HIV-1 Tat immunization restores immune homeostasis and attacks the HAART-resistant blood HIV DNA: results of a randomized phase II exploratory clinical trial

Fabrizio Ensoli, Aurelio Cafaro, Anna Casabianca, Antonella Tripiciano, Stefania Bellino, Olimpia Longo, Vittorio Francavilla, Orietta Picconi, Cecilia Sgadari, Sonia Moretti, Maria R Pavone Cossut, Angela Arancio, Chiara Orlandi, Leonardo Sernicola, Maria T Maggiorella, Giovanni Paniccia, Cristina Mussini, Adriano Lazzarin, Laura Sighinolfi, Guido Palamara, Andrea Gori, Gioacchino Angarano, Massimo Di Pietro, Massimo Galli, Vito S Mercurio, Francesco Castelli, Giovanni Di Perri, Paolo Monini, Mauro Magnani, Enrico Garaci, Barbara Ensoli, Fabrizio Ensoli, Aurelio Cafaro, Anna Casabianca, Antonella Tripiciano, Stefania Bellino, Olimpia Longo, Vittorio Francavilla, Orietta Picconi, Cecilia Sgadari, Sonia Moretti, Maria R Pavone Cossut, Angela Arancio, Chiara Orlandi, Leonardo Sernicola, Maria T Maggiorella, Giovanni Paniccia, Cristina Mussini, Adriano Lazzarin, Laura Sighinolfi, Guido Palamara, Andrea Gori, Gioacchino Angarano, Massimo Di Pietro, Massimo Galli, Vito S Mercurio, Francesco Castelli, Giovanni Di Perri, Paolo Monini, Mauro Magnani, Enrico Garaci, Barbara Ensoli

Abstract

Background: The phase II multicenter, randomized, open label, therapeutic trial (ISS T-002, Clinicaltrials.gov NCT00751595) was aimed at evaluating the immunogenicity and the safety of the biologically active HIV-1 Tat protein administered at 7.5 or 30 μg, given 3 or 5 times monthly, and at exploring immunological and virological disease biomarkers. The study duration was 48 weeks, however, vaccinees were followed until the last enrolled subject reached the 48 weeks. Reported are final data up to 144 weeks of follow-up. The ISS T-002 trial was conducted in 11 clinical centers in Italy on 168 HIV positive subjects under Highly Active Antiretroviral Therapy (HAART), anti-Tat Antibody (Ab) negative at baseline, with plasma viremia <50 copies/mL in the last 6 months prior to enrollment, and CD4(+) T-cell number ≥200 cells/μL. Subjects from a parallel observational study (ISS OBS T-002, Clinicaltrials.gov NCT0102455) enrolled at the same clinical sites with the same criteria constituted an external reference group to explore biomarkers of disease.

Results: The vaccine was safe and well tolerated and induced anti-Tat Abs in most patients (79%), with the highest frequency and durability in the Tat 30 μg groups (89%) particularly when given 3 times (92%). Vaccination promoted a durable and significant restoration of T, B, natural killer (NK) cells, and CD4(+) and CD8(+) central memory subsets. Moreover, a significant reduction of blood proviral DNA was seen after week 72, particularly under PI-based regimens and with Tat 30 μg given 3 times (30 μg, 3x), reaching a predicted 70% decay after 3 years from vaccination with a half-life of 88 weeks. This decay was significantly associated with anti-Tat IgM and IgG Abs and neutralization of Tat-mediated entry of oligomeric Env in dendritic cells, which predicted HIV-1 DNA decay. Finally, the 30 μg, 3x group was the only one showing significant increases of NK cells and CD38(+)HLA-DR(+)/CD8(+) T cells, a phenotype associated with increased killing activity in elite controllers.

Conclusions: Anti-Tat immune responses are needed to restore immune homeostasis and effective anti-viral responses capable of attacking the virus reservoir. Thus, Tat immunization represents a promising pathogenesis-driven intervention to intensify HAART efficacy.

Trial registration: ClinicalTrials.gov NCT01024556.

Figures

Figure 1
Figure 1
Study flow chart. Two hundred seventy-two HAART-treated patients were assessed for eligibility. One hundred three patients were excluded from the study (n = 94 for either not meeting the inclusion criteria or for meeting exclusion criteria; n = 8 declined to participate, and 1 was lost to follow-up). Thus, 169 subjects were randomized to one of the 4 treatment groups. One subject was excluded after randomization for concomitant treatment with a contraindicated medication, therefore 168 volunteers were allocated to intervention and analyzed for safety (safety population). Since 11 subjects (from the different treatment groups) received less than 3 immunizations and 2 subjects were non compliant with antiretroviral therapy, 155 subjects were considered for the analyses of immunogenicity (immunogenicity population).
Figure 2
Figure 2
Anti-Tat humoral immune response in vaccinees. (A) Percentage of subjects producing Anti-Tat Abs (responders) after Tat immunization (7.5 μg, 3x n = 40; 7.5 μg, 5x n = 40; 30 μg, 3x n = 38; 30 μg, 5x n = 37). (B) Kaplan-Meier estimates showing the cumulative probability of anti-Tat Ab durability in responders stratified according to treatment groups and up to week 144 of follow-up (n = 123, median follow-up of 96 weeks). (C) Percentage of subjects in each treatment group producing anti-Tat IgM (light blue), IgG (red), or IgA (white) Abs. (D) Percentage of subjects in each treatment group producing two (IgM and IgG, in blue) or three (IgM, IgG and IgA, in red) anti-Tat Ab classes. (E) Anti-Tat IgM (light blue) and IgA (white) peak titers between 4 and 24 weeks since the first immunization in subjects positive for IgM (7.5 μg, 3x n = 16; 7.5 μg, 5x n = 20; 30 μg, 3x n = 24; 30 μg, 5x n = 23) or IgA anti-Tat Abs (7.5 μg, 3x n = 7; 7.5 μg, 5x n = 9; 30 μg, 3x n = 19; 30 μg, 5x n = 22). (F) Anti-Tat IgG Ab peak titers between 4 and 24 weeks since the first immunization in subjects positive for IgG anti-Tat Abs (7.5 μg, 3x n = 24; 7.5 μg, 5x n = 24; 30 μg, 3x n = 32; 30 μg, 5x n = 31). Box plots represent the median, 25th and 75th percentile, with the minimum and maximum values; the outliers are not represented in the graphs.
Figure 3
Figure 3
Anti-Tat cellular immune response. IFN-γ, IL-2 or IL-4 production and CD4+ or CD8+ T cell proliferation to Tat in (A) vaccinees or (C) OBS subjects. The percentages of responders at baseline (white bars) and up to 48 weeks from baseline (blue bars) (vaccinees: IFN-γ, IL-2, or IL-4 n = 151, proliferation n = 140; OBS: IFN-γ, IL-2, or IL-4 n = 60, proliferation n = 54) are shown. Peak intensity of anti-Tat cellular responses in (B) vaccinees or (D) OBS subjects at baseline (white bars) and up to 48 weeks (blue bars) in responders (subjects with at least one positive response after baseline) (vaccinees: IFN-γ n = 39, IL-2 n = 38, IL-4 n = 12, CD4+ proliferation n = 86, CD8+ proliferation n = 77; OBS: IFN-γ n = 12, IL-2 n = 11, IL-4 n = 6, CD4+ proliferation n = 33, CD8+ proliferation n = 34). Box plots represent the median, 25th and 75th percentile, with the minimum and maximum values; the outliers are not represented in the graphs. The McNemar’s and Wilcoxon signed-rank tests were applied. P-values assess the values of frequencies and intensity, respectively, up to 48 weeks after immunization versus baseline values.
Figure 4
Figure 4
CD4+, CD8+, B and NK cell numbers. Changes from baseline of (A, B) CD4+, (C, D) CD8+, (E, F) B and (G, H) NK cells at years 1, 2 and 3 in vaccinees (left panels) (n = 152 at year 1 ; n = 114 at year 2 ; n = 69 at year 3) and in OBS subjects (right panels) (n = 79 at year 1 ; n = 42 at year 2 ; n = 30 at year 3), respectively. Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values.
Figure 5
Figure 5
CD4+, CD8+, B and NK cell numbers stratified by antiretroviral regimens. Changes from baseline of (A, B) CD4+, (C, D) CD8+, (E, F) B and (G, H) NK cells at years 1, 2 and 3 according to antiretroviral regimens in vaccinees (left panels) (NNRTI- or NRTI-based n = 104 at year 1 ; n = 80 at year 2 ; n = 49 at year 3; PI-based n = 48 at year 1 ; n = 34 at year 2 ; n = 20 at year 3) and OBS subjects (right panels) (NNRTI- or NRTI-based n = 49 at year 1 ; n = 21 at year 2 ; n = 14 at year 3; PI-based n = 30 at year 1 ; n = 20 at year 2 ; n = 16 at year 3). Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values, both overall and stratified by drug regimens (NNRTI/NRTI- or PI-based).
Figure 6
Figure 6
CD4+T cells stratified by CD4+nadir and CD4+/CD8+T cell ratio. Changes from baseline of (A) CD4+ by CD4+ nadir and (B) CD4+/CD8+ T cell ratio at years 1, 2 and 3 in vaccinees (left panels) (n = 152 at year 1 ; n = 114 at year 2 ; n = 69 at year 3) and in OBS subjects (right panels) (n = 79 at year 1 ; n = 42 at year 2 ; n = 30 at year 3), respectively. Vaccinees with CD4+ T cell nadir ≤250 cells/μl: n = 32, >250 cells/μl n = 120; OBS subjects with CD4+ T cell nadir ≤250 cells/μl: n = 45 ; >250 cells/μl n = 34. Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values.
Figure 7
Figure 7
Naïve, central and effector memory CD4+and CD8+T cells. Changes from baseline of naïve, Tem and Tcm (A, B) CD4+ and (C, D) CD8+ T cell percentage at years 1, 2 and 3 for vaccinees (left panels) (n = 94 at year 1 ; n = 75 at year 2 ; n = 32 at year 3) and OBS subjects (right panels) (n = 32 at year 1 ; n = 20 at year 2). Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values.
Figure 8
Figure 8
Naïve, central and effector memory CD4+and CD8+T cells stratified by antiretroviral regimens. Changes from baseline of naïve, Tem and Tcm (A, B) CD4+ and (C, D) CD8+ T-cell percentage at years 1, 2 and 3 according to antiretroviral regimens in vaccinees (left panels) (NNRTI- or NRTI-based n = 59 at year 1 ; n = 49 at year 2 ; n = 21 at year 3; PI-based n = 35 at year 1 ; n = 26 at year 2 ; n = 11 at year 3) and OBS subjects (right panels) (NNRTI- or NRTI-based n = 19 at year 1 ; n = 9 at year 2; PI-based n = 13 at year 1 ; n = 11 at year 2). Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values, both overall and stratified by drug regimens (NNRTI/NRTI- or PI-based).
Figure 9
Figure 9
Blood HIV-1 DNA load (expressed as log10copies/μg DNA) stratified by antiretroviral regimens. (A) Changes of HIV-1 DNA (expressed as log10 copies/μg DNA) versus baseline according to antiretroviral regimens in vaccinees (NNRTI- or NRTI-based n = 102; PI-based n = 45) or OBS subjects (NNRTI- or NRTI-based n = 36; PI-based n = 26). (B) Changes of HIV-1 DNA versus baseline in vaccinees of each treatment group stratified according to ARV regimens (7.5 μg, 3x n = 38; 7.5 μg, 5x n = 37; 30 μg, 3x n = 37; 30 μg, 5x n = 35). The dotted lines represent the 99% confidence interval of HIV-1 DNA mean change from baseline (−0.21, 0.04 log10 copies/μg) in OBS subjects (all time points). In all panels data are presented as mean values with standard error. Median time of follow-up was 96 weeks for vaccinees and 120 weeks for OBS subjects. A longitudinal analysis for repeated measurements was applied. P-values assess the values at each week after immunization versus baseline values stratified by drug regimens (NNRTI/NRTI- or PI-based).
Figure 10
Figure 10
Blood HIV-1 DNA load (expressed as log10copies/106CD4+T cells) stratified by antiretroviral regimens. (A) Changes of HIV-1 DNA (expressed as log10 copies/106 CD4+ T cells) in vaccinees (NNRTI- or NRTI-based n = 97; PI-based n = 45) or OBS subjects (NNRTI- or NRTI-based n = 30; PI-based n = 24). (B) Changes of HIV-1 DNA in vaccinees of each treatment group stratified according to ARV regimens (7.5 μg, 3x n = 36; 7.5 μg, 5x n = 36; 30 μg, 3x n = 36; 30 μg, 5x n = 34). The dotted lines represent the 99% confidence interval of HIV-1 DNA mean change from baseline (−0.24, 0.02 log10 copies/106 CD4+ T cells) in OBS subjects (all time points). In all panels data are presented as mean values with standard error. Median time of follow-up was 96 weeks for vaccinees and 120 weeks for OBS subjects. A longitudinal analysis for repeated measurements was applied. P-values assess the values at each week after immunization versus baseline values stratified by drug regimens (NNRTI/NRTI- or PI-based).
Figure 11
Figure 11
HIV-1 DNA decay in individuals immunized with Tat at 30 μg, 3x or OBS subjects. (A) Longitudinal regression analysis of HIV-1 DNA (log10 copies/106 CD4+ T cells) using a random-effect regression model up to 144 weeks since the first immunization in vaccinees (Tat 30 μg, 3x, n = 37) or OBS subjects (n = 62). All longitudinal data were included in the analysis with a median of 108 weeks of follow-up in vaccinees and 120 weeks in OBS subjects, respectively. (B) Estimates of HIV-1 DNA decay based on the regression model after 1, 2 and 3 years since the first immunization in vaccinees (Tat 30 μg, 3x in red) and in OBS subjects (in blue). (C) Estimates of HIV-1 DNA annual decay in vaccinees immunized with Tat 30 μg, 3x stratified according to NNRTI- or NRTI-based (in blue, n = 25) or PI-based (in red, n = 12) regimens. Results in panels B and C are expressed as the percentage of HIV-1 DNA decay with 95% confidence interval (CI).
Figure 12
Figure 12
Neutralization of Tat-mediated Env entry in DC and HIV-1 DNA in vaccinees. (A) Relationship between Tat-mediated entry of trimeric Env in DC and anti-Tat IgM (blue), IgG (red) and IgA (violet) Ab binding titers in subjects immunized with Tat 30 μg, 3x (generalized estimating equations with adjustment for repeated measures in the same patient; longitudinal samples from 31 individuals). (B) Percentage of Env entry in DC in the presence or absence of Tat in subjects immunized with Tat 30 μg, 3x at 48 weeks since the first immunization (n = 32) (Student’s t-test). Data are expressed as mean changes from baseline with standard error. (C) Inhibition of Tat-mediated Env entry in DC by anti-Tat Ab positive (n = 20) and anti-Tat Ab negative (n = 12) sera at week 48 from subjects immunized with Tat 30 μg, 3x. Of the 32 subjects evaluated, 30 were positive for anti-Tat Abs before week 48. Data are expressed as the percentage of subjects showing neutralization of Tat-mediated Env entry at 48 weeks as compared to baseline. (Fisher's Exact test) (D) Pearson correlation between anti-Tat IgM and IgG Ab titers at 48 weeks and Tat-mediated Env entry in DC at 48 weeks versus baseline in subjects immunized with Tat 30 μg, 3x (n = 32). (E) Reduction from baseline of HIV-1 DNA (log10 copies/106 CD4+ T cells) over time in sera from vaccinees (Tat 30 μg, 3x) positive for neutralization (≥50%) of Env entry in DC at week 48 since vaccination (n = 20). Data in panels are presented as mean values with standard error. Changes from baseline of HIV-1 DNA in subjects with anti-Tat Abs and neutralizing activity were evaluated with a longitudinal analysis for repeated measurements. P-values assess the values at each week after immunization versus values at baseline.
Figure 13
Figure 13
CD38+HLA-DR+/CD8+T cells in individuals immunized with Tat at the 30 μg doses or in OBS subjects. Changes from baseline of CD38+HLA-DR+/CD8+ at years 1, 2 and 3 in (A) vaccinees of the Tat 30 μg, 3x group (year 1: n = 28; year 2: n = 20; year 3: n = 10) and the Tat 30 μg, 5x group (year 1: n = 28; year: 2 n = 19; year 3: n = 9); (B) OBS subjects (year 1: n = 38; year 2: n = 25). (C) Changes from baseline of CD38+HLA-DR+/CD8+ at week 8, 12, 20 and 48 in vaccinees of the Tat 30 μg, 3x group (n = 28); and the Tat 30 μg, 5x group (n = 28). Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2, or 3 or week 8, 12, 20 or 48 after immunization versus baseline values.
Figure 14
Figure 14
NK cells in immunized with Tat at the 30 μg doses or in OBS subjects. Changes from baseline of NK cells/μl at years 1, 2 and 3 in (A) vaccinees of the Tat 30 μg, 3x (year 1 n = 37; year 2 n = 27; year 3: n = 18); and the Tat 30 μg, 5x group (year 1: n = 37; year 2: n = 26; year 3: n = 16); and (B) OBS subjects (year 1: n = 79; year 2: n = 42; year 3: n = 30). Data are presented as mean values with standard error. A longitudinal analysis for repeated measurements was applied. P-values assess the values at year 1, 2 or 3 after immunization versus baseline values.

References

    1. Klatt NR, Chomont N, Douek DC, Deeks SG. Immune activation and HIV persistence: implications for curative approaches to HIV infection. Immunol Rev. 2013;254:326–42. doi: 10.1111/imr.12065.
    1. Tsiara CG, Nikolopoulos GK, Bagos PG, Goujard C, Katzenstein TL, Minga AK, et al. Impact of HIV type 1 DNA levels on spontaneous disease progression: a meta-analysis. AIDS Res Hum Retroviruses. 2012;28:366–73. doi: 10.1089/aid.2011.0032.
    1. Deeks SG, Phillips AN. HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. BMJ. 2009;338:a3172. doi: 10.1136/bmj.a3172.
    1. Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol. 2008;214:231–41. doi: 10.1002/path.2276.
    1. Alexaki A, Liu Y, Wigdahl B. Cellular reservoirs of HIV-1 and their role in viral persistence. Curr HIV Res. 2008;6:388–400. doi: 10.2174/157016208785861195.
    1. Sigal A, Kim JT, Balazs AB, Dekel E, Mayo A, Milo R, et al. Cell-to-cell spread of HIV permits ongoing replication despite antiretroviral therapy. Nature. 2011;477:95–8. doi: 10.1038/nature10347.
    1. Kostrikis LG, Touloumi G, Karanicolas R, Pantazis N, Anastassopoulou C, Karafoulidou A, et al. Quantitation of human immunodeficiency virus type 1 DNA forms with the second template switch in peripheral blood cells predicts disease progression independently of plasma RNA load. J Virol. 2002;76:10099–108. doi: 10.1128/JVI.76.20.10099-10108.2002.
    1. Nicastri E, Palmisano L, Sarmati L, D’ettorre G, Parisi S, Andreotti M, et al. HIV-1 residual viremia and proviral DNA in patients with suppressed plasma viral load (<400 HIV-RNA cp/mL) during differential antiretroviral regimens. Curr HIV Res. 2008;6:261–6. doi: 10.2174/157016208784325010.
    1. Katlama C, Deeks SG, Autran B, Martinez-Picado J, van Lunzen J, Rouzioux C, et al. Barriers to a cure for HIV: new ways to target and eradicate HIV-1 reservoirs. Lancet. 2013;381:2109–17. doi: 10.1016/S0140-6736(13)60104-X.
    1. Drechsler H, Powderly WG. Switching effective antiretroviral therapy: a review. Clin Infect Dis. 2002;35:1219–30. doi: 10.1086/343050.
    1. McKinnon JE, Mellors JW, Swindells S. Simplification strategies to reduce antiretroviral drug exposure: progress and prospects. Antivir Ther. 2009;14:1–12.
    1. Volberding PA, Deeks SG. Antiretroviral therapy and management of HIV infection. Lancet. 2010;376:49–62. doi: 10.1016/S0140-6736(10)60676-9.
    1. Weinberger LS, Burnett JC, Toettcher JE, Arkin AP, Schaffer DV. Stochastic gene expression in a lentiviral positive-feedback loop: HIV-1 Tat fluctuations drive phenotypic diversity. Cell. 2005;122:169–82. doi: 10.1016/j.cell.2005.06.006.
    1. Ensoli B, Barillari G, Salahuddin SZ, Gallo RC, Wong-Staal F. Tat protein of HIV-1 stimulates growth of cells derived from Kaposi's sarcoma lesions of AIDS patients. Nature. 1990;345:84–6. doi: 10.1038/345084a0.
    1. Ensoli B, Buonaguro L, Barillari G, Fiorelli V, Gendelman R, Morgan RA, et al. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. J Virol. 1993;67:277–87.
    1. Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, Raffeld M, et al. Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi's sarcoma. Nature. 1994;371:674–680. doi: 10.1038/371674a0.
    1. Chang HC, Samaniego F, Nair BC, Buonaguro L, Ensoli B. HIV-1 Tat protein exits from cells via a leaderless secretory pathway and binds to extracellular matrix-associated heparan sulfate proteoglycans through its basic region. AIDS. 1997;11:1421–31. doi: 10.1097/00002030-199712000-00006.
    1. Rayne F, Debaisieux S, Yezid H, Lin YL, Mettling C, Konate K, et al. Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J. 2010;29:1348–1362. doi: 10.1038/emboj.2010.32.
    1. Ott M, Emiliani S, Van Lint C, Herbein G, Lovett J, Chirmule N, et al. Immune hyperactivation of HIV-1-infected T cells mediated by Tat and the CD28 pathway. Science. 1997;275:1481–1485. doi: 10.1126/science.275.5305.1481.
    1. Wu Y, Marsh JW. Selective transcription and modulation of resting T cell activity by preintegrated HIV DNA. Science. 2001;293:1503–6. doi: 10.1126/science.1061548.
    1. Mediouni S, Darque A, Baillat G, Ravaux I, Dhiver C, Tissot-Dupont H, et al. Antiretroviral therapy does not block the secretion of the human immunodeficiency virus Tat protein. Infect Disord Drug Targets. 2012;12:81–6. doi: 10.2174/187152612798994939.
    1. Ensoli B, Bellino S, Tripiciano A, Longo O, Francavilla V, Marcotullio S, et al. Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART. PLoS ONE. 2010;5:e13540. doi: 10.1371/journal.pone.0013540.
    1. Johnson TP, Patel K, Johnson KR, Maric D, Calabresi PA, Hasbun R, et al. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci USA. 2013;110:13588–93. doi: 10.1073/pnas.1308673110.
    1. Fanales-Belasio E, Moretti S, Nappi F, Barillari G, Micheletti F, Cafaro A, et al. Native HIV-1 Tat protein targets monocyte-derived dendritic cells and enhances their maturation, function, and antigen-specific T cell responses. J Immunol. 2002;168:197–206. doi: 10.4049/jimmunol.168.1.197.
    1. Gavioli R, Gallerani E, Fortini C, Fabris M, Bottoni A, Canella A, et al. HIV-1 Tat protein modulates the generation of cytotoxic T cell epitopes by modifying proteasome composition and enzymatic activity. J Immunol. 2004;173:3838–3843. doi: 10.4049/jimmunol.173.6.3838.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, Narciso P, et al. The therapeutic phase I trial of the recombinant native HIV-1 Tat protein. AIDS. 2008;22:2207–2209. doi: 10.1097/QAD.0b013e32831392d4.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, Narciso P, et al. The preventive phase I trial with the HIV-1 Tat-based vaccine. Vaccine. 2009;28:371–378. doi: 10.1016/j.vaccine.2009.10.038.
    1. Longo O, Tripiciano A, Fiorelli V, Bellino S, Scoglio A, Collacchi B, et al. Phase I therapeutic trial of the HIV-1 Tat protein and long term follow-up. Vaccine. 2009;27:3306–3312. doi: 10.1016/j.vaccine.2009.01.090.
    1. Huang L, Bosch I, Hofmann W, Sodroski J, Pardee AB. Tat protein induces human immunodeficiency virus type 1 (HIV-1) coreceptors and promotes infection with both macrophage-tropic and T-lymphotropic HIV-1 strains. J Virol. 1998;72:8952–8960.
    1. Li CJ, Ueda Y, Shi B, Borodyansky L, Huang L, Li YZ, et al. Tat protein induces self-perpetuating permissivity for productive HIV-1 infection. Proc Natl Acad Sci USA. 1997;94:8116–20. doi: 10.1073/pnas.94.15.8116.
    1. Nappi F, Chiozzini C, Bordignon V, Borsetti A, Bellino S, Cippitelli M, et al. Immobilized HIV-1 Tat protein promotes gene transfer via a transactivation-independent mechanism which requires binding of Tat to viral particles. J Gene Med. 2009;11:955–965. doi: 10.1002/jgm.1381.
    1. Monini P, Cafaro A, Srivastava IK, Moretti S, Sharma VA, Andreini C, et al. HIV-1 Tat Promotes Integrin-Mediated HIV Transmission to Dendritic Cells by Binding Env Spikes and Competes Neutralization by Anti-HIV Antibodies. PLoS ONE. 2012;7:e48781. doi: 10.1371/journal.pone.0048781.
    1. Chertova E, Chertov O, Coren LV, Roser JD, Trubey CM, Bess JW, Jr, et al. Proteomic and biochemical analysis of purified human immunodeficiency virus type 1 produced from infected monocyte-derived macrophages. J Virol. 2006;80:9039–52. doi: 10.1128/JVI.01013-06.
    1. Cafaro A, Caputo A, Fracasso C, Maggiorella MT, Goletti D, Baroncelli S, et al. Control of SHIV-89.6P-infection of cynomolgus monkeys by HIV-1 Tat protein vaccine. Nat Med. 1999;5:643–50. doi: 10.1038/9488.
    1. Cafaro A, Bellino S, Titti F, Maggiorella MT, Sernicola L, Wiseman RW, et al. Impact of viral dose and major histocompatibility complex class IB haplotype on viral outcome in Tat-vaccinated mauritian cynomolgus monkeys upon challenge with SHIV89.6P. J Virol. 2010;84:8953–8958. doi: 10.1128/JVI.00377-10.
    1. Bachler BC, Humbert M, Palikuqi B, Siddappa NB, Lakhashe SK, Rasmussen RA, et al. Novel biopanning strategy to identify epitopes associated with vaccine protection. J Virol. 2013;87:4403–16. doi: 10.1128/JVI.02888-12.
    1. Reiss P, de Wolf F, Kuiken CL, de Ronde A, Dekker J, Boucher CA, et al. Contribution of antibody response to recombinant HIV-1 gene-encoded products nef, rev, tat, and protease in predicting development of AIDS in HIV-1-infected individuals. J Acquir Immune Defic Syndr. 1991;4:165–72.
    1. Re MC, Furlini G, Vignoli M, Ramazzotti E, Roderigo G, De Rosa V, et al. Effect of antibody to HIV-1 Tat protein on viral replication in vitro and progression of HIV-1 disease in vivo. J Acquir Immune Defic Syndr Hum Retrovirol. 1995;10:408–16. doi: 10.1097/00042560-199512000-00003.
    1. Richardson MW, Mirchandani J, Duong J, Grimaldo S, Kocieda V, Hendel H, et al. Antibodies to Tat and Vpr in the GRIV cohort: differential association with maintenance of long-term non-progression status in HIV-1 infection. Biomed Pharmacother. 2003;57:4–14. doi: 10.1016/S0753-3322(02)00327-X.
    1. Rezza G, Fiorelli V, Dorrucci M, Ciccozzi M, Tripiciano A, Scoglio A, et al. The presence of anti-Tat antibodies is predictive of long-term nonprogression to AIDS or severe immunodeficiency: findings in a cohort of HIV-1 seroconverters. J Infect Dis. 2005;191:1321–1324. doi: 10.1086/428909.
    1. Bellino S, Tripiciano A, Picconi O, Francavilla V, Longo O, Sgadari C, et al. The presence of anti-Tat antibodies in HIV-infected individuals is associated with containment of CD4+ T-cell decay and viral load, and with delay of disease progression: results of a 3-year cohort study. Retrovirology. 2014;11:49. doi: 10.1186/1742-4690-11-49.
    1. Fanales-Belasio E, Moretti S, Fiorelli V, Tripiciano A, Pavone Cossut MR, Scoglio A, et al. HIV-1 Tat addresses dendritic cells to induce a predominant Th1-type adaptive immune response that appears prevalent in the asymptomatic stage of infection. J Immunol. 2009;182:2888–2897. doi: 10.4049/jimmunol.0711406.
    1. Gavioli R, Cellini S, Castaldello A, Voltan R, Gallerani E, Gagliardoni F, et al. The Tat protein broadens T cell responses directed to the HIV-1 antigens Gag and Env: implications for the design of new vaccination strategies against AIDS. Vaccine. 2008;26:727–737. doi: 10.1016/j.vaccine.2007.11.040.
    1. Sforza F, Nicoli F, Gallerani E, Finessi V, Reali E, Cafaro A, et al. HIV-1 Tat affects the programming and functionality of human CD8+ T cells by modulating the expression of T-box transcription factors. AIDS. 2014;28:1729–38. doi: 10.1097/QAD.0000000000000315.
    1. Nicoli F, Finessi V, Sicurella M, Rizzotto L, Gallerani E, Destro F, et al. The HIV-1 Tat protein induces the activation of CD8+ T cells and affects in vivo the magnitude and kinetics of antiviral responses. PLoS ONE. 2013;8:e77746. doi: 10.1371/journal.pone.0077746.
    1. Migueles SA, Weeks KA, Nou E, Berkley A, Rood JE, Osborne CM, et al. Defective human immunodeficiency virus-specific CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not restored by antiretroviral therapy. J Virol. 2009;83:11876–89. doi: 10.1128/JVI.01153-09.
    1. Sáez-Cirión A, Sinet M, Shin SY, Urrutia A, Versmisse P, Lacabaratz C, et al. Heterogeneity in HIV suppression by CD8 T cells from HIV controllers: association with Gag-specific CD8 T cell responses. J Immunol. 2009;182:7828–37. doi: 10.4049/jimmunol.0803928.
    1. Shan L, Deng K, Shroff NS, Durand CM, Rabi SA, Yang HC, et al. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity. 2012;36:491–501. doi: 10.1016/j.immuni.2012.01.014.
    1. Chiozzini C, Collacchi B, Nappi F, Bauer T, Arenaccio C, Tripiciano A, et al. Surface-bound Tat inhibits antigen-specific CD8+ T-cell activation in an integrin-dependent manner. AIDS. 2014;28:2189–200. doi: 10.1097/QAD.0000000000000389.
    1. European Medicines Agency. Note for Guidance on Choice of Control Group in Clinical Trials (CPMP/ICH/364/96). January 2001.
    1. Kelley CF, Kitchen CM, Hunt PW, Rodriguez B, Hecht FM, Kitahata M, et al. Incomplete peripheral CD4+ cell count restoration in HIV-infected patients receiving long-term antiretroviral treatment. Clin Infect Dis. 2009;48:787–94. doi: 10.1086/597093.
    1. Robbins GK, Spritzler JG, Chan ES, Asmuth DM, Gandhi RT, Rodriguez BA, et al. Incomplete reconstitution of T cell subsets on combination antiretroviral therapy in the AIDS Clinical Trials Group protocol 384. Clin Infect Dis. 2009;48:350–61. doi: 10.1086/595888.
    1. Rallón N, Sempere-Ortells JM, Soriano V, Benito JM. Central memory CD4 T cells are associated with incomplete restoration of the CD4 T cell pool after treatment-induced long-term undetectable HIV viraemia. J Antimicrob Chemoter. 2013;68:2616–25. doi: 10.1093/jac/dkt245.
    1. Casazza JP, Betts MR, Picker LJ, Koup RA. Decay kinetics of human immunodeficiency virus-specific CD8+ T cells in peripheral blood after initiation of highly active antiretroviral therapy. J Virol. 2001;75:6508–16. doi: 10.1128/JVI.75.14.6508-6516.2001.
    1. Serrano-Villar Serrano-Villar S, Gutiérrez C, Vallejo A, Hernández-Novoa B, Díaz L, Abad Fernández M, et al. The CD4/CD8 ratio in HIV-infected subjects is independently associated with T-cell activation despite long-term viral suppression. J Infect. 2013;66:57–66. doi: 10.1016/j.jinf.2012.09.013.
    1. Serrano-Villar S, Sainz T, Lee SA, Hunt PW, Sinclair E, Shacklett BL, et al. HIV-infected individuals with low CD4/CD8 ratio despite effective antiretroviral therapy exhibit altered T cell subsets, heightened CD8+ T cell activation, and increased risk of non-AIDS morbidity and mortality. PLoS Pathogens. 2014;10:e1004078. doi: 10.1371/journal.ppat.1004078.
    1. Brunetta E, Hudspeth KL, Mavilio D. Pathologic natural killer cell subset redistribution in HIV-1 infection: new insights in pathophysiology and clinical outcomes. J Leukoc Biol. 2010;88:1119–1130. doi: 10.1189/jlb.0410225.
    1. Pensieroso S, Galli L, Nozza S, Ruffin N, Castagna A, Tambussi G, et al. B-cell subset alterations and correlated factors in HIV-1 infection. AIDS. 2013;27:1209–17. doi: 10.1097/QAD.0b013e32835edc47.
    1. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9:727–8. doi: 10.1038/nm880.
    1. Costiniuk CT, Jenabian MA. HIV Reservoir Dynamics in the Face of Highly Active Antiretroviral Therapy. AIDS Patient Care STDs. 2015;29:55–68. doi: 10.1089/apc.2014.0173.
    1. Medina-Ramírez M, Sánchez-Merino V, Sánchez-Palomino S, Merino-Mansilla A, Ferreira CB, Pérez I, et al. Broadly cross-neutralizing antibodies in HIV-1 patients with undetectable viremia. J Virol. 2011;85:5804–13. doi: 10.1128/JVI.02482-10.
    1. Sáez-Cirión A, Lacabaratz C, Lambotte O, Versmisse P, Urrutia A, Boufassa F, et al. HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc Natl Acad Sci USA. 2007;104:6776–81. doi: 10.1073/pnas.0611244104.
    1. Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol. 2014;12:750–64. doi: 10.1038/nrmicro3352.
    1. McKinstry KK, Strutt TM, Swain SL. The potential of CD4 T-cell memory. Immunology. 2010;130:1–9. doi: 10.1111/j.1365-2567.2010.03259.x.
    1. Cui W, Kaech SM. Generation of effector CD8+ T cells and their conversion to memory T cells. Immunol Rev. 2010;236:151–66. doi: 10.1111/j.1600-065X.2010.00926.x.
    1. Hoxie JA, June CH. Novel cell and gene therapies for HIV. Cold Spring Harb Perspect Med. 2012; 2(10).
    1. García F, León A, Gatell JM, Plana M, Gallart T. Therapeutic vaccines against HIV infection. Hum Vaccin Immunother. 2012;8:569–81. doi: 10.4161/hv.19555.
    1. Ensoli B, Cafaro A, Monini P, Marcotullio S, Ensoli F. Challenges in HIV Vaccine Research for Treatment and Prevention. Frontiers Immunol. 2014;5:417. doi: 10.3389/fimmu.2014.00417.
    1. Walker B, McMichael A. The T-cell response to HIV. Cold Spring Harb Perspect Med. 2012;2:a007054. doi: 10.1101/cshperspect.a007054.
    1. Carrington M, Alter G. Innate immune control of HIV. Cold Spring Harb Perspect Med. 2012;2:a007070. doi: 10.1101/cshperspect.a007070.
    1. Castells M. Rapid desensitization for hypersensitivity reactions to medications. Immunol Allergy Clin North Am. 2009;29:585–606. doi: 10.1016/j.iac.2009.04.012.
    1. Keet CA, Wood RA. Emerging therapies for food allergy. J Clin Invest. 2014;124:1880–1886. doi: 10.1172/JCI72061.
    1. Kenney RT, Frech SA, Muenz LR, Villar CP, Glenn GM. Dose sparing with intradermal injection of influenza vaccine. N Engl J Med. 2004;351:2295–301. doi: 10.1056/NEJMoa043540.
    1. Leroux-Roels I, Borkowski A, Vanwolleghem T, Dramé M, Clement F, Hons E, et al. Antigen sparing and cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet. 2007;370:580–9. doi: 10.1016/S0140-6736(07)61297-5.
    1. Pope M, Haase AT. Transmission, acute HIV-1 infection and the quest for strategies to prevent infection. Nat Med. 2003;9:847–52. doi: 10.1038/nm0703-847.
    1. Haase A. Targeting early infection to prevent HIV-1 mucosal transmission. Nature. 2010;464:217–23. doi: 10.1038/nature08757.
    1. Coleman CM, Wu L. HIV interactions with monocytes and dendritic cells: viral latency and reservoirs. Retrovirology. 2009;6:51. doi: 10.1186/1742-4690-6-51.
    1. Saksena NK, Wang B, Zhou L, Soedjono M, Ho YS, Conceicao V. HIV reservoirs in vivo and new strategies for possible eradication of HIV from the reservoir sites. HIV/AIDS (Auckl) 2010;2:103–22.
    1. Iglesias-Ussel MD, Romerio F. HIV reservoirs: the new frontier. AIDS Rev. 2011;13:13–29.
    1. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med. 1999;5:512–7. doi: 10.1038/8394.
    1. Buttò S, Fiorelli V, Tripiciano A, Ruiz-Alvarez MJ, Scoglio A, Ensoli F, et al. Sequence conservation and antibody cross-recognition of clade B human immunodeficiency virus (HIV) type 1 Tat protein in HIV-1-infected Italians, Ugandans, and South Africans. J Infect Dis. 2003;188:1171–80. doi: 10.1086/378412.
    1. Gregory TR. Animal Genome Size Database 2013.
    1. Casabianca A, Orlandi C, Canovari B, Scotti M, Acetoso M, Valentini M, et al. A real time PCR platform for the simultaneous quantification of total and extrachromosomal HIV DNA forms in blood of HIV-1 infected patients. PLoS ONE. 2014;9:e111919. doi: 10.1371/journal.pone.0111919.
    1. Bannert N, Kurth R. The evolutionary dynamics of Human Endogenous Retroviral families. Annu Rev Genomics Hum Genet. 2006;7:149–73. doi: 10.1146/annurev.genom.7.080505.115700.

Source: PubMed

3
Abonnere