, Mechanism of action of licensed vaccine adjuvants, Vaccine, vol.27, issue.25-26, pp.3331-3334, 2009.
DOI : 10.1016/j.vaccine.2009.01.084

J. L. Imler and L. Zheng, Biology of Toll receptors: lessons from insects and mammals, Journal of Leukocyte Biology, vol.25, issue.1, pp.18-26, 2004.
DOI : 10.1093/nar/25.24.4876

C. Pasare and R. Medzhitov, Toll-like receptors: linking innate and adaptive immunity, Microbes and Infection, vol.6, issue.15, pp.1382-1387, 2004.
DOI : 10.1016/j.micinf.2004.08.018

R. Medzhitov, P. Preston-hurlburt, E. Kopp, A. Stadlen, C. Chen et al., MyD88 Is an Adaptor Protein in the hToll/IL-1 Receptor Family Signaling Pathways, Molecular Cell, vol.2, issue.2, pp.253-258, 1998.
DOI : 10.1016/S1097-2765(00)80136-7

K. A. Fitzgerald, D. C. Rowe, and D. T. Golenbock, Endotoxin recognition and signal transduction by the TLR4/MD2-complex, Microbes and Infection, vol.6, issue.15, pp.1361-1367, 2004.
DOI : 10.1016/j.micinf.2004.08.015

K. A. Fitzgerald, D. C. Rowe, B. J. Barnes, D. R. Caffrey, A. Visintin et al., LPS-TLR4 Signaling to IRF-3/7 and NF-??B Involves the Toll Adapters TRAM and TRIF, The Journal of Experimental Medicine, vol.18, issue.7, pp.1043-1055, 2003.
DOI : 10.1038/nature01889

D. N. Toussi and P. Massari, Immune Adjuvant Effect of Molecularly-defined Toll-Like Receptor Ligands, Vaccines, vol.8, issue.2, pp.323-353, 2014.
DOI : 10.1007/s12272-009-2100-6

M. Schnare, G. M. Barton, A. C. Holt, K. Takeda, S. Akira et al., Toll-like receptors control activation of adaptive immune responses, Nature Immunology, vol.20, issue.10, pp.947-950, 2001.
DOI : 10.1016/S0167-5699(99)01475-9

N. W. Palm and R. Medzhitov, Pattern recognition receptors and control of adaptive immunity, Immunological Reviews, vol.72, issue.1, pp.221-233, 2009.
DOI : 10.4049/jimmunol.178.4.2415

C. Olive, Pattern recognition receptors: sentinels in innate immunity and targets of new vaccine adjuvants, Expert Review of Vaccines, vol.126, issue.2, pp.237-256, 2012.
DOI : 10.1111/j.1600-065X.2011.01048.x

M. A. West, R. P. Wallin, S. P. Matthews, H. G. Svensson, R. Zaru et al., Enhanced Dendritic Cell Antigen Capture via Toll-Like Receptor-Induced Actin Remodeling, Science, vol.305, issue.5687, pp.1153-1157, 2004.
DOI : 10.1126/science.1099153

J. M. Blander and R. Medzhitov, Toll-dependent selection of microbial antigens for presentation by dendritic cells, Nature, vol.80, issue.7085, pp.808-812, 2006.
DOI : 10.1073/pnas.80.1.273

F. Sallusto, P. Schaerli, P. Loetscher, C. Schaniel, D. Lenig et al., Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation, European Journal of Immunology, vol.184, issue.9, pp.2760-2769, 1998.
DOI : 10.1084/jem.184.2.569

M. C. Dieu, B. Vanbervliet, A. Vicari, J. M. Bridon, E. Oldham et al., Selective Recruitment of Immature and Mature Dendritic Cells by Distinct Chemokines Expressed in Different Anatomic Sites, The Journal of Experimental Medicine, vol.160, issue.2, pp.373-386, 1998.
DOI : 10.1126/science.279.5349.381

S. Akira, K. Takeda, and T. Kaisho, Toll-like receptors: critical proteins linking innate and acquired immunity, Nature Immunology, vol.291, issue.8, pp.675-680, 2001.
DOI : 10.1126/science.291.5508.1544

A. H. Rahman, D. K. Taylor, and L. A. Turka, The contribution of direct TLR signaling to T cell responses, Immunologic Research, vol.9, issue.Suppl 1, pp.25-36, 2009.
DOI : 10.4049/jimmunol.176.12.7589

Z. Hua and B. Hou, TLR signaling in B-cell development and activation, Cellular & Molecular Immunology, vol.178, issue.2, pp.103-106, 2013.
DOI : 10.1084/jem.20110200

J. Dupont, J. Altclas, A. Lepetic, M. Lombardo, V. Vazquez et al., A controlled clinical trial comparing the safety and immunogenicity of a new adjuvanted hepatitis B vaccine with a standard hepatitis B vaccine, Vaccine, vol.24, issue.49-50, pp.7167-7174, 2006.
DOI : 10.1016/j.vaccine.2006.06.053

M. Kundi, New hepatitis B vaccine formulated with an improved adjuvant system, Expert Review of Vaccines, vol.68, issue.2, pp.133-140, 2007.
DOI : 10.1111/j.1523-1755.2005.00689.x

C. D. Scallan, D. W. Tingley, J. D. Lindbloom, J. S. Toomey, and S. N. Tucker, ABSTRACT, Clinical and Vaccine Immunology, vol.20, issue.1, pp.85-94, 2013.
DOI : 10.1128/CVI.00552-12

J. J. Treanor, B. Essink, S. Hull, S. Reed, R. Izikson et al., Evaluation of safety and immunogenicity of recombinant influenza hemagglutinin (H5/Indonesia/05/2005) formulated with and without a stable oil-in-water emulsion containing glucopyranosyl-lipid A (SE+GLA) adjuvant, Vaccine, vol.31, issue.48, pp.5760-5765, 2013.
DOI : 10.1016/j.vaccine.2013.08.064

C. L. Cooper, H. L. Davis, M. L. Morris, S. M. Efler, A. M. Krieg et al., Safety and immunogenicity of CPG 7909 injection as an adjuvant to Fluarix influenza vaccine, Vaccine, vol.22, issue.23-24, pp.3136-3143, 2004.
DOI : 10.1016/j.vaccine.2004.01.058

S. Pichyangkul, M. Gettayacamin, R. S. Miller, J. A. Lyon, E. Angov et al., Pre-clinical evaluation of the malaria vaccine candidate . MSP1 formulated with novel adjuvants or with alum, Vaccine, vol.22, issue.29-30, pp.3831-3840, 2004.
DOI : 10.1016/j.vaccine.2004.07.023

M. Schellenberg, P. Sillman, S. C. Vansadia, and . Rts, First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children, N Engl J Med, vol.365, pp.1863-1875, 2011.

T. Tougan, T. Aoshi, C. Coban, Y. Katakai, C. Kai et al., TLR9 adjuvants enhance immunogenicity and protective efficacy of the SE36/AHG malaria vaccine in nonhuman primate models, Human Vaccines & Immunotherapeutics, vol.174, issue.2, pp.283-290, 2013.
DOI : 10.4049/jimmunol.174.1.475

R. D. Ellis, Y. Wu, L. B. Martin, D. Shaffer, K. Miura et al., Phase 1 Study in Malaria Na??ve Adults of BSAM2/Alhydrogel??+CPG 7909, a Blood Stage Vaccine against P. falciparum Malaria, Phase 1 study in malaria naive adults of BSAM2/Alhydrogel(R)+CPG 7909, a blood stage vaccine against P. falciparum malaria, p.46094, 2012.
DOI : 10.1371/journal.pone.0046094.s002

E. Nascimento, D. F. Fernandes, E. P. Vieira, A. Campos-neto, J. A. Ashman et al., A clinical trial to evaluate the safety and immunogenicity of the LEISH-F1+MPL-SE vaccine when used in combination with meglumine antimoniate for the treatment of cutaneous leishmaniasis, Vaccine, vol.28, issue.40, pp.6581-6587, 2010.
DOI : 10.1016/j.vaccine.2010.07.063

V. Pulko, X. Liu, C. J. Krco, K. J. Harris, X. Frigola et al., TLR3-Stimulated Dendritic Cells Up-regulate B7-H1 Expression and Influence the Magnitude of CD8 T Cell Responses to Tumor Vaccination, The Journal of Immunology, vol.183, issue.6, pp.3634-3641, 2009.
DOI : 10.4049/jimmunol.0900974

N. Butowski, S. M. Chang, L. Junck, L. M. Deangelis, L. Abrey et al., A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05), Journal of Neuro-Oncology, vol.5, issue.2, pp.175-182, 2009.
DOI : 10.1089/jir.1991.11.199

N. Isambert, P. Fumoleau, C. Paul, C. Ferrand, S. Zanetta et al., Phase I study of OM-174, a lipid A analogue, with assessment of immunological response, in patients with refractory solid tumors, BMC Cancer, vol.563, issue.1, p.172, 2013.
DOI : 10.1016/j.ejphar.2007.02.018

URL : https://hal.archives-ouvertes.fr/inserm-00813522

J. Karbach, A. Neumann, A. Atmaca, C. Wahle, K. Brand et al., Efficient In vivo Priming by Vaccination with Recombinant NY-ESO-1 Protein and CpG in Antigen Naive Prostate Cancer Patients, Clinical Cancer Research, vol.17, issue.4, pp.861-870, 2011.
DOI : 10.1158/1078-0432.CCR-10-1811

C. Manegold, D. Gravenor, D. Woytowitz, J. Mezger, V. Hirsh et al., Randomized Phase II Trial of a Toll-Like Receptor 9 Agonist Oligodeoxynucleotide, PF-3512676, in Combination With First-Line Taxane Plus Platinum Chemotherapy for Advanced-Stage Non???Small-Cell Lung Cancer, Journal of Clinical Oncology, vol.26, issue.24, pp.3979-3986, 2008.
DOI : 10.1200/JCO.2007.12.5807

P. Pellegrino, E. Clementi, and S. Radice, On vaccine's adjuvants and autoimmunity: Current evidence and future perspectives, Autoimmunity Reviews, vol.14, issue.10, pp.880-888, 2015.
DOI : 10.1016/j.autrev.2015.05.014

D. Gregorio, E. , E. Tritto, and R. Rappuoli, Alum adjuvanticity: Unraveling a century old mystery, European Journal of Immunology, vol.320, issue.8, pp.2068-2071, 2008.
DOI : 10.4049/jimmunol.178.8.5271

R. L. Rubin, F. L. Tang, A. H. Lucas, H. L. Spiegelberg, and E. M. Tan, IgG subclasses of anti-tetanus toxoid antibodies in adult and newborn normal subjects and in patients with 151, 1986.

C. J. Clements and E. Griffiths, The global impact of vaccines containing aluminium adjuvants, Vaccine, vol.20, issue.3, pp.24-33, 2002.
DOI : 10.1016/S0264-410X(02)00168-8

N. L. Letvin, B. R. Bloom, and S. L. Hoffman, Prospects for Vaccines to Protect Against AIDS, Tuberculosis, and Malaria, JAMA, vol.285, issue.5, pp.606-611, 2001.
DOI : 10.1001/jama.285.5.606

C. Moser, M. Muller, M. D. Kaeser, U. Weydemann, and M. Amacker, Influenza virosomes as vaccine adjuvant and carrier system, Expert Review of Vaccines, vol.7, issue.7, pp.779-791, 2013.
DOI : 10.1016/0264-410X(96)00042-4

A. Huckriede, L. Bungener, T. Daemen, and J. Wilschut, Influenza Virosomes in Vaccine Development, Methods Enzymol, vol.373, pp.74-91, 2003.
DOI : 10.1016/S0076-6879(03)73005-5

P. A. Bovier, : a virosomal vaccine to prevent hepatitis A infection, Expert Review of Vaccines, vol.13, issue.8, pp.1141-1150, 2008.
DOI : 10.1016/0264-410X(92)90506-F

G. Ott, G. L. Barchfeld, D. Chernoff, R. Radhakrishnan, P. Van-hoogevest et al., MF59 Design and Evaluation of a Safe and Potent Adjuvant for Human Vaccines, Pharm Biotechnol, vol.6, pp.277-296, 1995.
DOI : 10.1007/978-1-4615-1823-5_10

S. L. Giannini, E. Hanon, P. Moris, M. Van-mechelen, S. Morel et al., Enhanced humoral and memory B cellular immunity using HPV16/18 L1 VLP vaccine formulated with the MPL/aluminium salt combination (AS04) compared to aluminium salt only, Vaccine, vol.24, issue.33-34, pp.5937-5949, 2006.
DOI : 10.1016/j.vaccine.2006.06.005

D. Carvalho, N. , J. Teixeira, C. M. Roteli-martins, P. Naud et al., Sustained efficacy and immunogenicity of the HPV-16/18 AS04-adjuvanted vaccine up to 7.3 years in young adult women, Vaccine, vol.28, issue.38, pp.6247-6255, 2010.
DOI : 10.1016/j.vaccine.2010.07.007

P. S. Naud, C. M. Roteli-martins, N. S. De-carvalho, J. C. Teixeira, P. C. De-borba et al., Sustained efficacy, immunogenicity, and safety of the HPV-16/18 AS04-adjuvanted vaccine, Human Vaccines & Immunotherapeutics, vol.10, issue.8, pp.2147-2162, 2014.
DOI : 10.1016/j.virol.2003.12.027

N. K. Tong, J. Beran, S. A. Kee, J. L. Miguel, C. Sanchez et al., Immunogenicity and safety of an adjuvanted hepatitis B vaccine in pre-hemodialysis and hemodialysis patients, Kidney International, vol.68, issue.5, pp.2298-2303, 2005.
DOI : 10.1111/j.1523-1755.2005.00689.x

T. Vesikari, M. Knuf, P. Wutzler, A. Karvonen, D. Kieninger-baum et al., Oil-in-Water Emulsion Adjuvant with Influenza Vaccine in Young Children, New England Journal of Medicine, vol.365, issue.15, pp.1406-1416, 2011.
DOI : 10.1056/NEJMoa1010331

E. M. Agger, I. Rosenkrands, A. W. Olsen, G. Hatch, A. Williams et al., Protective immunity to tuberculosis with Ag85B-ESAT-6 in a synthetic cationic adjuvant system IC31, Vaccine, vol.24, issue.26, pp.5452-5460, 2006.
DOI : 10.1016/j.vaccine.2006.03.072

T. A. Olafsdottir, K. Lingnau, E. Nagy, and I. Jonsdottir, , a Two-Component Novel Adjuvant Mixed with a Conjugate Vaccine Enhances Protective Immunity against Pneumococcal Disease in Neonatal Mice, Scandinavian Journal of Immunology, vol.129, issue.Pt 2, pp.194-202, 2009.
DOI : 10.1042/bj3020535

J. T. Van-dissel, S. A. Joosten, S. T. Hoff, D. Soonawala, C. Prins et al., A novel liposomal adjuvant system, CAF01, promotes long-lived Mycobacterium tuberculosis-specific T-cell responses in human, Vaccine, vol.32, issue.52, pp.7098-7107, 2014.
DOI : 10.1016/j.vaccine.2014.10.036

S. Bertholet, G. C. Ireton, M. Kahn, J. Guderian, R. Mohamath et al., Identification of Human T Cell Antigens for the Development of Vaccines against Mycobacterium tuberculosis, The Journal of Immunology, vol.181, issue.11, pp.7948-7957, 2008.
DOI : 10.4049/jimmunol.181.11.7948

S. L. Sampson, Mycobacterial PE/PPE Proteins at the Host-Pathogen Interface, Clinical and Developmental Immunology, vol.5, issue.7, p.497203, 2011.
DOI : 10.1186/1471-2172-11-18

S. Kohli, Y. Singh, K. Sharma, A. Mittal, N. Z. Ehtesham et al., Comparative genomic and proteomic analyses of PE/PPE multigene family of Mycobacterium tuberculosis H37Rv and H37Ra reveal novel and interesting differences with implications in virulence, Nucleic Acids Research, vol.188, issue.15, pp.7113-7122, 2012.
DOI : 10.1128/JB.01212-06

F. Sayes, L. Sun, M. D. Luca, R. Simeone, N. Degaiffier et al., Strong Immunogenicity and Cross-Reactivity of Mycobacterium tuberculosis ESX-5 Type VII Secretion -Encoded PE-PPE Proteins Predicts Vaccine Potential, Cell Host & Microbe, vol.11, issue.4, pp.352-363, 2012.
DOI : 10.1016/j.chom.2012.03.003

URL : https://hal.archives-ouvertes.fr/pasteur-01104794

P. Brodin, I. Rosenkrands, P. Andersen, S. T. Cole, and R. Brosch, ESAT-6 proteins: protective antigens and virulence factors?, Trends in Microbiology, vol.12, issue.11, pp.500-508, 2004.
DOI : 10.1016/j.tim.2004.09.007

G. J. Jones, S. V. Gordon, R. G. Hewinson, and H. M. Vordermeier, Screening of Predicted Secreted Antigens from Mycobacterium bovis Reveals the Immunodominance of the ESAT-6 Protein Family, Infection and Immunity, vol.78, issue.3, pp.1326-1332, 2010.
DOI : 10.1128/IAI.01246-09

A. S. Mustafa, Y. A. Skeiky, R. Al-attiyah, M. R. Alderson, R. G. Hewinson et al., Immunogenicity of Mycobacterium tuberculosis Antigens in Mycobacterium bovis BCG-Vaccinated and M. bovis-Infected Cattle, Infection and Immunity, vol.74, issue.8, pp.4566-4572, 2006.
DOI : 10.1128/IAI.01660-05

R. L. Skjot, T. Oettinger, I. Rosenkrands, P. Ravn, I. Brock et al., Comparative Evaluation of Low-Molecular-Mass Proteins from Mycobacterium tuberculosis Identifies Members of the ESAT-6 Family as Immunodominant T-Cell Antigens, Infection and Immunity, vol.68, issue.1, pp.214-220, 2000.
DOI : 10.1128/IAI.68.1.214-220.2000

O. Neill, L. A. , and A. G. Bowie, The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling, Nature Reviews Immunology, vol.19, issue.5, pp.353-364, 2007.
DOI : 10.4049/jimmunol.175.2.839

J. C. Chow, D. W. Young, D. T. Golenbock, W. J. Christ, and F. Gusovsky, Toll-like Receptor-4 Mediates Lipopolysaccharide-induced Signal Transduction, Journal of Biological Chemistry, vol.392, issue.16, pp.10689-10692, 1999.
DOI : 10.1038/sj.bjp.0702596

URL : http://www.jbc.org/content/274/16/10689.full.pdf

J. T. Evans, C. W. Cluff, D. A. Johnson, M. J. Lacy, D. H. Persing et al., Enhancement of antigen-specific immunity via the TLR4 ligands MPL??? adjuvant and Ribi.529, Expert Review of Vaccines, vol.1, issue.2, pp.219-229, 2003.
DOI : 10.1038/35021228

M. R. Alderson, P. Mcgowan, J. R. Baldridge, and P. Probst, TLR4 agonists as immunomodulatory agents, Journal of Endotoxin Research, vol.174, issue.5, pp.313-319, 2006.
DOI : 10.4049/jimmunol.174.10.6416

K. Johansen, U. Schroder, and L. Svensson, Immunogenicity and protective efficacy of a formalin-inactivated rotavirus vaccine combined with lipid adjuvants, Vaccine, vol.21, issue.5-6, pp.368-375, 2003.
DOI : 10.1016/S0264-410X(02)00617-5

M. Qiao, K. Murata, A. R. Davis, S. H. Jeong, and T. J. Liang, Hepatitis C virus???like particles combined with novel adjuvant systems enhance virus-specific immune responses, Hepatology, vol.10, issue.1, pp.52-59, 2003.
DOI : 10.1053/jhep.2003.50000

R. N. Coler, Y. A. Skeiky, K. Bernards, K. Greeson, D. Carter et al., Immunization with a polyprotein vaccine consisting of the T-Cell antigens thiol-specific antioxidant, Leishmania major stress-inducible protein 1, and 153, 2002.

L. Brandt, M. Elhay, I. Rosenkrands, E. B. Lindblad, and P. Andersen, ESAT-6 Subunit Vaccination against Mycobacterium tuberculosis, Infection and Immunity, vol.68, issue.2, pp.791-795, 2000.
DOI : 10.1128/IAI.68.2.791-795.2000

URL : http://iai.asm.org/content/68/2/791.full.pdf

R. L. Richards, M. Rao, N. M. Wassef, G. M. Glenn, S. W. Rothwell et al., Liposomes containing lipid A serve as an adjuvant for induction of antibody and cytotoxic Tcell responses against RTS,S malaria antigen, Infect Immun, vol.66, pp.2859-2865, 1998.

L. S. Rickman, D. M. Gordon, R. Wistar, J. , U. Krzych et al., Use of adjuvant containing mycobacterial cell-wall skeleton, monophosphoryl lipid A, and squalane in malaria circumsporozoite protein vaccine, The Lancet, vol.337, issue.8748, pp.998-1001, 1991.
DOI : 10.1016/0140-6736(91)92659-P

D. M. Gordon, T. W. Mcgovern, U. Krzych, J. C. Cohen, I. Schneider et al., Safety, Immunogenicity, and Efficacy of a Recombinantly Produced Plasmodium falciparum Circumsporozoite Protein-Hepatitis B Surface Antigen Subunit Vaccine, Journal of Infectious Diseases, vol.171, issue.6, pp.1576-1585, 1995.
DOI : 10.1093/infdis/171.6.1576

A. Vafai, Boosting immune response with a candidate varicella-zoster virus glycoprotein subunit vaccine, Vaccine, vol.13, issue.14, pp.1336-1338, 1995.
DOI : 10.1016/0264-410X(94)00073-V

S. Thoelen, P. Van-damme, C. Mathei, G. Leroux-roels, I. Desombere et al., Safety and immunogenicity of a hepatitis B vaccine formulated with a novel adjuvant system, Vaccine, vol.16, issue.7, pp.708-714, 1998.
DOI : 10.1016/S0264-410X(97)00254-5

S. Mccormack, A. Tilzey, A. Carmichael, F. Gotch, J. Kepple et al., A phase I trial in HIV negative healthy volunteers evaluating the effect of potent adjuvants on immunogenicity of a recombinant gp120W61D derived from dual tropic R5X4 HIV-1ACH320, Vaccine, vol.18, issue.13, pp.1166-1177, 2000.
DOI : 10.1016/S0264-410X(99)00388-6

C. Cekic, C. R. Casella, C. A. Eaves, A. Matsuzawa, H. Ichijo et al., Selective Activation of the p38 MAPK Pathway by Synthetic Monophosphoryl Lipid A, Journal of Biological Chemistry, vol.181, issue.46, pp.31982-31991, 2009.
DOI : 10.4049/jimmunol.181.5.3464

P. Bandyopadhyay and M. Johnson, Fatty alcohols or fatty acids as niosomal hybrid carrier: Effect on vesicle size, encapsulation efficiency and in vitro dye release, Colloids and Surfaces B: Biointerfaces, vol.58, issue.1, pp.68-71, 2007.
DOI : 10.1016/j.colsurfb.2007.01.014

P. Rallabhandi, A. Awomoyi, K. E. Thomas, A. Phalipon, Y. Fujimoto et al., Differential Activation of Human TLR4 by Escherichia coli and Shigella flexneri 2a Lipopolysaccharide: Combined Effects of Lipid A Acylation State and TLR4 Polymorphisms on Signaling, The Journal of Immunology, vol.180, issue.2, pp.1139-1147, 2008.
DOI : 10.4049/jimmunol.180.2.1139

D. M. Cataldo and G. Van-nest, The adjuvant MF59 increases the immunogenicity and protective efficacy of subunit influenza vaccine in mice, Vaccine, vol.15, issue.16, pp.1710-1715, 1997.
DOI : 10.1016/S0264-410X(97)00115-1

C. B. Fox, C. Huynh, M. K. O-'hara, and A. Onu, Technology transfer of oil-in-water emulsion adjuvant manufacturing for pandemic influenza vaccine production in Romania, Vaccine, vol.31, issue.12, pp.1633-1640, 2013.
DOI : 10.1016/j.vaccine.2012.10.048

X. Li, L. Du, C. Wang, Y. Liu, X. Mei et al., Highly efficient and lowly toxic docetaxel nanoemulsions for intravenous injection to animals, Pharmazie, vol.66, pp.479-483, 2011.

R. N. Coler, T. Hudson, S. Hughes, P. W. Huang, E. A. Beebe et al., Vaccination Produces CD4 T Cells with a Novel CD154???CD40-Dependent Cytolytic Mechanism, The Journal of Immunology, vol.195, issue.7, pp.3190-3197, 2015.
DOI : 10.4049/jimmunol.1501118

S. L. Baldwin, S. Bertholet, V. Reese, L. K. Ching, S. G. Reed et al., The Importance of Adjuvant Formulation in the Development of a Tuberculosis Vaccine, The Journal of Immunology, vol.188, issue.5, pp.2189-2197, 2012.
DOI : 10.4049/jimmunol.1102696

S. L. Baldwin, V. A. Reese, P. W. Huang, E. A. Beebe, B. K. Podell et al., ABSTRACT, Clinical and Vaccine Immunology, vol.23, issue.2, pp.137-147, 2015.
DOI : 10.1128/CVI.00458-15

R. N. Coler, S. Bertholet, S. O. Pine, M. T. Orr, V. Reese et al., Therapeutic Immunization against Mycobacterium tuberculosis Is an Effective Adjunct to Antibiotic Treatment, The Journal of Infectious Diseases, vol.74, issue.8, pp.1242-1252, 2013.
DOI : 10.1128/IAI.74.5.2817-2822.2006

S. L. Baldwin, V. Reese, B. Granger, M. T. Orr, G. C. Ireton et al., ABSTRACT, Clinical and Vaccine Immunology, vol.21, issue.9, pp.1309-1313, 2014.
DOI : 10.1128/CVI.00372-14

M. T. Orr, C. B. Fox, S. L. Baldwin, S. J. Sivananthan, E. Lucas et al., Adjuvant formulation structure and composition are critical for the development of an effective vaccine against tuberculosis, Journal of Controlled Release, vol.172, issue.1, pp.190-200, 2013.
DOI : 10.1016/j.jconrel.2013.07.030

A. Seubert, S. Calabro, L. Santini, B. Galli, A. Genovese et al., Adjuvanticity of the oil-in-water emulsion MF59 is independent of Nlrp3 inflammasome but requires the adaptor protein MyD88, Proceedings of the National Academy of Sciences of the United States of America, pp.11169-11174, 2011.
DOI : 10.1126/science.287.5459.1816

A. Pantel, C. Cheong, D. Dandamudi, E. Shrestha, S. Mehandru et al., A new synthetic TLR4 agonist, GLA, allows dendritic cells targeted with antigen to elicit Th1 T-cell immunity in vivo, European Journal of Immunology, vol.30, issue.1, pp.101-109, 2012.
DOI : 10.1016/j.it.2008.09.006

S. L. Lambert, C. F. Yang, Z. Liu, R. Sweetwood, J. Zhao et al., Molecular and Cellular Response Profiles Induced by the TLR4 Agonist-Based Adjuvant Glucopyranosyl Lipid A, PLoS ONE, vol.265, issue.12, 2012.
DOI : 10.1371/journal.pone.0051618.s002

M. T. Orr, M. S. Duthie, H. P. Windish, E. A. Lucas, J. A. Guderian et al., MyD88 and TRIF synergistic interaction is required for TH1-cell polarization with a synthetic TLR4 agonist adjuvant, European Journal of Immunology, vol.8, issue.9, pp.2398-2408, 2013.
DOI : 10.1371/journal.ppat.1002963

D. Gregorio, E. , E. Caproni, and J. B. Ulmer, Vaccine Adjuvants: Mode of Action, Frontiers in Immunology, vol.4, p.214, 2013.
DOI : 10.3389/fimmu.2013.00214

A. Zumla, P. Nahid, and S. T. Cole, Advances in the development of new tuberculosis drugs and treatment regimens, Nature Reviews Drug Discovery, vol.109, issue.5, pp.388-404, 2013.
DOI : 10.1073/pnas.1205735109

B. Castelnuovo, A review of compliance to anti tuberculosis treatment and risk factors for defaulting treatment in Sub Saharan Africa, African health sciences, vol.10, pp.320-324, 2010.

D. Steenwinkel, J. E. , G. J. De-knegt, M. T. Ten-kate, H. Verbrugh et al., Relapse of tuberculosis versus primary tuberculosis; course, pathogenesis and therapy in mice, Tuberculosis, vol.93, issue.2, pp.213-221, 2013.
DOI : 10.1016/j.tube.2012.11.006

A. Zumla, I. Abubakar, M. Raviglione, M. Hoelscher, L. Ditiu et al., Drug-Resistant Tuberculosis???Current Dilemmas, Unanswered Questions, Challenges, and Priority Needs, The Journal of Infectious Diseases, vol.12, issue.suppl_2, pp.228-240, 2012.
DOI : 10.1016/j.prrv.2010.09.002

URL : https://academic.oup.com/jid/article-pdf/205/suppl_2/S228/18067515/jir858.pdf

J. L. Johnson, R. M. Kamya, A. Okwera, A. M. Loughlin, S. Nyole et al., Immunotherapy in Non???Human Immunodeficiency Virus???Infected Ugandan Adults with Newly Diagnosed Pulmonary Tuberculosis, The Journal of Infectious Diseases, vol.181, issue.4, pp.1304-1312, 2000.
DOI : 10.1086/315393

D. Dlugovitzky, G. Fiorenza, M. Farroni, C. Bogue, C. Stanford et al., Immunological consequences of three doses of heat-killed Mycobacterium vaccae in the immunotherapy of tuberculosis, Respiratory Medicine, vol.100, issue.6, pp.1079-1087, 2006.
DOI : 10.1016/j.rmed.2005.09.026

R. M. Mccune, W. Jr, R. Mcdermott, and . Tompsett, THE FATE OF MYCOBACTERIUM TUBERCULOSIS IN MOUSE TISSUES AS DETERMINED BY THE MICROBIAL ENUMERATION TECHNIQUE: II. THE CONVERSION OF TUBERCULOUS INFECTION TO THE LATENT STATE BY THE ADMINISTRATION OF PYRAZINAMIDE AND A COMPANION DRUG, Journal of Experimental Medicine, vol.104, issue.5, pp.763-802, 1956.
DOI : 10.1084/jem.104.5.763

R. M. Mccune, R. Jr, and . Tompsett, FATE OF MYCOBACTERIUM TUBERCULOSIS IN MOUSE TISSUES AS DETERMINED BY THE MICROBIAL ENUMERATION TECHNIQUE: I. THE PERSISTENCE OF DRUG-SUSCEPTIBLE TUBERCLE BACILLI IN THE TISSUES DESPITE PROLONGED ANTIMICROBIAL THERAPY, Journal of Experimental Medicine, vol.104, issue.5, pp.737-762, 1956.
DOI : 10.1084/jem.104.5.737

A. M. Lenaerts, S. E. Chase, A. J. Chmielewski, and M. H. Cynamon, Evaluation of rifapentine in long-term treatment regimens for tuberculosis in mice, Antimicrob Agents Chemother, vol.43, pp.2356-2360, 1999.

A. M. Lenaerts, S. E. Chase, and M. H. Cynamon, Evaluation of Rifalazil in a Combination Treatment Regimen as an Alternative to Isoniazid-Rifampin Therapy in a Mouse Tuberculosis Model, Antimicrobial Agents and Chemotherapy, vol.44, issue.11, pp.3167-3168, 2000.
DOI : 10.1128/AAC.44.11.3167-3168.2000

A. A. Bachmanov, D. R. Reed, G. K. Beauchamp, and M. G. Tordoff, Food intake, water intake, and drinking spout side preference of 28 mouse strains, Behavior Genetics, vol.32, issue.6, pp.435-443, 2002.
DOI : 10.1023/A:1020884312053

R. C. Anderson, C. B. Fox, T. S. Dutill, N. Shaverdian, T. L. Evers et al., Physicochemical characterization and biological activity of synthetic TLR4 agonist formulations, Colloids and Surfaces B: Biointerfaces, vol.75, issue.1, pp.123-132, 2010.
DOI : 10.1016/j.colsurfb.2009.08.022

S. Buccheri, R. Reljic, N. Caccamo, S. Meraviglia, J. Ivanyi et al., Prevention of the post-chemotherapy relapse of tuberculous infection by combined immunotherapy, Tuberculosis, vol.89, issue.1, pp.91-94, 2009.
DOI : 10.1016/j.tube.2008.09.001

B. P. Kelly, S. K. Furney, M. T. Jessen, and I. M. Orme, Low-dose aerosol infection model for testing drugs for efficacy against Mycobacterium tuberculosis, Antimicrob Agents Chemother, vol.40, pp.2809-2812, 1996.

W. P. Gill, N. S. Harik, M. R. Whiddon, R. P. Liao, J. E. Mittler et al., A replication clock for Mycobacterium tuberculosis, Nature Medicine, vol.181, issue.2, pp.211-214, 2009.
DOI : 10.1371/journal.pone.0001502

S. A. Musa, Y. Kim, R. Hashim, G. Z. Wang, C. Dimmer et al., Response of inbred mice to aerosol challenge with Mycobacterium tuberculosis, Infection and Immunity, vol.55, pp.1862-1866, 1987.

O. C. Turner, R. G. Keefe, I. Sugawara, H. Yamada, and I. M. Orme, SWR Mice Are Highly Susceptible to Pulmonary Infection with Mycobacterium tuberculosis, Infection and Immunity, vol.71, issue.9, pp.5266-5272, 2003.
DOI : 10.1128/IAI.71.9.5266-5272.2003

P. E. Andersen, [Endovascular interventional treatment of hemoptysis], Ugeskr Laeger, vol.167, pp.3160-3163, 2005.

P. Ravn, I. Brock, P. Andersen, and K. Weldingh, [A possible successor of the Mantoux test after 97 years], Ugeskr Laeger, vol.167, pp.2905-2906, 2005.

C. H. Clegg, R. Roque, N. Van-hoeven, L. Perrone, S. L. Baldwin et al., Adjuvant solution for pandemic influenza vaccine production, Proceedings of the National Academy of Sciences, pp.17585-17590, 2012.
DOI : 10.1016/j.vaccine.2010.12.128

URL : http://www.pnas.org/content/109/43/17585.full.pdf

J. Kovarik and C. A. Siegrist, The search for novel adjuvants for early life vaccinations: can "danger" motifs show us the way?, Arch Immunol Ther Exp (Warsz), vol.49, pp.209-215, 2001.

F. V. Chisari and C. Ferrari, Hepatitis B Virus Immunopathogenesis, Annual Review of Immunology, vol.13, issue.1, pp.29-60, 1995.
DOI : 10.1146/annurev.iy.13.040195.000333

K. Dredge, J. B. Marriott, S. M. Todryk, and A. G. Dalgleish, Adjuvants and the promotion of Th1-type cytokines in tumour immunotherapy, Cancer Immunology, Immunotherapy, vol.51, issue.10, pp.521-531, 2002.
DOI : 10.1007/s00262-002-0309-z

K. Rubtsova, A. V. Rubtsov, L. F. Van-dyk, J. W. Kappler, and P. Marrack, T-box transcription factor T-bet, a key player in a unique type of B-cell activation essential for effective viral clearance, Proceedings of the National Academy of Sciences, vol.110, issue.34, pp.3216-3224, 2013.
DOI : 10.1016/j.virol.2008.07.031

T. Yoshimoto, K. Takeda, T. Tanaka, K. Ohkusu, S. Kashiwamura et al., IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-gamma production, J Immunol, vol.161, pp.3400-3407, 1998.

Y. S. Kang, Y. Do, H. K. Lee, S. H. Park, C. Cheong et al., A Dominant Complement Fixation Pathway for Pneumococcal Polysaccharides Initiated by SIGN-R1 Interacting with C1q, Cell, vol.125, issue.1, pp.47-58, 2006.
DOI : 10.1016/j.cell.2006.01.046

T. Junt, E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang et al., Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells, Nature, vol.9, issue.7166, pp.110-114, 2007.
DOI : 10.1038/nature06287

M. Vono, M. Taccone, P. Caccin, M. Gallotta, G. Donvito et al., The adjuvant MF59 induces ATP release from muscle that potentiates response to vaccination, Proceedings of the National Academy of Sciences, vol.186, issue.7, pp.21095-21100, 2013.
DOI : 10.4049/jimmunol.1003020

N. D. Cauwelaert, A. L. Desbien, T. E. Hudson, S. O. Pine, S. G. Reed et al., The TLR4 Agonist Vaccine Adjuvant, GLA-SE, Requires Canonical and Atypical Mechanisms of Action for TH1 Induction, PLOS ONE, vol.188, issue.2, p.146372, 2016.
DOI : 10.1371/journal.pone.0146372.g004