A Randomized Placebo-Controlled Efficacy Study of a Prime Boost Therapeutic Vaccination Strategy in HIV-1-Infected Individuals: VRI02 ANRS 149 LIGHT Phase II Trial

Y Lévy, C Lacabaratz, E Lhomme, A Wiedemann, C Bauduin, C Fenwick, E Foucat, M Surenaud, L Guillaumat, V Boilet, V Rieux, O Bouchaud, P-M Girard, J-M Molina, P Morlat, L Hocqueloux, L Richert, G Pantaleo, J D Lelièvre, R Thiébaut, Y Lévy, C Lacabaratz, E Lhomme, A Wiedemann, C Bauduin, C Fenwick, E Foucat, M Surenaud, L Guillaumat, V Boilet, V Rieux, O Bouchaud, P-M Girard, J-M Molina, P Morlat, L Hocqueloux, L Richert, G Pantaleo, J D Lelièvre, R Thiébaut

Abstract

In this placebo-controlled phase II randomized clinical trial, 103 human immunodeficiency virus type 1 (HIV-1)-infected patients under cART (combined antiretroviral treatment) were randomized 2:1 to receive either 3 doses of DNA GTU-MultiHIV B (coding for Rev, Nef, Tat, Gag, and gp160) at week 0 (W0), W4, and W12, followed by 2 doses of LIPO-5 vaccine containing long peptides from Gag, Pol, and Nef at W20 and W24, or placebo. Analytical treatment interruption (ATI) was performed between W36 to W48. At W28, vaccinees experienced an increase in functional CD4+ T-cell responses (P < 0.001 for each cytokine compared to W0) measured, predominantly against Gag and Pol/Env, and an increase in HIV-specific CD8+ T cells producing interleukin 2 (IL-2) and tumor necrosis factor alpha (TNF-α) (P = 0.001 and 0.013, respectively), predominantly against Pol/Env and Nef. However, analysis of T-cell subsets by mass cytometry in a subpopulation showed an increase in the W28/W0 ratio for memory CD8+ T cells coexpressing exhaustion and senescence markers such as PD-1/TIGIT (P = 0.004) and CD27/CD57 (P = 0.044) in vaccinees compared to the placebo group. During ATI, all patients experienced viral rebound, with the maximum observed HIV RNA level at W42 (median, 4.63 log10 copies [cp]/ml; interquartile range [IQR], 4.00 to 5.09), without any difference between arms. No patient resumed cART for CD4 cell count drop. Globally, the vaccine strategy was safe. However, a secondary HIV transmission during ATI was observed. These data show that the prime-boost combination of DNA and LIPO-5 vaccines elicited broad and polyfunctional T cells. The contrast between the quality of immune responses and the lack of potent viral control underscores the need for combined immunomodulatory strategies. (This study has been registered at ClinicalTrials.gov under registration no. NCT01492985.)IMPORTANCE In this placebo-controlled phase II randomized clinical trial, we evaluated the safety and immunogenicity of a therapeutic prime-boost vaccine strategy using a recombinant DNA vaccine (GTU-MultiHIV B clade) followed by a boost vaccination with a lipopeptide vaccine (HIV-LIPO-5) in HIV-infected patients on combined antiretroviral therapy. We show here that this prime-boost strategy is well tolerated, consistently with previous studies in HIV-1-infected individuals and healthy volunteers who received each vaccine component individually. Compared to the placebo group, vaccinees elicited strong and polyfunctional HIV-specific CD4+ and CD8+ T-cell responses. However, these immune responses presented some qualitative defects and were not able to control viremia following antiretroviral treatment interruption, as no difference in HIV viral rebound was observed in the vaccine and placebo groups. Several lessons were learned from these results, pointing out the urgent need to combine vaccine strategies with other immune-based interventions.

Keywords: HIV; antiretroviral therapy interruption; clinical trials; human immunodeficiency virus; therapeutic vaccine.

Copyright © 2021 Lévy et al.

Figures

FIG 1
FIG 1
Trial design. (A) Schematics of study design. Blue arrows indicate time of DNA GTU-MultiHIV B or placebo administrations. Red arrows indicate time of HIV LIPO-5 or placebo administrations. ART, antiretroviral therapy; ATI, analytical treatment interruption; (B) Consolidated Standards of Reporting Trials (CONSORT) flow diagram for the trial. CONSORT diagram delineates the study enrollment of 103 participants who underwent randomization to the placebo or vaccine groups.
FIG 2
FIG 2
Functional profile of CD4+ and CD8+ T-cell responses. Production of interleukin 2 (IL-2), gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α) as measured by intracellular cytokine staining (ICS) using multiparametric flow cytometry after cell stimulation before (W0) and after vaccination (W28) in placebo (red) and therapeutic vaccine (blue) groups. (A) HIV-specific CD3+ CD4+ T-cell frequency; (B) HIV-specific CD3+ CD8+ T-cell frequency; (C) heatmap of P values between W28 and W0 of CD3+ CD4+ and CD3+ CD8+ marginal responses against Gag, Pol/Env, Nef, and the sum of HIV peptides (total HIV); (D) frequency of HIV-specific CD3+ CD4+ T cells producing 1, 2, or 3 cytokines in the vaccine group at W0 (light gray) and W28 (dark gray); (E) frequency of HIV-specific CD3+ CD8+ T cells producing 1, 2, or 3 cytokines in the vaccine group at W0 (light gray) and W28 (dark gray); (F) heatmap of P values between W28 and W0 of CD3+ CD4+ and CD3+ CD8+ polyfunctionality responses against Gag, Pol/Env, Nef, and total HIV peptides.
FIG 3
FIG 3
Mass cytometry (CyTOF) phenotyping. Ratio of memory CD8+ T cells at W28 compared to W0 for several subsets according to PD-1 and TIGIT (A), CD27 and CD57 (B), or HLA-DR and CD38 (C) in placebo (red) and therapeutic vaccine (blue) groups. Ratio of memory CD4+ T cells at W28 compared to W0 for HLA-DR and CD38 (D) in placebo (red) and therapeutic vaccine (blue) groups. P values were calculated using the Mann-Whitney test; *, P = 0.017; **, P = 0.0022.
FIG 4
FIG 4
Plasma HIV viral load and CD4+ T-cell count changes throughout the study. (A) Levels of plasma HIV RNA in the placebo (red) and therapeutic vaccine (blue) groups before and after ATI (weeks 36 to 48); (B) CD4+ T-cell count changes during the vaccination phase and following ATI in the placebo (red) and therapeutic vaccine (blue) groups before and after ATI (weeks 36 to 48).
FIG 5
FIG 5
Integrative analysis of immune response to vaccine. Principal-component analysis of ICS responses at W28. Log-transformed marginal CD4+ and CD8+ T-cell responses at W28 were included as active variables; virological markers during ATI (highest viral load, viral load slope, viral load area under the curve [AUC], and time to rebound) were included as supplementary variables. (A) Projection of variables; (B) projection of individuals represented into placebo (red) and therapeutic vaccine (blue) groups.

References

    1. Siliciano RF. 2014. Targeting reservoirs to clear and cure. Nat Med 20:480–481. 10.1038/nm.3550.
    1. Margolis DM, Garcia JV, Hazuda DJ, Haynes BF. 2016. Latency reversal and viral clearance to cure HIV-1. Science 353:aaf6517. 10.1126/science.aaf6517.
    1. Pantaleo G, Levy Y. 2016. Therapeutic vaccines and immunological intervention in HIV infection: a paradigm change. Curr Opin HIV AIDS 11:576–584. 10.1097/COH.0000000000000324.
    1. Stephenson KE. 2018. Therapeutic vaccination for HIV. Curr Opin HIV AIDS 13:408–415. 10.1097/COH.0000000000000491.
    1. Vardas E, Stanescu I, Leinonen M, Ellefsen K, Pantaleo G, Valtavaara M, Ustav M, Reijonen K. 2012. Indicators of therapeutic effect in FIT-06, a phase II trial of a DNA vaccine, GTU®-Multi-HIVB, in untreated HIV-1 infected subjects. Vaccine 30:4046–4054. 10.1016/j.vaccine.2012.04.007.
    1. Haidari G, Day S, Wood M, Ridgers H, Cope AV, Fleck S, Yan C, Reijonen K, Hannaman D, Spentzou A, Hayes P, Vogt A, Combadiere B, Cook A, McCormack S, Shattock RJ. 2019. The safety and immunogenicity of GTU®MultiHIV DNA vaccine delivered by transcutaneous and intramuscular injection with or without electroporation in HIV-1 positive subjects on suppressive ART. Front Immunol 10:2911. 10.3389/fimmu.2019.02911.
    1. Salmon-Ceron D, Durier C, Desaint C, Cuzin L, Surenaud M, Hamouda NB, Lelievre JD, Bonnet B, Pialoux G, Poizot-Martin I, Aboulker JP, Levy Y, Launay O. 2010. Immunogenicity and safety of an HIV-1 lipopeptide vaccine in healthy adults: a phase 2 placebo-controlled ANRS trial. AIDS 24:2211–2223. 10.1097/QAD.0b013e32833ce566.
    1. Richert L, Hue S, Hocini H, Raimbault M, Lacabaratz C, Surenaud M, Wiedemann A, Tisserand P, Durier C, Salmon D, Lelievre JD, Chene G, Thiebaut R, Levy Y, ANRS Vaccine Network/Vaccine Research Institute. 2013. Cytokine and gene transcription profiles of immune responses elicited by HIV lipopeptide vaccine in HIV-negative volunteers. AIDS 27:1421–1431. 10.1097/QAD.0b013e32835f5b60.
    1. Hansen SG, Marshall EE, Malouli D, Ventura AB, Hughes CM, Ainslie E, Ford JC, Morrow D, Gilbride RM, Bae JY, Legasse AW, Oswald K, Shoemaker R, Berkemeier B, Bosche WJ, Hull M, Womack J, Shao J, Edlefsen PT, Reed JS, Burwitz BJ, Sacha JB, Axthelm MK, Früh K, Lifson JD, Picker LJ. 2019. A live-attenuated RhCMV/SIV vaccine shows long-term efficacy against heterologous SIV challenge. Sci Transl Med 11:eaaw2607. 10.1126/scitranslmed.aaw2607.
    1. Julg B, Dee L, Ananworanich J, Barouch DH, Bar K, Caskey M, Colby DJ, Dawson L, Dong KL, Dubé K, Eron J, Frater J, Gandhi RT, Geleziunas R, Goulder P, Hanna GJ, Jefferys R, Johnston R, Kuritzkes D, Li JZ, Likhitwonnawut U, van Lunzen J, Martinez-Picado J, Miller V, Montaner LJ, Nixon DF, Palm D, Pantaleo G, Peay H, Persaud D, Salzwedel J, Salzwedel K, Schacker T, Sheikh V, Søgaard OS, Spudich S, Stephenson K, Sugarman J, Taylor J, Tebas P, Tiemessen CT, Tressler R, Weiss CD, Zheng L, Robb ML, Michael NL, Mellors JW, Deeks SG, Walker BD. 2019. Recommendations for analytical antiretroviral treatment interruptions in HIV research trials—report of a consensus meeting. Lancet HIV 6:e259–e268. 10.1016/S2352-3018(19)30052-9.
    1. Lelièvre J-D, Hocqueloux L. 2019. Unintended HIV-1 Transmission to a Sex Partner in a Study of a Therapeutic Vaccine candidate. J Infect Dis 220:S5–S6. 10.1093/infdis/jiz012.
    1. Lévy Y, Gahéry-Ségard H, Durier C, Lascaux AS, Goujard C, Meiffrédy V, Rouzioux C, El Habib R, Beumont-Mauviel M, Guillet JG, Delfraissy JF, Aboulker JP. 2005. Immunological and virological efficacy of a therapeutic immunization combined with interleukin-2 in chronically HIV-1 infected patients. AIDS 19:279–286.
    1. Goujard C, Marcellin F, Hendel-Chavez H, Burgard M, Meiffrédy V, Venet A, Rouzioux C, Taoufik Y, El Habib R, Beumont-Mauviel M, Aboulker J-P, Lévy Y, Delfraissy J-F, for the PRIMOVAC-ANRS 095 Study Group. 2007. Interruption of antiretroviral therapy initiated during primary HIV-1 infection: impact of a therapeutic vaccination strategy combined with interleukin (IL)-2 compared with IL-2 alone in the ANRS 095 randomized study. AIDS Res Hum Retroviruses 23:1105–1113. 10.1089/aid.2007.0047.
    1. Palich R, Ghosn J, Chaillon A, Boilet V, Nere M-L, Chaix M-L, Delobel P, Molina J-M, Lucht F, Bouchaud O, Rieux V, Thiebaut R, Levy Y, Delaugerre C, Lelievre J-D, VRI02/ANRS149 LIGHT Vaccine Trial Group. 2019. Viral rebound in semen after antiretroviral treatment interruption in an HIV therapeutic vaccine double-blind trial. AIDS 33:279–284. 10.1097/QAD.0000000000002058.
    1. Lévy Y, Durier C, Lascaux AS, Meiffrédy V, Gahéry-Ségard H, Goujard C, Rouzioux C, Resch M, Guillet JG, Kazatchkine M, Delfraissy JF, Aboulker JP, ANRS 093 Study Group. 2006. Sustained control of viremia following therapeutic immunization in chronically HIV-1-infected individuals. AIDS 20:405–413. 10.1097/01.aids.0000206504.09159.d3.
    1. Lévy Y, Thiébaut R, Montes M, Lacabaratz C, Sloan L, King B, Pérusat S, Harrod C, Cobb A, Roberts LK, Surenaud M, Boucherie C, Zurawski S, Delaugerre C, Richert L, Chêne G, Banchereau J, Palucka K. 2014. Dendritic cell-based therapeutic vaccine elicits polyfunctional HIV-specific T-cell immunity associated with control of viral load. Eur J Immunol 44:2802–2810. 10.1002/eji.201344433.
    1. Surenaud M, Lacabaratz C, Zurawski G, Lévy Y, Lelièvre JD. 2017. Development of an epitope-based HIV-1 vaccine strategy from HIV-1 lipopeptide to dendritic-based vaccines. Expert Rev Vaccines 16:955–972. 10.1080/14760584.2017.1374182.
    1. Surenaud M, Montes M, Lindestam Arlehamn CS, Sette A, Banchereau J, Palucka K, Lelièvre J-D, Lacabaratz C, Lévy Y. 2019. Anti-HIV potency of T-cell responses elicited by dendritic cell therapeutic vaccination. PLoS Pathog 15:e1008011. 10.1371/journal.ppat.1008011.
    1. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA, Abraham J, Lederman MM, Benito JM, Goepfert PA, Connors M, Roederer M, Koup RA. 2006. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood 107:4781–4789. 10.1182/blood-2005-12-4818.
    1. Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, Park S, Javinal V, Chiu H, Irving B, Eaton DL, Grogan JL. 2014. The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T-cell effector function. Cancer Cell 26:923–937. 10.1016/j.ccell.2014.10.018.
    1. Chauvin JM, Pagliano O, Fourcade J, Sun Z, Wang H, Sander C, Kirkwood JM, Chen THT, Maurer M, Korman AJ, Zarour HM. 2015. TIGIT and PD-1 impair tumor antigen-specific CD8+ T cells in melanoma patients. J Clin Invest 125:2046–2058. 10.1172/JCI80445.
    1. Chew GM, Fujita T, Webb GM, Burwitz BJ, Wu HL, Reed JS, Hammond KB, Clayton KL, Ishii N, Abdel-Mohsen M, Liegler T, Mitchell BI, Hecht FM, Ostrowski M, Shikuma CM, Hansen SG, Maurer M, Korman AJ, Deeks SG, Sacha JB, Ndhlovu LC. 2016. TIGIT marks exhausted T cells, correlates with disease progression, and serves as a target for immune restoration in HIV and SIV infection. PLoS Pathog 12:e1005349. 10.1371/journal.ppat.1005349.
    1. Sáez-Cirión A, Lacabaratz C, Lambotte O, Versmisse P, Urrutia A, Boufassa F, Barré-Sinoussi F, Delfraissy JF, Sinet M, Pancino G, Venet A, Agence Nationale de Recherches sur le Sida EP36 HIV Controllers Study Group. 2007. 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 104:6776–6781. 10.1073/pnas.0611244104.
    1. Thiébaut R, Hejblum BP, Hocini H, Bonnabau H, Skinner J, Montes M, Lacabaratz C, Richert L, Palucka K, Banchereau J, Lévy Y. 2019. Gene expression signatures associated with immune and virological responses to therapeutic vaccination with dendritic cells in HIV-infected individuals. Front Immunol 10:874. 10.3389/fimmu.2019.00874.
    1. Obermoser G, Presnell S, Domico K, Xu H, Wang Y, Anguiano E, Thompson-Snipes LA, Ranganathan R, Zeitner B, Bjork A, Anderson D, Speake C, Ruchaud E, Skinner J, Alsina L, Sharma M, Dutartre H, Cepika A, Israelsson E, Nguyen P, Nguyen QA, Harrod AC, Zurawski SM, Pascual V, Ueno H, Nepom GT, Quinn C, Blankenship D, Palucka K, Banchereau J, Chaussabel D. 2013. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity 38:831–844. 10.1016/j.immuni.2012.12.008.
    1. Kazmin D, Nakaya HI, Lee EK, Johnson MJ, Van Der Most R, Van Den Berg RA, Ballou WR, Jongert E, Wille-Reece U, Ockenhouse C, Aderem A, Zak DE, Sadoff J, Hendriks J, Wrammert J, Ahmed R, Pulendran B. 2017. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. Proc Natl Acad Sci U S A 114:2425–2430. 10.1073/pnas.1621489114.
    1. Sneller MC, Clarridge KE, Seamon C, Shi V, Zorawski MD, Justement JS, Blazkova J, Huiting ED, Proschan MA, Mora JR, Shetzline M, Moir S, Lane HC, Chun TW, Fauci AS. 2019. An open-label phase 1 clinical trial of the anti-α4β7 monoclonal antibody vedolizumab in HIV-infected individuals. Sci Transl Med 11:eaax3447. 10.1126/scitranslmed.aax3447.
    1. Lelièvre J-D. 2019. Preexposure prophylaxis for mitigating risk of HIV transmission during HIV cure–related clinical trials with a treatment interruption. J Infect Dis 220:S16–S18. 10.1093/infdis/jiz036.
    1. Moore CL, Stöhr W, Crook AM, Richert L, Leliévre JD, Pantaleo G, García F, Vella S, Lévy Y, Thiébaut R, McCormack S. 2019. Multi-arm, multi-stage randomised controlled trials for evaluating therapeutic HIV cure interventions. Lancet HIV 6:e334–e340. 10.1016/S2352-3018(19)30082-7.

Source: PubMed

3
Abonneren