, Fact sheet-latest statistics on the status of the AIDS epidemic, 2017.

K. V. Ramana, Effect of highly active antiretroviral therapy (HAART) on human immunodeficiency virus disease pathogenesis and progression, Am. J. Public Health Res. Am. J. Public Health Res, vol.2, pp.68-74, 2014.

M. G. Sension, Long-term suppression of HIV infection: benefits and limitations of current treatment options, J. Assoc. Nurses AIDS Care, vol.18, 2007.

P. B. Gilbert, Correlation between immunologic responses to a recombinant glycoprotein 120 vaccine and incidence of HIV-1 infection in a phase 3 HIV-1 preventive vaccine trial, J. Infect. Dis, vol.191, pp.666-677, 2005.

S. P. Buchbinder, Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-ofconcept trial, Lancet, vol.372, pp.1881-1893, 2008.

M. J. Mcelrath, HIV-1 vaccine-induced immunity in the test-of-concept Step Study: a case-cohort analysis, Lancet, vol.372, pp.1894-1905, 2008.

Z. Moodie, Continued follow-up of phambili phase 2b randomized HIV-1 vaccine trial participants supports increased HIV-1 acquisition among vaccinated men, PLoS ONE, vol.10, p.137666, 2015.

B. F. Haynes, Immune-correlates analysis of an HIV-1 vaccine efficacy trial, N. Engl. J. Med, vol.366, pp.1275-1286, 2012.

S. Zolla-pazner, Vaccine-induced IgG antibodies to V1V2 regions of multiple HIV-1 subtypes correlate with decreased risk of HIV-1 infection, PLoS ONE, vol.9, p.87572, 2014.

L. Lin, COMPASS identifies T-cell subsets correlated with clinical outcomes, Nat. Biotechnol, vol.33, pp.610-616, 2015.

S. Rerks-ngarm, Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand, N. Engl. J. Med, vol.361, pp.2209-2220, 2009.

G. D. Tomaras, Vaccine-induced plasma IgA specific for the C1 region of the HIV-1 envelope blocks binding and effector function of IgG, Proc. Natl Acad. Sci, vol.110, pp.9019-9024, 2013.

J. H. Kim, S. Rerks-ngarm, J. Excler, and N. L. Michael, HIV vaccines-lessons learned and the way forward, Curr. Opin. HIV AIDS, vol.5, pp.428-434, 2010.

A. J. Mcmichael and B. F. Haynes, Lessons learned from HIV-1 vaccine trials: new priorities and directions, Nat. Immunol, vol.13, pp.423-427, 2012.

L. Zhao, Nanoparticle vaccines, Vaccine, vol.32, pp.327-337, 2014.

A. Roldão, M. C. Mellado, L. R. Castilho, M. J. Carrondo, and P. M. Alves, Virus-like particles in vaccine development, Expert. Rev. Vaccin, vol.9, pp.1149-1176, 2010.

M. F. Bachmann and G. T. Jennings, Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns, Nat. Rev. Immunol, vol.10, pp.787-796, 2010.

S. T. Reddy, Exploiting lymphatic transport and complement activation in nanoparticle vaccines, Nat. Biotechnol, vol.25, pp.1159-1164, 2007.

W. Zhang, Immunopotentiator-loaded polymeric microparticles as robust adjuvant to improve vaccine efficacy, Pharm. Res, vol.32, pp.2837-2850, 2015.

T. Courant, Tailoring nanostructured lipid carriers for the delivery of protein antigens: physicochemical properties versus immunogenicity studies, Biomaterials, vol.136, pp.29-42, 2017.
URL : https://hal.archives-ouvertes.fr/cea-02202450

M. Rolland, D. C. Nickle, and J. I. Mullins, HIV-1 group M conserved elements vaccine, PLoS Pathog, vol.3, p.157, 2007.

Y. Ataman-Önal, Surfactant-free anionic PLA nanoparticles coated with HIV-1 p24 protein induced enhanced cellular and humoral immune responses in various animal models, J. Control. Release, vol.112, pp.175-185, 2006.

B. J. Flynn, Immunization with HIV Gag targeted to dendritic cells followed by recombinant New York vaccinia virus induces robust T-cell immunity in nonhuman primates, Proc. Natl Acad. Sci, vol.108, pp.7131-7136, 2011.

G. Romain, CD34-derived dendritic cells transfected ex vivo with HIV-Gag mRNA induce polyfunctional T-cell responses in nonhuman primates, Eur. J. Immunol, vol.42, 2012.

V. Kulkarni, HIV-1 conserved elements p24CE DNA vaccine induces humoral immune responses with broad epitope recognition in macaques, PLoS ONE, vol.9, p.111085, 2014.

O. Epaulard, Macrophage-and neutrophil-derived TNF-? instructs skin langerhans cells to prime antiviral immune responses, J. Immunol, vol.193, pp.2416-2426, 2014.

A. Flamar, Delivering HIV Gagp24 to DCIR induces strong antibody responses in vivo, PLoS. One, vol.10, p.135513, 2015.
URL : https://hal.archives-ouvertes.fr/cea-01825887

N. Salabert, Intradermal injection of an anti-langerin-HIVGag fusion vaccine targets epidermal Langerhans cells in nonhuman primates and can be tracked in vivo, Eur. J. Immunol, vol.46, pp.689-700, 2016.

C. Li, Protection against SHIV-KB9 infection by combining rDNA and rFPV vaccines based on HIV multiepitope and p24 protein in Chinese rhesus macaques, Clin. Dev. Immunol, vol.2012, pp.1-9, 2012.

P. Kiepiela, CD8+ T-cell responses to different HIV proteins have discordant associations with viral load, Nat. Med, vol.13, pp.46-53, 2007.

M. Rolland, Broad and Gag-biased HIV-1 epitope repertoires are associated with lower viral loads, PLoS ONE, vol.3, p.1424, 2008.

R. Zuniga, Relative dominance of Gag p24-specific cytotoxic T lymphocytes is associated with human immunodeficiency virus control, J. Virol, vol.80, pp.3122-3125, 2006.

C. Bode, G. Zhao, F. Steinhagen, T. Kinjo, and D. M. Klinman, CpG DNA as a vaccine adjuvant, Expert. Rev. Vaccin, vol.10, pp.499-511, 2011.

O. Borges, Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles, Eur. J. Pharm. Biopharm, vol.69, pp.405-416, 2008.

K. K. Van-rompay, Tackling HIV and AIDS: contributions by non-human primate models, Lab. Anim, vol.46, pp.259-270, 2017.

M. Vaccari, Adjuvant-dependent innate and adaptive immune signatures of risk of SIV mac251 acquisition, Nat. Med, vol.22, pp.762-770, 2016.

G. Hartmann, G. J. Weiner, and A. M. Krieg, CpG DNA: a potent signal for growth, activation, and maturation of human dendritic cells, Proc. Natl. Acad. Sci, vol.96, pp.9305-9310, 1999.

C. Barnier-quer, A. Elsharkawy, S. Romeijn, A. Kros, and W. Jiskoot, Adjuvant effect of cationic liposomes for subunit influenza vaccine: influence of antigen loading method, cholesterol and immune modulators, Pharmaceutics, vol.5, pp.392-410, 2013.

S. L. Demento, TLR9-targeted biodegradable nanoparticles as immunization vectors protect against West Nile encephalitis, J. Immunol, vol.185, pp.2989-2997, 2010.

S. Fischer, Concomitant delivery of a CTL-restricted peptide antigen and CpG ODN by PLGA microparticles induces cellular immune response, J. Drug. Target, vol.17, pp.652-661, 2009.

E. Zupan?i?, Rational design of nanoparticles towards targeting antigenpresenting cells and improved T cell priming, J. Control. Release Off. J. Control. Release Soc, vol.258, pp.182-195, 2017.

J. R. Baldridge, Taking a Toll on human disease: toll-like receptor 4 agonists as vaccine adjuvants and monotherapeutic agents, Expert. Opin. Biol. Ther, vol.4, pp.1129-1138, 2004.

H. Bentala, Removal of phosphate from lipid A as a strategy to detoxify lipopolysaccharide, Shock, vol.18, pp.561-566, 2002.

C. R. Alving, K. K. Peachman, M. Rao, and S. G. Reed, Adjuvants for human vaccines, Curr. Opin. Immunol, vol.24, pp.310-315, 2012.

J. K. Bohannon, A. Hernandez, P. Enkhbaatar, W. L. Adams, and E. R. Sherwood, The immunobiology of toll-like receptor 4 agonists: from endotoxin tolerance to immunoadjuvants, Shock, vol.40, pp.451-462, 2013.

K. Johansen, U. Schröder, and L. Svensson, Immunogenicity and protective efficacy of a formalin-inactivated rotavirus vaccine combined with lipid adjuvants, Vaccine, vol.21, pp.368-375, 2003.

V. Snoeck, I. R. Peters, and E. Cox, The IgA system: a comparison of structure and function in different species, Vet. Res, vol.37, pp.455-467, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00903033

M. A. Otten and M. Van-egmond, The Fc receptor for IgA (Fc?RI, CD89), Immunol. Lett, vol.92, pp.23-31, 2004.

A. Cerutti, The regulation of IgA class switching, Nat. Rev. Immunol, vol.8, pp.421-434, 2008.

M. F. Bachmann and A. Oxenius, Interleukin 2: from immunostimulation to immunoregulation and back again, EMBO Rep, vol.8, pp.1142-1148, 2007.

G. E. Kaiko, J. C. Horvat, K. W. Beagley, and P. M. Hansbro, Immunological decisionmaking: how does the immune system decide to mount a helper T-cell response?, Immunology, vol.123, pp.326-338, 2008.

J. R. Schoenborn and C. B. Wilson, Regulation of interferon-gamma during innate and adaptive immune responses, Adv. Immunol, vol.96, pp.41-101, 2007.

J. J. Glass, S. J. Kent, and R. De, Enhancing dendritic cell activation and HIV vaccine effectiveness through nanoparticle vaccination, Expert. Rev. Vaccin, vol.15, pp.719-729, 2016.

S. M. Bal, S. Hortensius, Z. Ding, W. Jiskoot, and J. A. Bouwstra, Co-encapsulation of antigen and Toll-like receptor ligand in cationic liposomes affects the quality of the immune response in mice after intradermal vaccination, Vaccine, vol.29, pp.1045-1052, 2011.

P. O. Ilyinskii, Adjuvant-carrying synthetic vaccine particles augment the immune response to encapsulated antigen and exhibit strong local immune activation without inducing systemic cytokine release, Vaccine, vol.32, pp.2882-2895, 2014.

M. O. Mohsen, Delivering adjuvants and antigens in separate nanoparticles eliminates the need of physical linkage for effective vaccination, J. Control. Release, vol.251, pp.92-100, 2017.

B. Ling, SIV(mac) pathogenesis in rhesus macaques of Chinese and Indian origin compared with primary HIV infections in humans, AIDS Lond. Engl, vol.16, pp.1489-1496, 2002.

A. J. Bett, Comparison of T cell immune responses induced by vectored HIV vaccines in non-human primates and humans, Vaccine, vol.28, pp.7881-7889, 2010.

T. W. Phares, Rhesus macaque and mouse models for down-selecting circumsporozoite protein based malaria vaccines differ significantly in immunogenicity and functional outcomes, Malar. J, vol.16, p.115, 2017.

D. Mohanan, Administration routes affect the quality of immune responses: a cross-sectional evaluation of particulate antigen-delivery systems, J. Control. Release Off. J. Control. Release Soc, vol.147, pp.342-349, 2010.

N. Dereuddre-bosquet, HIV specific responses induced in nonhuman primates with ANRS HIV-Lipo-5 vaccine combined with rMVA-HIV prime or boost immunizations, Vaccine, vol.33, pp.2354-2359, 2015.

A. Nanda, Immunogenicity of recombinant fiber-chimeric adenovirus serotype 35 vector-based vaccines in mice and rhesus monkeys, J. Virol, vol.79, pp.14161-14168, 2005.

F. P. Navarro, Lipid nanoparticle vectorization of indocyanine green improves fluorescence imaging for tumor diagnosis and lymph node resection, J. Biomed. Nanotechnol, vol.8, pp.730-741, 2012.

P. C. Demuth, J. J. Moon, H. Suh, P. T. Hammond, and D. J. Irvine, Releasable layerby-layer assembly of stabilized lipid nanocapsules on microneedles for enhanced transcutaneous vaccine delivery, ACS Nano, vol.6, pp.8041-8051, 2012.

T. Delmas, H. Piraux, and A. Couffin, How to prepare and stabilize very small nanoemulsions, Langmuir, vol.27, pp.1683-1692, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00605087

S. Elsen, Cryptic O 2 -generating NADPH oxidase in dendritic cells, J. Cell. Sci, vol.117, pp.2215-2226, 2004.

C. Villiers, H. Freitas, R. Couderc, M. Villiers, and P. Marche, Analysis of the toxicity of gold nano particles on the immune system: effect on dendritic cell functions, J. Nanopart. Res. Interdiscip. Forum Nanoscale Sci. Technol, vol.12, pp.55-60, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00458282

N. D. Russell, M. G. Hudgens, R. Ha, C. Havenar-daughton, and M. J. Mcelrath, Moving to human immunodeficiency virus type 1 vaccine efficacy trials: defining T cell responses as potential correlates of immunity, J. Infect. Dis, vol.187, pp.226-242, 2003.

H. Streeck, N. Frahm, and B. D. Walker, The role of IFN-gamma elispot assay in HIV vaccine research, Nat. Protoc, vol.4, pp.461-469, 2009.

B. J. Macatangay, Comparison of immunologic assays for detecting immune responses in HIV immunotherapeutic studies: AIDS clinical trials group trial A5181, Clin. Vaccin. Immunol. CVI, vol.17, pp.1452-1459, 2010.

R. L. Felts, 3D visualization of HIV transfer at the virological synapse between dendritic cells and T cells, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.13336-13341, 2010.

. Blausen, com staff & staff, B. com. Medical gallery of Blausen medical, WikiJournal Med, vol.1, p.10, 2014.