F. Barré-sinoussi, A. L. Ross, and J. Delfraissy, Past, present and future: 30 years of HIV research, Nat. Rev. Microbiol, vol.11, pp.877-883, 2013.

, International AIDS Society Scientific Working Group on HIV Cure

B. Autran, M. Berkhout, S. Benkirane, N. Cairns, T. Chomont et al.,

M. Margolis, J. Markowitz, J. I. Martinez-picado, J. Mullins, S. Mellors et al., Towards an HIV cure: a global scientific strategy, Nat. Rev. Immunol, vol.12, pp.607-614, 2012.

J. D. Siliciano and R. F. Siliciano, Recent developments in the effort to cure HIV infection: going beyond N = 1, J. Clin. Invest, vol.126, pp.409-414, 2016.

G. Maartens, C. Celum, and S. R. Lewin, HIV infection: epidemiology, pathogenesis, treatment, and prevention, Lancet, vol.384, pp.60164-60165, 2014.

H. M. Delagrèverie, C. Delaugerre, S. R. Lewin, S. G. Deeks, and J. Z. Li, Ongoing Clinical Trials of Human Immunodeficiency Virus Latency-Reversing and Immunomodulatory Agents, Open Forum Infect. Dis, vol.3, p.189, 2016.

A. Isaacs and J. Lindenmann, Virus interference. I. The interferon, Proc. R. Soc. Lond. B Biol. Sci, vol.147, pp.258-267, 1957.

M. R. Capobianchi, E. Uleri, C. Caglioti, and A. Dolei, Type I IFN family members: similarity, differences and interaction, Cytokine Growth Factor Rev, vol.26, pp.103-111, 2015.

J. I. Gallin, J. M. Farber, S. M. Holland, and T. B. Nutman, Interferon-gamma in the management of infectious diseases, Ann. Intern. Med, vol.123, pp.216-224, 1995.

S. V. Kotenko, G. Gallagher, V. V. Baurin, A. Lewis-antes, M. Shen et al.,

F. Langer, H. Sheikh, R. P. Dickensheets, and . Donnelly, IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex, Nat. Immunol, vol.4, pp.69-77, 2003.

P. Sheppard, W. Kindsvogel, W. Xu, K. Henderson, S. Schlutsmeyer et al.,

C. Mcknight, D. Clegg, K. M. Foster, and . Klucher, IL-28, IL-29 and their class II cytokine receptor IL28R, Nat. Immunol, vol.4, pp.63-68, 2003.

E. A. Hemann, M. Gale, and R. Savan, Interferon Lambda Genetics and Biology in Regulation of Viral Control, Front. Immunol, vol.8, p.1707, 2017.

T. Doyle, C. Goujon, and M. H. Malim, HIV-1 and interferons: who's interfering with whom?, Nat. Rev. Microbiol, vol.13, pp.403-413, 2015.

W. M. Schneider, M. D. Chevillotte, and C. M. Rice, Interferon-stimulated genes: a complex web of host defenses, Annu. Rev. Immunol, vol.32, pp.513-545, 2014.

A. Murira and A. Lamarre, Type-I Interferon Responses: From Friend to Foe in the Battle against Chronic Viral Infection, Front. Immunol, vol.7, p.609, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01534446

M. J. De-veer, M. Holko, M. Frevel, E. Walker, S. Der et al., Functional classification of interferon-stimulated genes identified using microarrays, J. Leukoc. Biol, vol.69, pp.912-920, 2001.

O. Falade-nwulia, C. Suarez-cuervo, D. R. Nelson, M. W. Fried, J. B. Segal et al., Oral Direct-Acting Agent Therapy for Hepatitis C Virus Infection: A Systematic Review, Ann. Intern. Med, vol.166, pp.637-648, 2017.

D. S. Reich, C. F. Lucchinetti, and P. A. Calabresi, Multiple Sclerosis, N. Engl. J. Med, vol.378, pp.169-180, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01480156

T. S. Sandstrom, N. Ranganath, and J. B. , Angel, Impairment of the type I interferon response by HIV-1: Potential targets for HIV eradication, Cytokine Growth Factor Rev, vol.37, pp.1-16, 2017.

R. De-wit, J. K. Schattenkerk, C. A. Boucher, P. J. Bakker, K. H. Veenhof et al., Clinical and virological effects of high-dose recombinant interferon-alpha in disseminated AIDS-related Kaposi's sarcoma, Lancet Lond. Engl, vol.2, pp.1214-1217, 1988.

H. C. Lane, J. A. Kovacs, J. Feinberg, B. Herpin, V. Davey et al.,

M. Metcalf, N. Baseler, and . Salzman, Anti-retroviral effects of interferon-alpha in AIDS-associated Kaposi's sarcoma, Lancet Lond. Engl, vol.2, pp.1218-1222, 1988.

H. C. Lane, V. Davey, J. A. Kovacs, J. Feinberg, J. A. Metcalf et al., Interferon-alpha in patients with asymptomatic human immunodeficiency virus (HIV) infection. A randomized, placebo-controlled trial, Ann. Intern. Med, vol.112, pp.805-811, 1990.

J. Rivero, M. Limonta, A. Aguilera, M. Fraga, and P. L. Saura, Use of recombinant interferon-alpha in human immunodeficiency virus (HIV)-infected individuals, Biotherapy Dordr. Neth, vol.8, pp.23-31, 1994.

J. Rivero, M. Fraga, I. Cancio, J. Cuervo, and P. López-saura, Long-term treatment with recombinant interferon alpha-2b prolongs survival of asymptomatic HIV-infected individuals, Biotherapy Dordr. Neth, vol.10, pp.107-113, 1997.

D. R. Skillman, J. L. Malone, C. F. Decker, K. F. Wagner, R. L. Mapou et al., Phase I trial of interferon alfa-n3 in early-stage human immunodeficiency virus type 1 disease: evidence for drug safety, tolerance, and antiviral activity, J. Infect. Dis, vol.173, pp.1107-1114, 1996.

G. J. Williams and C. B. Colby, Recombinant human interferon-beta suppresses the replication of HIV and acts synergistically with AZT, J. Interferon Res, vol.9, pp.709-718, 1989.

B. Michaelis and J. A. Levy, HIV replication can be blocked by recombinant human interferon beta, AIDS Lond. Engl, vol.3, pp.27-31, 1989.

S. Oka, Y. Hirabayashi, H. Mouri, S. Sakurada, H. Goto et al., Beta-interferon and early stage HIV infection, J. Acquir. Immune Defic. Syndr, vol.2, pp.125-128, 1989.

S. A. Miles, H. J. Wang, E. Cortes, J. Carden, S. Marcus et al., Beta-interferon therapy in patients with poor-prognosis Kaposi sarcoma related to the acquired immunodeficiency syndrome (AIDS). A phase II trial with preliminary evidence of antiviral activity and low incidence of opportunistic infections, Ann. Intern. Med, vol.112, pp.582-589, 1990.

R. L. Krigel, K. A. Padavic-shaller, A. R. Rudolph, B. J. Poiesz, and R. L. Comis, Exacerbation of epidemic Kaposi's sarcoma with a combination of interleukin-2 and beta-interferon: results of a phase 2 study, J. Biol. Response Mod, vol.8, pp.359-365, 1989.

S. Miles, A. Levine, M. Feldstein, J. Carden, S. Cabriallas et al., Open-label phase I study of combination therapy with zidovudine and interferon-beta in patients with AIDS-related Kaposi's sarcoma: AIDS Clinical Trials Group Protocol 057, Cytokines Cell. Mol. Ther, vol.4, pp.17-23, 1998.

W. M. El-sadr, J. D. Lundgren, J. D. Neaton, F. Gordin, D. Abrams et al., CD4+ count-guided interruption of antiretroviral treatment, vol.355, pp.2283-2296, 2006.

T. W. Chun, D. Engel, S. B. Mizell, L. A. Ehler, and A. S. Fauci, Induction of HIV-1 replication in latently infected CD4+ T cells using a combination of cytokines, J. Exp. Med, vol.188, pp.83-91, 1998.

J. D. Siliciano and R. F. Siliciano, Recent developments in the search for a cure for HIV-1 infection: targeting the latent reservoir for HIV-1, J. Allergy Clin. Immunol, vol.134, pp.12-19, 2014.

M. W. Fried, M. L. Shiffman, K. R. Reddy, C. Smith, G. Marinos et al., Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection, N. Engl. J. Med, vol.347, pp.975-982, 2002.

H. L. Chan, N. W. Leung, and A. Y. Hui,

L. C. Chan, Y. Hung, J. S. Lee, C. W. Tam, J. J. Lam et al., A randomized, controlled trial of combination therapy for chronic hepatitis B: comparing pegylated interferon-alpha2b and lamivudine with lamivudine alone, Ann. Intern. Med, vol.142, pp.240-250, 2005.

D. M. Asmuth, R. L. Murphy, S. L. Rosenkranz, J. J. Lertora, S. Kottilil et al.,

R. T. Chan, C. R. Schooley, N. Rinaldo, X. Thielman, S. M. Li et al., Safety, tolerability, and mechanisms of antiretroviral activity of pegylated interferon Alfa-2a in HIV-1monoinfected participants: a phase II clinical trial, AIDS Clinical Trials Group A5192 Team, vol.201, pp.1686-1696, 2010.

S. S. Iyer, F. Bibollet-ruche, S. Sherrill-mix, G. H. Learn, L. Plenderleith et al., Resistance to type 1 interferons is a major determinant of HIV-1 transmission fitness, Proc. Natl. Acad. Sci. U. S. A, vol.114, pp.590-599, 2017.

F. Dianzani, G. Rozera, I. Abbate, G. D'offizi, A. Abdeddaim et al., Interferon may prevent HIV viral rebound after HAART interruption in HIV patients, J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res, vol.28, pp.1-3, 2008.
DOI : 10.1089/jir.2007.0076

F. Boué, J. Reynes, C. Rouzioux, D. Emilie, F. Souala et al., Alpha interferon administration during structured interruptions of combination antiretroviral therapy in patients with chronic HIV-1 infection: INTERVAC ANRS 105 trial, AIDS Lond. Engl, vol.25, pp.115-118, 2011.

C. Goujard, D. Emilie, C. Roussillon, V. Godot, C. Rouzioux et al., Continuous versus intermittent treatment strategies during primary HIV-1 infection: the randomized ANRS INTERPRIM Trial, vol.26, pp.1895-1905, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00875175

L. Azzoni, A. S. Foulkes, E. Papasavvas, A. M. Mexas, K. M. Lynn et al., Pegylated Interferon alfa-2a monotherapy results in suppression of HIV type 1 replication and decreased cell-associated HIV DNA integration, J. Infect. Dis, vol.207, pp.213-222, 2013.

H. Sun, M. J. Buzon, A. Shaw, R. K. Berg, X. G. Yu et al., Hepatitis C therapy with interferon-? and ribavirin reduces CD4 Tcell-associated HIV-1 DNA in HIV-1/hepatitis C virus-coinfected patients, J. Infect. Dis, vol.209, pp.1315-1320, 2014.
DOI : 10.1093/infdis/jit628

URL : https://academic.oup.com/jid/article-pdf/209/9/1315/18069917/jit628.pdf

Y. Jiao, W. Weng, Q. Gao, W. Zhu, W. Cai et al., Hepatitis C therapy with interferon-? and ribavirin reduces the CD4 cell count and the total, 2LTR circular and integrated HIV-1 DNA in HIV/HCV co-infected patients, Antiviral Res, vol.118, pp.118-122, 2015.

N. Huot, P. Rascle, T. Garcia-tellez, B. Jacquelin, and M. Müller-trutwin, Innate immune cell responses in non pathogenic versus pathogenic SIV infections, Curr. Opin. Virol, vol.19, pp.37-44, 2016.
DOI : 10.1016/j.coviro.2016.06.011

URL : https://hal.archives-ouvertes.fr/pasteur-01419560

T. Garcia-tellez, N. Huot, M. J. Ploquin, P. Rascle, B. Jacquelin et al., Non-human primates in HIV research: Achievements, limits and alternatives, vol.46, pp.324-332, 2016.
DOI : 10.1016/j.meegid.2016.07.012

URL : https://hal.archives-ouvertes.fr/pasteur-01419521

S. E. Bosinger, Q. Li, S. N. Gordon, N. R. Klatt, L. Duan et al., Global genomic analysis reveals rapid control of a robust innate response in SIV-infected sooty mangabeys, J. Clin. Invest, vol.119, pp.3556-3572, 2009.

B. Jacquelin, V. Mayau, B. Targat, A. Liovat, D. Kunkel et al., Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response, J. Clin. Invest, vol.119, pp.3544-3555, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-01968918

L. D. Harris, B. Tabb, D. L. Sodora, M. Paiardini, N. R. Klatt et al., Downregulation of robust acute type I interferon responses distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection of natural hosts from pathogenic SIV infection of rhesus macaques, J. Virol, vol.84, pp.7886-7891, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01968882

B. Jacquelin, G. Petitjean, D. Kunkel, A. Liovat, S. P. Jochems et al., Innate immune responses and rapid control of inflammation in African green monkeys treated or not with interferon-alpha during primary SIVagm infection, PLoS Pathog, vol.10, p.1004241, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01960800

T. H. Vanderford, C. Slichter, K. A. Rogers, B. O. Lawson, R. Obaede et al., Treatment of SIV-infected sooty mangabeys with a type-I IFN agonist results in decreased virus replication without inducing hyperimmune activation, Blood, vol.119, pp.5750-5757, 2012.

N. G. Sandler, S. E. Bosinger, J. D. Estes, R. T. Zhu, G. K. Tharp et al.,

J. D. Langer, G. Lifson, D. C. Schreiber, and . Douek, Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression, Nature, vol.511, pp.601-605, 2014.

S. Fernandez, S. Tanaskovic, K. Helbig, R. Rajasuriar, M. Kramski et al., CD4+ T-cell deficiency in HIV patients responding to antiretroviral therapy is associated with increased expression of interferonstimulated genes in CD4+ T cells, J. Infect. Dis, vol.204, pp.1927-1935, 2011.

G. A. Hardy, S. Sieg, B. Rodriguez, D. Anthony, R. Asaad et al.,

N. T. Schacker, H. A. Funderburg, R. Pilch-cooper, R. L. Debernardo, M. M. Rabin et al., Interferon-? is the primary plasma type-I IFN in HIV-1 infection and correlates with immune activation and disease markers, PloS One, vol.8, p.56527, 2013.

N. Noel, F. Boufassa, C. Lécuroux, A. Saez-cirion, C. Bourgeois et al., Elevated IP10 levels are associated with immune activation and low CD4 + T-cell counts in HIV controller patients, AIDS Lond. Engl, vol.28, pp.467-476, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01420524

M. J. Ploquin, Y. Madec, A. Casrouge, N. Huot, C. Passaes et al., Elevated Basal Pre-infection CXCL10 in Plasma and in the Small Intestine after Infection Are Associated with More Rapid HIV/SIV Disease Onset, vol.12, p.1005774, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01380604

E. Canoui, N. Noël, C. Lécuroux, F. Boufassa, A. Sáez-cirión et al., Strong ifitm1 Expression in CD4 T Cells in HIV Controllers Is Correlated With Immune Activation, J. Acquir. Immune Defic. Syndr, vol.74, pp.56-59, 1999.
URL : https://hal.archives-ouvertes.fr/pasteur-01420412

C. Lehmann, M. Lafferty, A. Garzino-demo, N. Jung, P. Hartmann et al., Plasmacytoid dendritic cells accumulate and secrete interferon alpha in lymph nodes of HIV-1 patients, PloS One, vol.5, p.11110, 2010.

M. Paiardini and M. Müller-trutwin, HIV-associated chronic immune activation, Immunol. Rev, vol.254, pp.78-101, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-02017999

G. A. Hardy, S. F. Sieg, B. Rodriguez, W. Jiang, R. Asaad et al.,

. Harding, Desensitization to type I interferon in HIV-1 infection correlates with markers of immune activation and disease progression, Blood, vol.113, pp.5497-5505, 2009.

A. Yadav, P. Fitzgerald, M. M. Sajadi, B. Gilliam, M. K. Lafferty et al., Increased expression of suppressor of cytokine signaling-1 (SOCS-1): A mechanism for dysregulated T helper-1 responses in HIV-1 disease, Virology, vol.385, pp.126-133, 2009.

L. N. Akhtar, H. Qin, M. T. Muldowney, L. L. Yanagisawa, O. Kutsch et al.,

. Benveniste, Suppressor of cytokine signaling 3 inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages, J. Immunol. Baltim. Md, vol.185, pp.2393-2404, 1950.

N. Ranganath, T. S. Sandstrom, S. Fadel, S. C. Côté, and J. B. , Angel, Type I interferon 33 responses are impaired in latently HIV infected cells, Retrovirology, vol.13, p.66, 2016.

M. P. Hardy, C. M. Owczarek, L. S. Jermiin, M. Ejdebäck, and P. J. Hertzog, Characterization of the type I interferon locus and identification of novel genes, Genomics, vol.84, pp.331-345, 2004.

C. Thomas, I. Moraga, D. Levin, P. O. Krutzik, Y. Podoplelova et al.,

S. E. Yarden, J. S. Vleck, G. P. Glenn, J. Nolan, G. Piehler et al., Structural linkage between ligand discrimination and receptor activation by type I interferons, Cell, vol.146, pp.621-632, 2011.

E. Jaks, M. Gavutis, G. Uzé, J. Martal, and J. Piehler, Differential receptor subunit affinities of type I interferons govern differential signal activation, J. Mol. Biol, vol.366, pp.525-539, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00167456

K. Gibbert, J. J. Joedicke, A. Meryk, M. Trilling, S. Francois et al., Interferon-alpha subtype 11 activates NK cells and enables control of retroviral infection, PLoS Pathog, vol.8, p.1002868, 2012.

W. S. Yeow, C. M. Lai, and M. W. Beilharz, The in vivo expression patterns of individual type I interferon genes in murine cytomegalovirus infections, Antiviral Res, vol.34, pp.17-26, 1997.

N. Gerlach, K. Gibbert, C. Alter, S. Nair, G. Zelinskyy et al., Antiretroviral effects of type I IFN subtypes in vivo, Eur. J. Immunol, vol.39, pp.136-146, 2009.

M. S. Harper, K. Guo, K. Gibbert, E. J. Lee, S. M. Dillon et al., Interferon-? Subtypes in an Ex Vivo Model of Acute HIV-1 Infection: Expression, Potency and Effector Mechanisms, PLoS Pathog, vol.11, p.1005254, 2015.

K. J. Lavender, K. Gibbert, K. E. Peterson, E. Van-dis, S. Francois et al.,

. Dittmer, Interferon Alpha Subtype-Specific Suppression of HIV-1 Infection In Vivo, J. Virol, vol.90, pp.6001-6013, 2016.

S. Abraham, J. Choi, N. M. Ortega, J. Zhang, P. Shankar et al., Gene therapy with plasmids encoding IFN-? or IFN-?14 confers long-term resistance to HIV-1 in humanized mice, Oncotarget, vol.7, pp.78412-78420, 2016.

K. Abel, M. J. Alegria-hartman, K. Rothaeusler, M. Marthas, and C. J. Miller, The relationship between simian immunodeficiency virus RNA levels and the mRNA levels of alpha/beta interferons (IFN-alpha/beta) and IFN-alpha/beta-inducible Mx in lymphoid tissues of rhesus macaques during acute and chronic infection, J. Virol, vol.76, pp.8433-8445, 2002.

C. Lehmann, D. Taubert, N. Jung, G. Fätkenheuer, J. Van-lunzen et al., Preferential upregulation of interferon-alpha subtype 2 expression in HIV-1 patients, AIDS Res. Hum. Retroviruses, vol.25, pp.577-581, 2009.

J. Dutrieux, V. Fabre-mersseman, B. Charmeteau-de-muylder, M. Rancez, R. Ponte et al., Modified interferon-? subtypes production and chemokine networks in the thymus during acute simian immunodeficiency virus infection, AIDS Lond. Engl, vol.28, pp.1101-1113, 2014.

Y. Li, B. Sun, S. Esser, H. Jessen, H. Streeck et al., Expression Pattern of Individual IFNA Subtypes in Chronic HIV Infection, J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res, vol.37, pp.541-549, 2017.

M. P. Rodero, J. Decalf, V. Bondet, D. Hunt, G. I. Rice et al., Detection of interferon alpha protein reveals differential levels and cellular sources in disease, J. Exp. Med, vol.214, pp.1547-1555, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01534181

R. S. Veazey, H. A. Pilch-cooper, T. J. Hope, G. Alter, A. M. Carias et al., Prevention of SHIV transmission by topical IFN-? treatment, Mucosal Immunol, vol.9, pp.1528-1536, 2016.

Y. Wang, K. Abel, K. Lantz, A. M. Krieg, M. B. Mcchesney et al., The Toll-like receptor 7 (TLR7) agonist, imiquimod, and the TLR9 agonist, CpG ODN, induce antiviral cytokines and chemokines but do not prevent vaginal transmission of simian immunodeficiency virus when applied intravaginally to rhesus macaques, J. Virol, vol.79, pp.14355-14370, 2005.

K. Y. Fung, N. E. Mangan, H. Cumming, J. C. Horvat, J. R. Mayall et al., Interferon-? protects the female reproductive tract from viral and bacterial infection, Science, vol.339, pp.1088-1092, 2013.

S. A. Stifter, A. Y. Matthews, N. E. Mangan, K. Y. Fung, A. Drew et al., Defining the distinct, intrinsic properties of the novel type I interferon, epsilon, J. Biol. Chem, 2017.

A. Garcia-minambres, S. G. Eid, N. E. Mangan, C. Pade, S. S. Lim et al., Interferon epsilon promotes HIV restriction at multiple steps of viral replication, Immunol. Cell Biol, vol.95, pp.478-483, 2017.

C. Tasker, S. Subbian, P. Gao, J. Couret, C. Levine et al., , p.35

N. Landau, W. Lu, and T. L. Chang, IFN-?protects primary macrophages against HIV infection, JCI Insight, vol.1, p.88255, 2016.

J. Pott, T. Mahlakõiv, M. Mordstein, C. U. Duerr, T. Michiels et al., IFN-lambda determines the intestinal epithelial antiviral host defense, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.7944-7949, 2011.

M. T. Baldridge, S. Lee, J. J. Brown, N. Mcallister, K. Urbanek et al., Expression ofIfnlr1on Intestinal Epithelial Cells Is Critical to the Antiviral Effects of Interferon Lambda against Norovirus and Reovirus, J. Virol, p.91, 2017.

H. M. Lazear, T. J. Nice, and M. S. Diamond, Interferon-?: Immune Functions at Barrier Surfaces and Beyond, Immunity, vol.43, pp.15-28, 2015.

W. Hou, X. Wang, L. Ye, L. Zhou, Z. Yang et al., Lambda interferon inhibits human immunodeficiency virus type 1 infection of macrophages, J. Virol, vol.83, pp.3834-3842, 2009.

M. Liu, D. Zhou, X. Wang, W. Zhou, L. Ye et al., IFN-?3 inhibits HIV infection of macrophages through the JAK-STAT pathway, PloS One, vol.7, p.35902, 2012.

Y. Wang, J. Li, X. Wang, Y. Zhou, T. Zhang et al., Comparison of antiviral activity of lambda-interferons against HIV replication in macrophages, J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res, vol.35, pp.213-221, 2015.

R. Tian, H. Guo, J. Wei, C. Yang, S. He et al., IFN-? inhibits HIV-1 integration and post-transcriptional events in vitro, but there is only limited in vivo repression of viral production, Antiviral Res, vol.95, pp.57-65, 2012.

J. Li, Y. Wang, X. Wang, L. Ye, Y. Zhou et al., Immune activation of human brain microvascular endothelial cells inhibits HIV replication in macrophages, Blood, vol.121, pp.2934-2942, 2013.

X. Wang, H. Wang, M. Liu, J. Li, R. Zhou et al.,

. Ho, IFN-? Inhibits Drug-Resistant HIV Infection of Macrophages, Front. Immunol, vol.8, p.210, 2017.

S. Phillips, S. Mistry, A. Riva, H. Cooksley, T. Hadzhiolova-lebeau et al., Peg-Interferon Lambda Treatment Induces Robust Innate and Adaptive Immunity in Chronic Hepatitis B Patients, Front. Immunol, vol.8, p.621, 2017.

J. Friborg, S. Levine, C. Chen, A. K. Sheaffer, S. Chaniewski et al.,

. Mcphee, Combinations of lambda interferon with direct-acting antiviral agents are highly efficient in suppressing hepatitis C virus replication, Antimicrob. Agents Chemother, vol.57, pp.1312-1322, 2013.

M. Nelson, R. Rubio, A. Lazzarin, S. Romanova, A. Luetkemeyer et al.,

D. Molina, S. Xu, S. Srinivasan, and . Portsmouth, Safety and Efficacy of Pegylated Interferon Lambda, Ribavirin, and Daclatasvir in HCV and HIV-Coinfected Patients, J. Interferon Cytokine Res. Off. J. Int. Soc. Interferon Cytokine Res, vol.37, pp.103-111, 2017.

R. J. Gilson, J. L. Shupack, A. E. Friedman-kien, M. A. Conant, J. N. Weber et al., A randomized, controlled, safety study using imiquimod for the topical treatment of anogenital warts in HIV-infected patients, AIDS Lond. Engl, vol.13, pp.2397-2404, 1999.

P. A. Fox, M. Nathan, N. Francis, N. Singh, J. Weir et al., A double-blind, randomized controlled trial of the use of imiquimod cream for the treatment of anal canal high-grade anal intraepithelial neoplasia in HIV-positive MSM on HAART, with long-term follow-up data including the use of open-label imiquimod, AIDS Lond. Engl, vol.24, pp.2331-2335, 2010.

H. Hofmann, B. Vanwalscappel, N. Bloch, and N. R. Landau, TLR7/8 agonist induces a postentry SAMHD1-independent block to HIV-1 infection of monocytes, Retrovirology, vol.13, p.83, 2016.

P. F. Mckay, D. F. King, J. F. Mann, G. Barinaga, D. Carter et al., TLR4 and TLR7/8 Adjuvant Combinations Generate Different Vaccine Antigen-Specific Immune Outcomes in Minipigs when Administered via the ID or IN Routes, PloS One, vol.11, p.148984, 2016.

E. Poteet, P. Lewis, C. Chen, S. O. Ho, T. Do et al., Toll-like receptor 3 adjuvant in combination with virus-like particles elicit a humoral response against HIV, Vaccine, vol.34, pp.5886-5894, 2016.

I. Rebbapragada, G. Birkus, J. Perry, W. Xing, H. Kwon et al., Molecular Determinants of GS-9620-Dependent TLR7 Activation, PloS One, vol.11, p.146835, 2016.

R. A. Bam, D. Hansen, A. Irrinki, A. Mulato, G. S. Jones et al., TLR7 Agonist GS-9620 Is a Potent Inhibitor of Acute HIV-1 Infection in Human Peripheral Blood Mononuclear Cells, Antimicrob. Agents Chemother, p.61, 2017.

A. Tsai, A. Irrinki, J. Kaur, T. Cihlar, G. Kukolj et al., Toll-Like Receptor

, Agonist GS-9620 Induces HIV Expression and HIV-Specific Immunity in Cells from HIVInfected Individuals on Suppressive Antiretroviral Therapy, p.91, 2017.

R. Offersen, S. K. Nissen, T. A. Rasmussen, L. Østergaard, P. W. Denton et al., A Novel Toll-Like Receptor 9 Agonist, MGN1703, Enhances HIV-1 Transcription and NK Cell-Mediated Inhibition of HIV-1-Infected Autologous CD4+ T Cells, J. Virol, vol.90, pp.4441-4453, 2016.

L. Vibholm, M. H. Schleimann, J. F. Højen, T. Benfield, R. Offersen et al., Short-Course Toll-Like Receptor 9 Agonist Treatment Impacts Innate Immunity and Plasma Viremia in Individuals With Human Immunodeficiency Virus Infection, vol.64, pp.1686-1695, 2017.

A. R. Krarup, M. Abdel-mohsen, M. H. Schleimann, L. Vibholm, P. A. Engen et al.,

S. K. Søgaard, M. Pillai, P. W. Tolstrup, and . Denton, The TLR9 agonist MGN1703 triggers a potent type I interferon response in the sigmoid colon, Mucosal Immunol, 2017.

P. W. Hunt, J. Brenchley, E. Sinclair, J. M. Mccune, M. Roland et al., Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy, J. Infect. Dis, vol.197, pp.126-133, 2008.

D. R. Boulware, K. H. Hullsiek, C. E. Puronen, A. Rupert, J. V. Baker et al., Higher levels of CRP, D-dimer, IL-6, and hyaluronic acid before initiation of antiretroviral therapy (ART) are associated with increased risk of AIDS or death, J. Infect. Dis, vol.203, pp.1637-1646, 2011.

Á. H. Borges, M. J. Silverberg, D. Wentworth, A. E. Grulich, G. Fätkenheuer et al., Predicting risk of cancer during HIV infection: the role of inflammatory and coagulation biomarkers, AIDS Lond. Engl, vol.27, pp.1433-1441, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01101192

A. D. Nordell, M. Mckenna, Á. H. Borges, D. Duprez, J. Neuhaus et al., ESPRIT Study Groups, SILCAAT Scientific Committee, Severity of cardiovascular disease outcomes among patients with HIV is related to markers of inflammation and coagulation, J. Am. Heart Assoc, vol.3, p.844, 2014.

I. Sereti, S. J. Krebs, N. Phanuphak, J. L. Fletcher, B. Slike et al., RV254/SEARCH 010, RV304/SEARCH 013 and SEARCH 011 protocol teams, Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection, Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am, vol.64, pp.124-131, 2017.

N. Noel, N. Lerolle, C. Lécuroux, C. Goujard, A. Venet et al., Immunologic and Virologic Progression in HIV Controllers: The Role of Viral "Blips" and Immune Activation in the ANRS CO21 CODEX Study, PloS One, vol.10, p.131922, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01415218

E. H. Loughlin, J. B. Rice, H. S. Wells, I. Rappaport, and A. A. Joseph, The treatment of Plasmodium falciparum malaria with a single dose antimalarial; a preliminary report of the use of hydroxychloroquine, Antibiot. Chemother. Northfield Ill, vol.2, issue.4, pp.171-174, 1952.

N. Costedoat-chalumeau, L. Galicier, O. Aumaître, C. Francès, V. Le-guern et al.,

G. Piette and . Plus, Hydroxychloroquine in systemic lupus erythematosus: results of a French multicentre controlled trial (PLUS Study), Ann. Rheum. Dis, vol.72, pp.1786-1792, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00933279

V. Ravindran and G. Alias, Efficacy of combination DMARD therapy vs. hydroxychloroquine monotherapy in chronic persistent chikungunya arthritis: a 24-week randomized controlled open label study, Clin. Rheumatol, vol.36, pp.1335-1340, 2017.

J. A. Martinson, C. J. Montoya, X. Usuga, R. Ronquillo, A. L. Landay et al., Chloroquine modulates HIV-1-induced plasmacytoid dendritic cell alpha interferon: implication for T-cell activation, Antimicrob. Agents Chemother, vol.54, pp.871-881, 2010.

S. M. Murray, C. M. Down, D. R. Boulware, W. M. Stauffer, W. P. Cavert et al., Reduction of immune activation with chloroquine therapy during chronic HIV infection, J. Virol, vol.84, pp.12082-12086, 2010.

S. Piconi, S. Parisotto, G. Rizzardini, S. Passerini, R. Terzi et al., Hydroxychloroquine drastically reduces immune activation in HIV-infected, antiretroviral therapy-treated immunologic nonresponders, Blood, vol.118, pp.3263-3272, 2011.

J. Routy, J. B. Angel, M. Patel, C. Kanagaratham, D. Radzioch et al., Assessment of chloroquine as a modulator of immune activation to improve CD4 recovery in immune nonresponding HIV-infected patients receiving antiretroviral therapy, HIV Med, vol.16, pp.48-56, 2015.

N. I. Paton, R. L. Goodall, D. T. Dunn, S. Franzen, Y. Collaco-moraes et al.,

M. J. Williams, A. Fisher, J. Winston, C. Fox, E. A. Orkin et al., Hydroxychloroquine Trial Team, Effects of hydroxychloroquine on immune activation and disease progression among HIV-infected patients not receiving antiretroviral therapy: a randomized controlled trial, JAMA, vol.308, pp.353-361, 2012.

A. Zhen, V. Rezek, C. Youn, B. Lam, N. Chang et al., Targeting type I interferon-mediated activation restores immune function in chronic HIV infection, J. Clin. Invest, vol.127, pp.260-268, 2017.
DOI : 10.1172/jci89488

URL : http://www.jci.org/articles/view/89488/files/pdf

L. Cheng, J. Ma, J. Li, D. Li, G. Li et al., Blocking type I interferon signaling enhances T cell recovery and reduces HIV1 reservoirs, J. Clin. Invest, vol.127, pp.269-279, 2017.
DOI : 10.1172/jci90745

URL : http://www.jci.org/articles/view/90745/files/pdf

S. G. Deeks, P. M. Odorizzi, and R. Sekaly, The interferon paradox: can inhibiting an antiviral mechanism advance an HIV cure?, J. Clin. Invest, vol.127, pp.103-105, 2017.
DOI : 10.1172/jci91916

URL : http://www.jci.org/articles/view/91916/files/pdf

R. Furie, M. Khamashta, J. T. Merrill, V. P. Werth, K. Kalunian et al., CD1013 Study Investigators, Anifrolumab, an Anti-Interferon-? Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus, vol.69, pp.376-386, 2017.
DOI : 10.1002/art.39962

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/art.39962

J. J. Kohler, D. L. Tuttle, C. R. Coberley, J. W. Sleasman, and M. M. Goodenow, Human immunodeficiency virus type 1 (HIV-1) induces activation of multiple STATs in CD4+ cells of lymphocyte or monocyte/macrophage lineages, J. Leukoc. Biol, vol.73, pp.407-416, 2003.

C. Bovolenta, L. Camorali, A. L. Lorini, S. Ghezzi, E. Vicenzi et al., Constitutive activation of STATs upon in vivo human immunodeficiency virus infection, Blood, vol.94, pp.4202-4209, 1999.

A. Alhetheel, Y. Yakubtsov, K. Abdkader, N. Sant, F. Diaz-mitoma et al., Amplification of the signal transducer and activator of transcription I signaling pathway and its association with apoptosis in monocytes from HIV-infected patients, AIDS Lond. Engl, vol.22, pp.1137-1144, 2008.

Y. Takahashi, A. E. Mayne, L. Khowawisetsut, K. Pattanapanyasat, D. Little et al., In vivo administration of a JAK3 inhibitor to chronically siv infected rhesus macaques leads to NK cell depletion associated with transient modest increase in viral loads, PloS One, vol.8, p.70992, 2013.

Y. Takahashi, S. N. Byrareddy, C. Albrecht, M. Brameier, L. Walter et al.,

R. Dunbar, D. M. Russo, T. Little, L. Villinger, K. Khowawisetsut et al., In vivo administration of a JAK3 inhibitor during acute SIV infection leads to significant increases in viral load during chronic infection, PLoS Pathog, vol.10, p.1003929, 2014.

C. Gavegnano, M. Detorio, C. Montero, A. Bosque, V. Planelles et al., Ruxolitinib and tofacitinib are potent and selective inhibitors of HIV-1 replication and virus reactivation in vitro, Antimicrob. Agents Chemother, vol.58, pp.1977-1986, 2014.

W. B. Haile, C. Gavegnano, S. Tao, Y. Jiang, R. F. Schinazi et al., The Janus kinase inhibitor ruxolitinib reduces HIV replication in human macrophages and ameliorates HIV encephalitis in a murine model, Neurobiol. Dis, vol.92, pp.137-143, 2016.

A. M. Spivak, E. T. Larragoite, M. L. Coletti, A. B. Macedo, L. J. Martins et al., Janus kinase inhibition suppresses PKC-induced cytokine release without affecting HIV-1 latency reversal ex vivo, Retrovirology, vol.13, p.88, 2016.

C. Gavegnano, J. H. Brehm, F. P. Dupuy, A. Talla, S. P. Ribeiro et al., Novel mechanisms to inhibit HIV reservoir seeding using Jak inhibitors, PLoS Pathog, vol.13, p.1006740, 2017.
DOI : 10.1371/journal.ppat.1006740

URL : https://journals.plos.org/plospathogens/article/file?id=10.1371/journal.ppat.1006740&type=printable

L. Micci, J. Harper, S. Paganini, C. King, E. Ryan et al., Combined IL21 and IFN? treatment limits inflammation and delay viral rebound in ARTtreated, SIV infected macaques. Abstracts of the IAS HIV Cure and Cancer Forum, pp.4-5, 2017.
DOI : 10.1172/jci81400

URL : http://www.jci.org/articles/view/81400/files/pdf