Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART

Barbara Ensoli, Stefania Bellino, Antonella Tripiciano, Olimpia Longo, Vittorio Francavilla, Simone Marcotullio, Aurelio Cafaro, Orietta Picconi, Giovanni Paniccia, Arianna Scoglio, Angela Arancio, Cristina Ariola, Maria J Ruiz Alvarez, Massimo Campagna, Donato Scaramuzzi, Cristina Iori, Roberto Esposito, Cristina Mussini, Florio Ghinelli, Laura Sighinolfi, Guido Palamara, Alessandra Latini, Gioacchino Angarano, Nicoletta Ladisa, Fabrizio Soscia, Vito S Mercurio, Adriano Lazzarin, Giuseppe Tambussi, Raffaele Visintini, Francesco Mazzotta, Massimo Di Pietro, Massimo Galli, Stefano Rusconi, Giampiero Carosi, Carlo Torti, Giovanni Di Perri, Stefano Bonora, Fabrizio Ensoli, Enrico Garaci, Barbara Ensoli, Stefania Bellino, Antonella Tripiciano, Olimpia Longo, Vittorio Francavilla, Simone Marcotullio, Aurelio Cafaro, Orietta Picconi, Giovanni Paniccia, Arianna Scoglio, Angela Arancio, Cristina Ariola, Maria J Ruiz Alvarez, Massimo Campagna, Donato Scaramuzzi, Cristina Iori, Roberto Esposito, Cristina Mussini, Florio Ghinelli, Laura Sighinolfi, Guido Palamara, Alessandra Latini, Gioacchino Angarano, Nicoletta Ladisa, Fabrizio Soscia, Vito S Mercurio, Adriano Lazzarin, Giuseppe Tambussi, Raffaele Visintini, Francesco Mazzotta, Massimo Di Pietro, Massimo Galli, Stefano Rusconi, Giampiero Carosi, Carlo Torti, Giovanni Di Perri, Stefano Bonora, Fabrizio Ensoli, Enrico Garaci

Abstract

Although HAART suppresses HIV replication, it is often unable to restore immune homeostasis. Consequently, non-AIDS-defining diseases are increasingly seen in treated individuals. This is attributed to persistent virus expression in reservoirs and to cell activation. Of note, in CD4(+) T cells and monocyte-macrophages of virologically-suppressed individuals, there is continued expression of multi-spliced transcripts encoding HIV regulatory proteins. Among them, Tat is essential for virus gene expression and replication, either in primary infection or for virus reactivation during HAART, when Tat is expressed, released extracellularly and exerts, on both the virus and the immune system, effects that contribute to disease maintenance. Here we report results of an ad hoc exploratory interim analysis (up to 48 weeks) on 87 virologically-suppressed HAART-treated individuals enrolled in a phase II randomized open-label multicentric clinical trial of therapeutic immunization with Tat (ISS T-002). Eighty-eight virologically-suppressed HAART-treated individuals, enrolled in a parallel prospective observational study at the same sites (ISS OBS T-002), served for intergroup comparison. Immunization with Tat was safe, induced durable immune responses, and modified the pattern of CD4(+) and CD8(+) cellular activation (CD38 and HLA-DR) together with reduction of biochemical activation markers and persistent increases of regulatory T cells. This was accompanied by a progressive increment of CD4(+) T cells and B cells with reduction of CD8(+) T cells and NK cells, which were independent from the type of antiretroviral regimen. Increase in central and effector memory and reduction in terminally-differentiated effector memory CD4(+) and CD8(+) T cells were accompanied by increases of CD4(+) and CD8(+) T cell responses against Env and recall antigens. Of note, more immune-compromised individuals experienced greater therapeutic effects. In contrast, these changes were opposite, absent or partial in the OBS population. These findings support the use of Tat immunization to intensify HAART efficacy and to restore immune homeostasis.

Trial registration: ClinicalTrials.gov NCT00751595.

Trial registration: ClinicalTrials.gov NCT00751595 NCT01024556.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Flow diagram of the study…
Figure 1. Flow diagram of the study participants.
One hundred and forty-four HAART-treated patients were screened for enrollment. Of them, 87 met the inclusion criteria and were randomized on schedule and dose of immunization. This represents all the study population prior to the protocol amendment. All recruited individuals were included in the safety analysis (n = 87). Six subjects discontinued the immunization schedule. Of them, 5 were evaluated only for safety, since received at least one immunization, and 1 was evaluated also for immunogenicity since received 3 immunizations out of 5 (as indicated in the Protocol S1). A total of 82 individuals completed the 20-weeks period of the study and 68 have completed the 48-weeks period after the first immunizations.
Figure 2. Anti-Tat humoral immune responses.
Figure 2. Anti-Tat humoral immune responses.
(A) Percentage of subjects developing anti-Tat IgM (blue bar), IgG (purple bar), IgA (violet bar), or total anti-Tat Ab (green bar), stratified by Tat dose and treatment groups. Responder frequency up to week 20 among groups was analyzed by the Cochran-Armitage Trend test (p = 0.0117 and p = 0.0051 for IgM and IgA, respectively). Tat 7.5 µg, 3x, n = 21; Tat 7.5 µg, 5x, n = 18; Tat 30 µg, 3x, n = 21; Tat 30 µg, 5x, n = 22 subjects, respectively. (B) Percentage of subjects positive for anti-Tat Ab at week 48 (Tat 7.5 µg, 3x, n = 20; Tat 7.5 µg, 5x, n = 15; Tat 30 µg, 3x, n = 17; Tat 30 µg, 5x, n = 16 subjects, respectively). Responder frequency among groups was analyzed as described above (p = 0.0024 and p = 0.0048 for IgM and IgG, respectively, p = 0.0007 for total Ig). (C) Peak of anti-Tat IgM, IgG or IgA titers (geometric mean and range) up to 20 weeks after the first immunization from subjects positive for anti-Tat Ab (blue diamond Tat 7.5 µg and red square Tat 30 µg). Anti-Tat IgM: Tat 7.5 µg, n = 14; Tat 30 µg, n = 28. Anti-Tat IgG: Tat 7.5 µg, n = 26; Tat 30 µg, n = 33. Anti-Tat IgA: Tat 7.5 µg, n = 7; Tat 30 µg, n = 24 subjects, respectively. (D) Anti-Tat IgM, IgG or IgA titers at 48 weeks after the first immunization from subjects positive for anti-Tat Ab. Anti-Tat IgM: Tat 7.5 µg, n = 6; Tat 30 µg, n = 20. Anti-Tat IgG: Tat 7.5 µg, n = 8; Tat 30 µg, n = 18. Anti-Tat IgA: Tat 7.5 µg, n = 5; Tat 30 µg, n = 10 subjects, respectively.
Figure 3. Anti-Tat cellular immune responses.
Figure 3. Anti-Tat cellular immune responses.
(A) Percentage of subjects showing anti-Tat specific production of IFN-γ, IL-2 or IL-4, respectively, measured at baseline (blue bar) and up to week 48 after the first immunization (red bar) and stratified by Tat dose (Tat 7.5 µg, n = 35; Tat 30 µg, n = 33 subjects, respectively). (B) Percentage of subjects showing anti-Tat CD4+ or CD8+ lymphoproliferative responses measured at baseline and up to week 48 and stratified by Tat dose (Tat 7.5 µg, n = 31; Tat 30 µg, n = 31). The analysis was performed using the McNemar's test: *p<0.05, **p<0.01.
Figure 4. Expression of activation markers on…
Figure 4. Expression of activation markers on CD8+ T cells after Tat immunization.
Changes from baseline of CD8+ T cells (gating on CD8+ T cells) expressing (A) CD38, (B) HLA-DR, or (C) both CD38 and HLA-DR. Results are shown according to Tat dose and time after the first immunization. Data are presented as the mean % changes (± standard error) at week 8, 12, 20 and 48. Blue bars: Tat 7.5 µg, n = 17 up to week 20 and n = 12 at week 48; red bars: Tat 30 µg, n = 21 up to week 20, n = 16 at week 48, respectively. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01. (D) Correlation between CD38+/CD8+ T cells (%) and anti-Tat IgA antibody titers (Multivariate regression model for repeated measures). Blue diamond: Tat 7.5 µg, n = 39; red square: Tat 30 µg, n = 43, respectively.
Figure 5. Expression of activation markers on…
Figure 5. Expression of activation markers on CD4+ T cells after Tat immunization.
Changes from baseline of CD4+ T cells (gating on CD4+ T cells) expressing (A) CD38, (B) HLA-DR, or (C) both CD38 and HLA-DR. Results are shown according to Tat dose and time after the first immunization. Data are presented as the mean % changes (± standard error) at week 8, 12, 20 and 48. Blue bars: Tat 7.5 µg, n = 17 up to week 20 and n = 12 at week 48; red bars: Tat 30 µg, n = 20 up to week 20, n = 15 at week 48, respectively; The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 6. Production of β2-microglobulin and neopterin…
Figure 6. Production of β2-microglobulin and neopterin after Tat immunization.
Changes from baseline, according to Tat dose, of (A) β2-microglobulin serum levels (mg/L) and (B) Neopterin (nmol/L). Blue bars: Tat 7.5 µg; red bars: Tat 30 µg. Data are presented as the mean changes (± standard error) at 4, 16 and 24 weeks (Tat 7.5 µg, n = 37, Tat 30 µg, n = 40 subjects, respectively). The t-Test for paired data was used for the analyses: **p<0.01.
Figure 7. Production of total Ig after…
Figure 7. Production of total Ig after Tat immunization.
Total IgM (A), IgG (B) and IgA (C) serum levels (mg/dL) are shown. Blue bars: Tat 7.5 µg, n = 37; red bars: Tat 30 µg, n = 40. Data are presented as the mean changes (± standard error). The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 8. CD25 and FOXP3 expression on…
Figure 8. CD25 and FOXP3 expression on CD4+ T cells after Tat immunization.
(A) Changes from baseline of CD4+ lymphocytes expressing CD25 are shown according to Tat dose and time after the first immunization (Tat 7.5 µg, n = 32 up to week 20 and n = 24 at week 48; Tat 30 µg, n = 34 up to week 20 and n = 28 at week 48, respectively). (B) Changes from baseline of the percentage of CD4+CD25+ lymphocytes expressing FOXP3+ (Tat 7.5 µg, n = 31 up to week 20 and n = 23 at week 48; Tat 30 µg, n = 29 up to week 20 and n = 28 at week 48, respectively). (C) Changes from baseline of the percentage of CD4+ T cells expressing CD25+FOXP3+ (Tat 7.5 µg, n = 31 up to week 20 and n = 23 at week 48; Tat 30 µg, n = 29 up to week 20 and n = 24 at week 48). (D) Changes from baseline of the absolute number of CD4+ lymphocytes expressing CD25+ FOXP3+ (Tat 7.5 µg, n = 30 up to week 20 and n = 20 at week 48; Tat 30 µg, n = 29 up to week 20 and n = 22 at week 48, respectively). Blue bars: Tat 7.5 µg; red bars: Tat 30 µg. Data are presented as the mean changes (± standard error) evaluated at 8, 12, 20 and 48 weeks after the first immunization. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 9. Evaluation of PBMC viability, CD4…
Figure 9. Evaluation of PBMC viability, CD4+ T cell and B cell counts after Tat immunization.
(A) Changes from baseline of in vitro PBMC viability, stratified by Tat dose. Blue bars: Tat 7.5 µg, n = 35 up to week 20 and n = 32 at week 48; red bars: Tat 30 µg, n = 40 up to week 20 and n = 33 at week 48, respectively. (B) Changes from baseline of CD4+ T cells/µL (data from clinical sites), stratified by Tat dose. Blue bars: Tat 7.5 µg, n = 39 up to week 24 and n = 30 at week 48; red bars Tat 30 µg, n = 43 up to week 24 and n = 34 at week 48. (C) Changes from baseline of B cells/µL, stratified by Tat dose. Blue bars: Tat 7.5 µg, n = 38 up to week 20 and n = 30 at week 48; red bars Tat 30 µg, n = 40 up to week 20 and n = 30 at week 48, respectively. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 10. Evaluation of the percentage of…
Figure 10. Evaluation of the percentage of CD4+, CD8+, NK and B cells in all Tat-immunized subjects and after stratification by Tat dose.
(A) Changes from baseline of CD4+, CD8+, NK and B cells (percentage) for all immunized subjects (n = 78 up to week 20, n = 60 at week 48). (B, C) Changes from baseline of CD4+, CD8+, NK and B cells (percentage), stratified by Tat doses. (B) Tat 7.5 µg, n = 38 up to week 20 and n = 30 at week 48; (C) Tat 30 µg, n = 40 up to week 20 and n = 30 at week 48, respectively. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 11. Evaluation of the percentage of…
Figure 11. Evaluation of the percentage of CD4+, CD8+, NK and B cells in all Tat-immunized subjects after stratification by HAART regimens.
Changes from baseline of CD4+, CD8+, NK and B cells (percentage) from all immunized patients are shown for NNRTI-based (A) and PI-based (B) treatments. NNRTI-based: n = 51 up to week 20 and n = 41 at week 48; PI-based: n = 19 up to week 20 and n = 12 at week 48, respectively. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 12. Effect of Tat immunization on…
Figure 12. Effect of Tat immunization on naïve, central and effector memory CD4+ and CD8+ T cells.
Percentage of naïve (CD45RA+/CD62L+), effector RA+ (CD45RA+/CD62L−, Temra) or RA- (CD45RA−/CD62L−, Temro) and central memory (CD45RA−/CD62L+, Tcm) CD4+ (A) or CD8+ (B) T cells at baseline and at week 8, 12, 20 and 48 after the first immunization for subjects immunized with both Tat doses (n = 18 up to week 20 and n = 10 at week 48). Asterisk indicates significant changes from baseline. The t-Test for paired data was used for the analyses: *p<0.05, **p<0.01.
Figure 13. Cellular immune responses against Env…
Figure 13. Cellular immune responses against Env or recall antigens after Tat immunization.
Percentage of responders at baseline (blue bar) and up to week 48 (red bar) are stratified by Tat dose. Percentage of subjects showing (A) anti-Env production of IFN-γ, IL-2 and IL-4 (Tat 7.5 µg, n = 31; Tat 30 µg, n = 29) and (B) CD4+ or CD8+ lymphoproliferative responses (Tat 7.5 µg, n = 31; Tat 30 µg, n = 30); (C) anti-Candida cytokines production (Tat 7.5 µg, n = 32; Tat 30 µg, n = 29), and (D) CD4+ or CD8+ lymphoproliferative responses (Tat 7.5 µg, n = 31; Tat 30 µg, n = 29); (E) anti-CEF production of IFN-γ, IL-2 and IL-4 (Tat 7.5 µg, n = 34; Tat 30 µg, n = 32), and (F) CD4+ or CD8+ lymphoproliferative responses (Tat 7.5 µg, n = 31; Tat 30 µg, n = 29). The analysis was performed using the McNemar's test: *p<0.05, **p<0.01.
Figure 14. Correlation of the reduction of…
Figure 14. Correlation of the reduction of the percentage of CD38+/CD8+T cells with the increase of anti-Tat IgA antibody titers, CD8+ T central memory and anti-Tat specific IL-2 production after Tat immunization.
Multivariate regression model for repeated measures was applied to CD38+/CD8+ T cells (%), including as explicative factors anti-Tat antibody titers (IgM, IgG, IgA), CD8+ central memory (%) and anti-Tat IFN-γ, IL-2 and IL-4 cytokines, for immunized subjects (n = 70).

References

    1. Walensky RP, Paltiel AD, Losina E, Mercincavage LM, Schackman BR, et al. The survival benefits of AIDS treatment in the United States. J Infect Dis. 2006;194:11–19.
    1. Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol. 2008;214:231–241.
    1. Deeks SG, Phillips AN. HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. BMJ. 2009;338:a3172.
    1. Kelley CF, Kitchen CM, Hunt PW, Rodriguez B, Hecht FM, et al. Incomplete peripheral CD4+ cell count restoration in HIV-infected patients receiving long-term antiretroviral treatment. Clin Infect Dis. 2009;48:787–794.
    1. Robbins GK, Spritzler JG, Chan ES, Asmuth DM, Gandhi RT, et al. Incomplete reconstitution of T cell subsets on combination antiretroviral therapy in the AIDS Clinical Trials Group protocol 384. Clin Infect Dis. 2009;48:350–361.
    1. Pensieroso S, Cagigi A, Palma P, Nilsson A, Capponi C, et al. Timing of HAART defines the integrity of memory B cells and the longevity of humoral responses in HIV-1 vertically-infected children. Proc Natl Acad Sci U S A. 2009;106:7939–7944.
    1. Cagigi A, Palma P, Nilsson A, Di CS, Pensieroso S, et al. The impact of active HIV-1 replication on the physiological age-related decline of immature-transitional B-cells in HIV-1 infected children. AIDS 2010
    1. Cagigi A, Nilsson A, Pensieroso S Chiodi F. Dysfunctional B-cell responses during HIV-1 infection: implication for influenza vaccination and highly active antiretroviral therapy. Lancet Infect Dis. 2010;10:499–503.
    1. Gathe J, Diaz R, Fatkenheuer G, Zeinecker J, Mak C, et al. Phase 3 Trials of Vicriviroc in Treatment-experienced Subjects Demonstrate Safety but Not Significantly Superior Efficacy over Potent Background Regimens Alone. 2010. Abstract from the 17th Conference on Retroviruses and Opportunistic Infections.
    1. Timothy W, Lalama C, Tenorio A, Landay A, Ribaudo H, et al. Maraviroc Intensification for Suboptimal CD4+ Cell Response Despite Sustained Virologic Suppression: ACTG 5256. 2010. Abstract from the 17th Conference on Retroviruses and Opportunistic Infections.
    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.
    1. Carter CC, Onafuwa-Nuga A, McNamara LA, Riddell J, Bixby D, et al. HIV-1 infects multipotent progenitor cells causing cell death and establishing latent cellular reservoirs. Nat Med. 2010;16:446–451.
    1. Chun TW, Carruth L, Finzi D, Shen X, Di Giuseppe JA, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387:183–188.
    1. Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 2009;106:9403–9408.
    1. Hunt PW, Deeks SG, Bangsberg DR, Moss A, Sinclair E, et al. The independent effect of drug resistance on T cell activation in HIV infection. AIDS. 2006;20:691–699.
    1. Lambotte O, Taoufik Y, de Goer MG, Wallon C, Goujard C, et al. Detection of infectious HIV in circulating monocytes from patients on prolonged highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2000;23:114–119.
    1. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, et al. The challenge of finding a cure for HIV infection. Science. 2009;323:1304–1307.
    1. Sharkey ME, Teo I, Greenough T, Sharova N, Luzuriaga K, et al. Persistence of episomal HIV-1 infection intermediates in patients on highly active anti-retroviral therapy. Nat Med. 2000;6:76–81.
    1. Trono D, Van LC, Rouzioux C, Verdin E, Barre-Sinoussi F, et al. HIV persistence and the prospect of long-term drug-free remissions for HIV-infected individuals. Science. 2010;329:174–180.
    1. Valentin A, Rosati M, Patenaude DJ, Hatzakis A, Kostrikis LG, et al. Persistent HIV-1 infection of natural killer cells in patients receiving highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 2002;99:7015–7020.
    1. Zhu T, Muthui D, Holte S, Nickle D, Feng F, et al. Evidence for human immunodeficiency virus type 1 replication in vivo in CD14(+) monocytes and its potential role as a source of virus in patients on highly active antiretroviral therapy. J Virol. 2002;76:707–716.
    1. Kelly J, Beddall MH, Yu D, Iyer SR, Marsh JW, et al. Human macrophages support persistent transcription from unintegrated HIV-1 DNA. Virology. 2008;372:300–312.
    1. Wu Y, Marsh JW. Selective transcription and modulation of resting T cell activity by preintegrated HIV DNA. Science. 2001;293:1503–1506.
    1. Lassen KG, Bailey JR, Siliciano RF. Analysis of human immunodeficiency virus type 1 transcriptional elongation in resting CD4+ T cells in vivo. J Virol. 2004;78:9105–9114.
    1. Lassen KG, Ramyar KX, Bailey JR, Zhou Y, Siliciano RF. Nuclear retention of multiply spliced HIV-1 RNA in resting CD4+ T cells. PLoS Pathog. 2006;2:e68.
    1. Furtado MR, Callaway DS, Phair JP, Kunstman KJ, Stanton JL, et al. Persistence of HIV-1 transcription in peripheral-blood mononuclear cells in patients receiving potent antiretroviral therapy. N Engl J Med. 1999;340:1614–1622.
    1. Zhang L, Ramratnam B, Tenner-Racz K, He Y, Vesanen M, et al. Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy. N Engl J Med. 1999;340:1605–1613.
    1. Natarajan V, Bosche M, Metcalf JA, Ward DJ, Lane HC, et al. HIV-1 replication in patients with undetectable plasma virus receiving HAART. Highly active antiretroviral therapy. Lancet. 1999;353:119–120.
    1. Wu Y, Marsh JW. Early transcription from nonintegrated DNA in human immunodeficiency virus infection. J Virol. 2003;77:10376–10382.
    1. Deeks SG. Immune dysfunction, inflammation, and accelerated aging in patients on antiretroviral therapy. Top HIV Med. 2009;17:118–123.
    1. Li JC, Yim HC, Lau AS. Role of HIV-1 Tat in AIDS pathogenesis: its effects on cytokine dysregulation and contributions to the pathogenesis of opportunistic infection. AIDS. 2010;24:1609–1623.
    1. Ott M, Emiliani S, Van LC, Herbein G, Lovett J, et al. Immune hyperactivation of HIV-1-infected T cells mediated by Tat and the CD28 pathway. Science. 1997;275:1481–1485.
    1. Cullen BR. Regulation of HIV gene expression. AIDS. 1995;9(Suppl A):S19–S32.
    1. Dayton AI, Sodroski JG, Rosen CA, Goh WC Haseltine WA. The trans-activator gene of the human T cell lymphotropic virus type III is required for replication. Cell. 1986;44:941–947.
    1. Hauber J, Perkins A, Heimer EP Cullen BR. Trans-activation of human immunodeficiency virus gene expression is mediated by nuclear events. Proc Natl Acad Sci U S A. 1987;84:6364–6368.
    1. Jordan A, Defechereux P, Verdin E. The site of HIV-1 integration in the human genome determines basal transcriptional activity and response to Tat transactivation. EMBO J. 2001;20:1726–1738.
    1. Lin X, Irwin D, Kanazawa S, Huang L, Romeo J, et al. Transcriptional profiles of latent human immunodeficiency virus in infected individuals: effects of Tat on the host and reservoir. J Virol. 2003;77:8227–8236.
    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;122:169–182.
    1. Jordan A, Bisgrove D, Verdin E. HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. EMBO J. 2003;22:1868–1877.
    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;11:1421–1431.
    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;345:84–86.
    1. Rayne F, Debaisieux S, Yezid H, Lin YL, Mettling C, et al. Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J. 2010.
    1. Ensoli B, Buonaguro L, Barillari G, Fiorelli V, Gendelman R, et al. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. J Virol. 1993;67:277–287.
    1. Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, et al. Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi's sarcoma. Nature. 1994;371:674–680.
    1. Buonaguro L, Buonaguro FM, Giraldo G, Ensoli B. The human immunodeficiency virus type 1 Tat protein transactivates tumor necrosis factor beta gene expression through a TAR-like structure. J Virol. 1994;68:2677–2682.
    1. Campbell GR, Loret EP. What does the structure-function relationship of the HIV-1 Tat protein teach us about developing an AIDS vaccine? Retrovirology. 2009;6:50.
    1. Fanales-Belasio E, Moretti S, Fiorelli V, Tripiciano A, Pavone Cossut MR, 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.
    1. Gavioli R, Gallerani E, Fortini C, Fabris M, Bottoni 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.
    1. Li CJ, Ueda Y, Shi B, Borodyansky L, Huang L, et al. Tat protein induces self-perpetuating permissivity for productive HIV-1 infection. Proc Natl Acad Sci U S A. 1997;94:8116–8120.
    1. Nappi F, Chiozzini C, Bordignon V, Borsetti A, Bellino S, 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.
    1. Zauli G, Gibellini D, Celeghini C, Mischiati C, Bassini A, et al. Pleiotropic effects of immobilized versus soluble recombinant HIV-1 Tat protein on CD3- mediated activation, induction of apoptosis, and HIV-1 long terminal repeat transactivation in purified CD4+ T lymphocytes. J Immunol. 1996;157:2216–2224.
    1. Fanales-Belasio E, Moretti S, Nappi F, Barillari G, Micheletti F, 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.
    1. Gavioli R, Cellini S, Castaldello A, Voltan R, Gallerani E, 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.
    1. Remoli AL, Marsili G, Perrotti E, Gallerani E, Ilari R, et al. Intracellular HIV-1 Tat protein represses constitutive LMP2 transcription increasing proteasome activity by interfering with the binding of IRF-1 to STAT1. Biochem J. 2006;396:371–380.
    1. Barillari G, Buonaguro L, Fiorelli V, Hoffman J, Michaels F, et al. Effects of cytokines from activated immune cells on vascular cell growth and HIV-1 gene expression. Implications for AIDS-Kaposi's sarcoma pathogenesis. J Immunol. 1992;149:3727–3734.
    1. Buonaguro L, Barillari G, Chang HK, Bohan CA, Kao V, et al. Effects of the human immunodeficiency virus type 1 Tat protein on the expression of inflammatory cytokines. J Virol. 1992;66:7159–7167.
    1. Lafrenie RM, Wahl LM, Epstein JS, Yamada KM, Dhawan S. Activation of monocytes by HIV-Tat treatment is mediated by cytokine expression. J Immunol. 1997;159:4077–4083.
    1. Puri RK, Aggarwal BB. Human immunodeficiency virus type 1 tat gene up-regulates interleukin 4 receptors on a human B-lymphoblastoid cell line. Cancer Res. 1992;52:3787–3790.
    1. Sastry KJ, Reddy HR, Pandita R, Totpal K, Aggarwal BB. HIV-1 tat gene induces tumor necrosis factor-beta (lymphotoxin) in a human B-lymphoblastoid cell line. J Biol Chem. 1990;265:20091–20093.
    1. Scala G, Ruocco MR, Ambrosino C, Mallardo M, Giordano V, et al. The expression of the interleukin 6 gene is induced by the human immunodeficiency virus 1 TAT protein. J Exp Med. 1994;179:961–971.
    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. Secchiero P, Zella D, Capitani S, Gallo RC, Zauli G. Extracellular HIV-1 tat protein up-regulates the expression of surface CXC-chemokine receptor 4 in resting CD4+ T cells. J Immunol. 1999;162:2427–2431.
    1. Chertova E, Chertov O, Coren LV, Roser JD, Trubey CM, et al. Proteomic and biochemical analysis of purified human immunodeficiency virus type 1 produced from infected monocyte-derived macrophages. J Virol. 2006;80:9039–9052.
    1. Fischer M, Joos B, Wong JK, Ott P, Opravil M, et al. Attenuated and nonproductive viral transcription in the lymphatic tissue of HIV-1-infected patients receiving potent antiretroviral therapy. J Infect Dis. 2004;189:273–285.
    1. Kelly J, Beddall MH, Yu D, Iyer SR, Marsh JW, et al. Human macrophages support persistent transcription from unintegrated HIV-1 DNA. Virology. 2008;372:300–312.
    1. Pasternak AO, Jurriaans S, Bakker M, Prins JM, Berkhout B, et al. Cellular levels of HIV unspliced RNA from patients on combination antiretroviral therapy with undetectable plasma viremia predict the therapy outcome. PLoS One. 2009;4:e8490.
    1. Allen TM, O'Connor DH, Jing P, Dzuris JL, Mothe BR, et al. Tat-specific cytotoxic T lymphocytes select for SIV escape variants during resolution of primary viraemia. Nature. 2000;407:386–390.
    1. Cao J, McNevin J, Malhotra U, McElrath MJ. Evolution of CD8+ T cell immunity and viral escape following acute HIV-1 infection. J Immunol. 2003;171:3837–3846.
    1. Ensoli B, Fiorelli V, Ensoli F, Cafaro A, Titti F, et al. Candidate HIV-1 Tat vaccine development: from basic science to clinical trials. AIDS. 2006;20:2245–2261.
    1. O'Connor DH, Allen TM, Vogel TU, Jing P, DeSouza IP, et al. Acute phase cytotoxic T lymphocyte escape is a hallmark of simian immunodeficiency virus infection. Nat Med. 2002;8:493–499.
    1. Rezza G, Fiorelli V, Dorrucci M, Ciccozzi M, Tripiciano 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.
    1. van Baalen CA, Pontesilli O, Huisman RC, Geretti AM, Klein MR, et al. Human immunodeficiency virus type 1 Rev- and Tat-specific cytotoxic T lymphocyte frequencies inversely correlate with rapid progression to AIDS. J Gen Virol. 1997;78((Pt 8)):1913–1918.
    1. Cafaro A, Bellino S, Titti F, Maggiorella MT, Sernicola L, 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.
    1. Reiss P, de WF, Kuiken CL, de RA, Dekker J, 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;4:165–172.
    1. Borsetti A, Baroncelli S, Maggiorella MT, Moretti S, Fanales-Belasio E, et al. Containment of infection in tat vaccinated monkeys after rechallenge with a higher dose of SHIV89.6P(cy243). Viral Immunol. 2009;22:117–124.
    1. Cafaro A, Caputo A, Fracasso C, Maggiorella MT, Goletti D, et al. Control of SHIV-89.6P-infection of cynomolgus monkeys by HIV-1 Tat protein vaccine. Nat Med. 1999;5:643–650.
    1. Maggiorella MT, Baroncelli S, Michelini Z, Fanales-Belasio E, Moretti S, et al. Long-term protection against SHIV89.6P replication in HIV-1 Tat vaccinated cynomolgus monkeys. Vaccine. 2004;22:3258–3269.
    1. Bellino S, Francavilla V, Longo O, Tripiciano A, Paniccia G, et al. Parallel conduction of the phase I preventive and therapeutic trials based on the Tat vaccine candidate. Reviews on Recent Clinical Trials. 2009;4:195–204.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, et al. The therapeutic phase I trial of the recombinant native HIV-1 Tat protein. AIDS. 2008;22:2207–2209.
    1. Ensoli B, Fiorelli V, Ensoli F, Lazzarin A, Visintini R, et al. The preventive phase I trial with the HIV-1 Tat-based vaccine. Vaccine. 2009;28:371–378.
    1. Longo O, Tripiciano A, Fiorelli V, Bellino S, Scoglio A, et al. Phase I therapeutic trial of the HIV-1 Tat protein and long term follow-up. Vaccine. 2009;27:3306–3312.
    1. Ensoli B. AIDS Vaccine Integrated Project (AVIP): A Novel Program and Developing Countries Partnership. 2005;9:38–40.
    1. Butto S, Fiorelli V, Tripiciano A, Ruiz-Alvarez MJ, Scoglio A, 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;188:1171–1180.
    1. Mullis KB, Faloona FA. Specific synthesis of DNA in vitro via a polymerase-catalyzed chain reaction. Methods Enzymol. 1987;155:335–350.
    1. Szabo SE, Monroe SL, Fiorino S, Bitzan J, Loper K. Evaluation of an automated instrument for viability and concentration measurements of cryopreserved hematopoietic cells. Lab Hematol. 2004;10:109–111.
    1. Burtis CA, Aswhood ER. Saunders WB, editor. Tietz Textbook of Clinical Chemistry, 2nd Edn. 1994
    1. Welles SL, Jackson JB, Yen-Lieberman B, Demeter L, Japour AJ, et al. Prognostic value of plasma human immunodeficiency virus type 1 (HIV-1) RNA levels in patients with advanced HIV-1 disease and with little or no prior zidovudine therapy. AIDS Clinical Trials Group Protocol 116A/116B/117 Team. J Infect Dis. 1996;174:696–703.
    1. Finkel TH, Tudor-Williams G, Banda NK, Cotton MF, Curiel T, et al. Apoptosis occurs predominantly in bystander cells and not in productively infected cells of HIV- and SIV-infected lymph nodes. Nat Med. 1995;1:129–134.
    1. Volberding PA, Deeks SG. Antiretroviral therapy and management of HIV infection. Lancet. 2010;376:49–62.
    1. Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 2008;28:710–722.
    1. Ho HN, Hultin LE, Mitsuyasu RT, Matud JL, Hausner MA, et al. Circulating HIV-specific CD8+ cytotoxic T cells express CD38 and HLA-DR antigens. J Immunol. 1993;150:3070–3079.
    1. Saez-Cirion A, Lacabaratz C, Lambotte O, Versmisse P, Urrutia A, 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 U S A. 2007;104:6776–6781.
    1. Savarino A, Bottarel F, Calosso L, Feito MJ, Bensi T, et al. Effects of the human CD38 glycoprotein on the early stages of the HIV-1 replication cycle. FASEB J. 1999;13:2265–2276.
    1. Biancotto A, Iglehart SJ, Vanpouille C, Condack CE, Lisco A, et al. HIV-1 induced activation of CD4+ T cells creates new targets for HIV-1 infection in human lymphoid tissue ex vivo. Blood. 2008;111:699–704.
    1. Liu Z, Cumberland WG, Hultin LE, Prince HE, Detels R, et al. Elevated CD38 antigen expression on CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count, soluble immune activation markers or combinations of HLA-DR and CD38 expression. J Acquir Immune Defic Syndr Hum Retrovirol. 1997;16:83–92.
    1. Alves NL, van Leeuwen EM, Remmerswaal EB, Vrisekoop N, Tesselaar K, et al. A new subset of human naive CD8+ T cells defined by low expression of IL-7R alpha. J Immunol. 2007;179:221–228.
    1. Jackson DG, Bell JI. Isolation of a cDNA encoding the human CD38 (T10) molecule, a cell surface glycoprotein with an unusual discontinuous pattern of expression during lymphocyte differentiation. J Immunol. 1990;144:2811–2815.
    1. Malavasi F, Deaglio S, Funaro A, Ferrero E, Horenstein AL, et al. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol Rev. 2008;88:841–886.
    1. Speiser DE, Migliaccio M, Pittet MJ, Valmori D, Lienard D, et al. Human CD8(+) T cells expressing HLA-DR and CD28 show telomerase activity and are distinct from cytolytic effector T cells. Eur J Immunol. 2001;31:459–466.
    1. Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity. 2009;30:626–635.
    1. Chase AJ, Yang HC, Zhang H, Blankson JN, Siliciano RF. Preservation of FoxP3+ regulatory T cells in the peripheral blood of human immunodeficiency virus type 1-infected elite suppressors correlates with low CD4+ T-cell activation. J Virol. 2008;82:8307–8315.
    1. Jiao Y, Fu J, Xing S, Fu B, Zhang Z, et al. The decrease of regulatory T cells correlates with excessive activation and apoptosis of CD8+ T cells in HIV-1-infected typical progressors, but not in long-term non-progressors. Immunology. 2009;128:e366–e375.
    1. Almeida JR, Price DA, Papagno L, Arkoub ZA, Sauce D, et al. Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. J Exp Med. 2007;204:2473–2485.
    1. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA, et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107:4781–4789.

Source: PubMed

3
Předplatit