Immunogenicity of personalized dendritic-cell therapy in HIV-1 infected individuals under suppressive antiretroviral treatment: interim analysis from a phase II clinical trial

Marcella Vassão de Almeida Baptista, Laís Teodoro da Silva, Sadia Samer, Telma Miyuki Oshiro, Iart Luca Shytaj, Leila B Giron, Nathalia Mantovani Pena, Nicolly Cruz, Gisele Cristina Gosuen, Paulo Roberto Abrão Ferreira, Edécio Cunha-Neto, Juliana Galinskas, Danilo Dias, Maria Cecilia Araripe Sucupira, Cesar de Almeida-Neto, Reinaldo Salomão, Alberto José da Silva Duarte, Luís Mário Janini, James R Hunter, Andrea Savarino, Maria Aparecida Juliano, Ricardo Sobhie Diaz, Marcella Vassão de Almeida Baptista, Laís Teodoro da Silva, Sadia Samer, Telma Miyuki Oshiro, Iart Luca Shytaj, Leila B Giron, Nathalia Mantovani Pena, Nicolly Cruz, Gisele Cristina Gosuen, Paulo Roberto Abrão Ferreira, Edécio Cunha-Neto, Juliana Galinskas, Danilo Dias, Maria Cecilia Araripe Sucupira, Cesar de Almeida-Neto, Reinaldo Salomão, Alberto José da Silva Duarte, Luís Mário Janini, James R Hunter, Andrea Savarino, Maria Aparecida Juliano, Ricardo Sobhie Diaz

Abstract

Background: We developed a personalized Monocyte-Derived Dendritic-cell Therapy (MDDCT) for HIV-infected individuals on suppressive antiretroviral treatment and evaluated HIV-specific T-cell responses.

Methods: PBMCs were obtained from 10 HIV+ individuals enrolled in trial NCT02961829. Monocytes were differentiated into DCs using IFN-α and GM-CSF. After sequencing each patient's HIV-1 Gag and determining HLA profiles, autologous Gag peptides were selected based on the predicted individual immunogenicity and used to pulse MDDCs. Three doses of the MDDCT were administered every 15 days. To assess immunogenicity, patients' cells were stimulated in vitro with autologous peptides, and intracellular IL-2, TNF, and interferon-gamma (IFN-γ) production were measured in CD4+ and CD8+ T-cells.

Results: The protocol of ex-vivo treatment with IFN-α and GM-CSF was able to induce maturation of MDDCs, as well as to preserve their viability for reinfusion. MDDCT administration was associated with increased expression of IL-2 in CD4+ and CD8+ T-cells at 15 and/or 30 days after the first MDDCT administration. Moreover, intracellular TNF and IFN-γ expression was significantly increased in CD4+ T-cells. The number of candidates that increased in vitro the cytokine levels in CD4+ and CD8+ T cells upon stimulation with Gag peptides from baseline to day 15 and from baseline to day 30 and day 120 after MDDCT was significant as compared to Gag unstimulated response. This was accompanied by an increasing trend in the frequency of polyfunctional T-cells over time, which was visible when considering both cells expressing two and three out of the three cytokines examined.

Conclusions: MDDC had a mature profile, and this MDDCT promoted in-vitro T-cell immune responses in HIV-infected patients undergoing long-term suppressive antiretroviral treatment. Trial registration NCT02961829: (Multi Interventional Study Exploring HIV-1 Residual Replication: a Step Towards HIV-1 Eradication and Sterilizing Cure, https://www.clinicaltrials.gov/ct2/show/NCT02961829 , posted November 11th, 2016).

Keywords: Dendritic-cell therapy; HIV GAG; HIV cure research; HLA Haplotypes; Precision medicine.

Conflict of interest statement

ILS, AS, and RSD have a patent application for Dendritic-cell therapy.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Phenotypic characterization of immature and mature MDDCs used for MDDCT. Panels A,B) PBMCs were isolated from the total blood of the enrolled individuals and induced to MDDC differentiation/maturation as depicted in Additional file 1: Fig. S1. Both immature MDDCs (iMDDC) and mature MDDCs (mMDDCs) were analyzed by flow cytometry for the expression of the activation/maturation markers HLA-DR, CD80, CD86, CD83, CD40, and CCR7 according to the gating strategy depicted in Additional file 1: Fig. S7A. Box plots and whiskers represent the median fluorescence intensity of each marker (A) or the percentage of cells expressing each marker (B). Data are expressed as median ± min/max and were analyzed by the non-parametric Wilcoxon test (N of patients = 10).** p < 0.01
Fig. 2
Fig. 2
Viability of MDDCs used for each MDDCT dose preparation. Viability was assessed by flow cytometry following staining with a LIVE/DEAD fixable stain according to the gating strategy shown in (A). The horizontal line in the graph of panel B indicates the median (N of patients for each dose = 10). Data were analyzed by Two-Way ANOVA. * p < 0.05
Fig. 3
Fig. 3
Immunogenicity of MDDCT. Ten recipients (Table 1) received three doses of the personalized MDDCT. PBMCs were collected upon administration of the first MDDCT dose (day 0), upon dose 2 (day 15), and dose 3 administration (day 30), as well as during the post-therapy follow-up (day 120). Isolated PBMCs were stimulated in vitro with the autologous Gag peptides that were used for MDDCT. Production of IFN-γ, IL-2, and TNF by CD4+ and CD8+ T-lymphocytes was evaluated by flow cytometry. After applying the logit transformation to restore normality, data of patients for whom all time points were available for a given cytokine were analyzed by one-way ANOVA followed by Dunnet’s post-test. *p < 0.05; **p < 0.01; ***p < 0.001
Fig. 4
Fig. 4
Frequency of polyfunctional CD4+ and CD8+ T-lymphocytes following MDDCT. PBMCs were collected upon administration of the first MDDCT dose (day 0), upon dose 2 (day 15), and dose 3 administration (day 30), as well as during the post-therapy follow-up (day 120). Isolated PBMCs were stimulated in vitro with the autologous Gag peptides that were used for MDDCT. Production of IFN-γ, IL-2, and TNF by CD4+ and CD8+ T-lymphocytes was evaluated by flow cytometry. Panels A,B) Frequency of cells expressing one or more immune-mediator cytokine (IL-2, TNF and IFN-γ). Pie charts show the relative percentage, among cells expressing at least one cytokine, of CD4+ (A) and CD8+ (B) T-lymphocytes expressing one, two, or three cytokines. Panels C, D) Bar graphs showing the absolute frequency of CD4+ (C) and CD8+ (D) T-lymphocytes expressing each cytokine combination analyzed. Data are expressed as mean ± SD and were analyzed by two-way ANOVA. (N of patients for each dose = 10)
Fig. 5
Fig. 5
The proportion of patients displaying an undetectable HIV-1 DNA level in rectal biopsies at the end of all treatments. Panels A,B) MDDCT recipients were subjected to rectal biopsy before all investigational interventions (baseline) and at the end of all treatments. Patients were stratified based on the concordance (or lack thereof, i.e. concordance for 50% or less of the epitopes administered) of predicted epitopes between the online tools NetMHCpan (http://www.cbs.dtu.dk/services/NetMHCpan/) and Custommune (www.custommune.com). The relative risk (RR) is referred to the risk of having detectable viral DNA as calculated by intention-to-treat (A) and per-treatment (B) analysis

References

    1. Chang JJ, Altfeld M. Innate immune activation in primary HIV-1 infection. J Infect Dis. 2010;202(Suppl 2):S297–301.
    1. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, et al. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol. 1994;68(7):4650–4655.
    1. Lu W, Arraes LC, Ferreira WT, Andrieu J-M. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat Med. 2004;10:1359–1365. doi: 10.1038/nm1147.
    1. Gray CM, Lawrence J, Schapiro JM, Altman JD, Winters MA, Crompton M, et al. Frequency of class I HLA-restricted anti-HIV CD8+ T cells in individuals receiving highly active antiretroviral therapy (HAART) J Immunol. 1999;162(3):1780–1788.
    1. Kalams SA, Goulder PJ, Shea AK, Jones NG, Trocha AK, Ogg GS, et al. Levels of human immunodeficiency virus type 1-specific cytotoxic T-lymphocyte effector and memory responses decline after suppression of viremia with highly active antiretroviral therapy. J Virol. 1999;73(8):6721–6728.
    1. Pitcher CJ, Quittner C, Peterson DM, Connors M, Koup RA, Maino VC, et al. HIV-1-specific CD4+ T cells are detectable in most individuals with active HIV-1 infection, but decline with prolonged viral suppression. Nat Med. 1999;5(5):518–525.
    1. Williams JP, Hurst J, Stohr W, Robinson N, Brown H, Fisher M, et al. HIV-1 DNA predicts disease progression and post-treatment virological control. Elife. 2014;3:e03821.
    1. O’Keeffe M, Mok WH, Radford KJ. Human dendritic cell subsets and function in health and disease. Cell Mol Life Sci. 2015;72(22):4309–4325.
    1. Surenaud M, Montes M, Arlehamn LCS, Sette A, Banchereau J, Palucka K, et al. Anti-HIV potency of T-cell responses elicited by dendritic cell therapeutic vaccination. PLoS Pathog. 2019;15(9):e1008011.
    1. García F, Climent N, Guardo AC, Gil C, León A, Autran B, et al. A dendritic cell-based vaccine elicits T cell responses associated with control of HIV-1 replication. Sci Transl Med. 2013;5(166):166ra2.
    1. Lévy Y, Thiébaut R, Montes M, Lacabaratz C, Sloan L, King B, et al. Dendritic cell-based therapeutic vaccine elicits polyfunctional HIV-specific T-cell immunity associated with control of viral load. Eur J Immunol. 2014;44(9):2802–2810.
    1. Gay CL, DeBenedette MA, Tcherepanova IY, Gamble A, Lewis WE, Cope AB, et al. Immunogenicity of AGS-004 dendritic cell therapy in patients treated during acute HIV infection. AIDS Res Hum Retroviruses. 2018;34(1):111–122.
    1. Gay CL, Kuruc JD, Falcinelli SD, Warren JA, Reifeis SA, Kirchherr JL, et al. Assessing the impact of AGS-004, a dendritic cell-based immunotherapy, and vorinostat on persistent HIV-1 Infection. Sci Rep. 2020;10(1):5134.
    1. Kundu SK, Engleman E, Benike C, Shapero MH, Dupuis M, van Schooten WC, et al. A pilot clinical trial of HIV antigen-pulsed allogeneic and autologous dendritic cell therapy in HIV-infected patients. AIDS Res Hum Retroviruses. 1998;14(7):551–560.
    1. Ide F, Nakamura T, Tomizawa M, Kawana-Tachikawa A, Odawara T, Hosoya N, et al. Peptide-loaded dendritic-cell vaccination followed by treatment interruption for chronic HIV-1 infection: a phase 1 trial. J Med Virol. 2006;78(6):711–718.
    1. Routy J-P, Boulassel M-R, Yassine-Diab B, Nicolette C, Healey D, Jain R, et al. Immunologic activity and safety of autologous HIV RNA-electroporated dendritic cells in HIV-1 infected patients receiving antiretroviral therapy. Clin Immunol. 2010;134(2):140–147.
    1. Van Gulck E, Vlieghe E, Vekemans M, Van Tendeloo VFI, Van De Velde A, Smits E, et al. mRNA-based dendritic cell vaccination induces potent antiviral T-cell responses in HIV-1-infected patients. AIDS. 2012;26(4):F1–12.
    1. Macatangay BJC, Riddler SA, Wheeler ND, Spindler J, Lawani M, Hong F, et al. Therapeutic vaccination with dendritic cells loaded with autologous HIV type 1-infected apoptotic cells. J Infect Dis. 2016;213(9):1400–1409.
    1. Jacobson JM, Routy J-P, Welles S, DeBenedette M, Tcherepanova I, Angel JB, et al. Dendritic cell immunotherapy for HIV-1 infection using autologous HIV-1 RNA: a randomized, double-blind, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;72(1):31–38.
    1. Gandhi RT, Kwon DS, Macklin EA, Shopis JR, McLean AP, McBrine N, et al. Immunization of HIV-1-infected persons with autologous dendritic cells transfected with mRNA encoding HIV-1 Gag and Nef: results of a randomized, placebo-controlled clinical trial. J Acquir Immune Defic Syndr. 2016;71(3):246–253.
    1. Imamichi H, Dewar RL, Adelsberger JW, Rehm CA, O’Doherty U, Paxinos EE, et al. Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc Natl Acad Sci U S A. 2016;113(31):8783–8788.
    1. Samer S, Namiyama G, Oshiro T, Arif MS, da Silva CW, Sucupira MCA, et al. Evidence of noncompetent HIV after ex vivo purging among ART-suppressed individuals. AIDS Res Hum Retroviruses. 2017;33(10):993–4.
    1. Williams TM. Human leukocyte antigen gene polymorphism and the histocompatibility laboratory. J Mol Diagn. 2001;3(3):98–104.
    1. Kiepiela P, Ngumbela K, Thobakgale C, Ramduth D, Honeyborne I, Moodley E, et al. CD8+ T-cell responses to different HIV proteins have discordant associations with viral load. Nat Med. 2007;13(1):46–53.
    1. Addo MM, Yu XG, Rathod A, Cohen D, Eldridge RL, Strick D, et al. Comprehensive epitope analysis of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome demonstrate broadly directed responses, but no correlation to viral load. J Virol. 2003;77(3):2081–2092.
    1. Edwards BH, Bansal A, Sabbaj S, Bakari J, Mulligan MJ, Goepfert PA. Magnitude of functional CD8+ T-cell responses to the gag protein of human immunodeficiency virus type 1 correlates inversely with viral load in plasma. J Virol. 2002;76(5):2298–2305.
    1. Zuñiga R, Lucchetti A, Galvan P, Sanchez S, Sanchez C, Hernandez A, et al. Relative dominance of Gag p24-specific cytotoxic T lymphocytes is associated with human immunodeficiency virus control. J Virol. 2006;80(6):3122–3125.
    1. Jansen CA, Kostense S, Vandenberghe K, Nanlohy NM, De Cuyper IM, Piriou E, et al. High responsiveness of HLA-B57-restricted Gag-specific CD8+ T cells in vitro may contribute to the protective effect of HLA-B57 in HIV-infection. Eur J Immunol. 2005;35(1):150–158.
    1. Julg B, Williams KL, Reddy S, Bishop K, Qi Y, Carrington M, et al. Enhanced anti-HIV functional activity associated with Gag-specific CD8 T-cell responses. J Virol. 2010;84(11):5540–5549.
    1. Pace MJ, Graf EH, Agosto LM, Mexas AM, Male F, Brady T, et al. Directly infected resting CD4+T cells can produce HIV Gag without spreading infection in a model of HIV latency. PLoS Pathog. 2012;8(7):e1002818.
    1. Graf EH, Pace MJ, Peterson BA, Lynch LJ, Chukwulebe SB, Mexas AM, et al. Gag-positive reservoir cells are susceptible to HIV-specific cytotoxic T lymphocyte mediated clearance in vitro and can be detected in vivo [corrected] PLoS One. 2013;8(8):e71879.
    1. Steers NJ, Peachman KK, McClain SR, Alving CR, Rao M. Human immunodeficiency virus type 1 Gag p24 alters the composition of immunoproteasomes and affects antigen presentation. J Virol. 2009;83(14):7049–7061.
    1. Zhao G, Perilla JR, Yufenyuy EL, Meng X, Chen B, Ning J, et al. Mature HIV-1 capsid structure by cryo-electron microscopy and all-atom molecular dynamics. Nature. 2013;497(7451):643–646.
    1. Shytaj IL, Savarino A. Cell-mediated anti-Gag immunity in pharmacologically induced functional cure of simian AIDS: a “bottleneck effect”? J Med Primatol. 2015;44:227–240. doi: 10.1111/jmp.12176.
    1. Burwitz BJ, Giraldo-Vela JP, Reed J, Newman LP, Bean AT, Nimityongskul FA, et al. CD8+ and CD4+ cytotoxic T cell escape mutations precede breakthrough SIVmac239 viremia in an elite controller. Retrovirology. 2012;9:91.
    1. Paiardini M, Müller-Trutwin M. HIV-associated chronic immune activation. Immunol Rev. 2013;254(1):78–101.
    1. Zitvogel L, Apetoh L, Ghiringhelli F, André F, Tesniere A, Kroemer G. The anticancer immune response: indispensable for therapeutic success? J Clin Invest. 2008;118(6):1991–2001.
    1. Benhar M, Shytaj IL, Stamler JS, Savarino A. Dual targeting of the thioredoxin and glutathione systems in cancer and HIV. J Clin Invest. 2016;126(5):1630–1639.
    1. Diaz RS, Shytaj IL, Giron LB, Obermaier B, Della Libera E, Galinskas J, Jr, et al. Potential impact of the antirheumatic agent auranofin on proviral HIV-1 DNA in individuals under intensified antiretroviral therapy: results from a randomised clinical trial. Int J Antimicrob Agents. 2019;54(5):592–600.
    1. Chirullo B, Sgarbanti R, Limongi D, Shytaj IL, Alvarez D, Das B, et al. A candidate anti-HIV reservoir compound, auranofin, exerts a selective “anti-memory” effect by exploiting the baseline oxidative status of lymphocytes. Cell Death Dis. 2013;4:e944.
    1. Shytaj IL, Norelli S, Chirullo B, Della Corte A, Collins M, Yalley-Ogunro J, et al. A highly intensified ART regimen induces long-term viral suppression and restriction of the viral reservoir in a simian AIDS model. PLoS Pathog. 2012;8:e1002774. doi: 10.1371/journal.ppat.1002774.
    1. Samer S, Arif MS, Giron LB, Zukurov JPL, Hunter J, Santillo BT, et al. nicotinamide activates latent HIV-1 ex vivo in ART suppressed individuals, revealing higher potency than the association of two methyltransferase inhibitors, chaetocin and BIX01294. Braz J Infect Dis. 2020;24(2):150–159.
    1. Shytaj IL, Procopio FA, Tarek M, Carlon-Andres I, Tang H-Y, Goldman AR, et al. Glycolysis downregulation is a hallmark of HIV-1 latency and sensitizes infected cells to oxidative stress. 2020. 10.1101/2020.12.30.424810
    1. Diaz RS, Zhang L, Busch MP, Mosley JW, Mayer A. Divergence of HIV-1 quasispecies in an epidemiologic cluster. AIDS. 1997;11:415–422. doi: 10.1097/00002030-199704000-00003.
    1. Sievers F, Wilm A, Dineen D, Gibson TJ, Karplus K, Li W, et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol. 2011;7:539.
    1. NetMHCpan 4.0 Server. (cited 2021 Jul 26).
    1. Automated Server Benchmarks. (cited 2021 Jul 26). .
    1. Dhanda SK, Mahajan S, Paul S, Yan Z, Kim H, Jespersen MC, et al. IEDB-AR: immune epitope database-analysis resource in 2019. Nucleic Acids Res. 2019;47(W1):W502–W506.
    1. M.T. Custommune. (cited 2021 Jul 26). .
    1. Gag CTL/CD8+ Epitope Map. (cited 2021 Jul 26). .
    1. Tarek M, Elhefnawi M, Maricato JT, Diaz RS, Shytaj IL, Savarino A. Custommune: a web tool to design personalized and population-targeted vaccine epitopes. medRxiv. 2020;2020.04.25.20079426.
    1. Komninakis SV, Santos DEM, Santos C, Oliveros MPR, Sanabani S, Diaz RS. HIV-1 proviral DNA loads (as determined by quantitative PCR) in patients subjected to structured treatment interruption after antiretroviral therapy failure. J Clin Microbiol. 2012;50(6):2132–2133.
    1. Buzón MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med. 2010;16(4):460–465.
    1. Kumar AM, Fernandez JB, Singer EJ, Commins D, Waldrop-Valverde D, Ownby RL, et al. Human immunodeficiency virus type 1 in the central nervous system leads to decreased dopamine in different regions of postmortem human brains. J Neurovirol. 2009;15(3):257–274.
    1. Sim WJ, Malinarich F, Fairhurst A-M, Connolly JE. Generation of immature, mature and tolerogenic dendritic cells with differing metabolic phenotypes. J Vis Exp. 2016 doi: 10.3791/54128.
    1. Zhou X, Fragala MS, McElhaney JE, Kuchel GA. Conceptual and methodological issues relevant to cytokine and inflammatory marker measurements in clinical research. Curr Opin Clin Nutr Metab Care. 2010;13(5):541–547.
    1. Ndhlovu ZM, Proudfoot J, Cesa K, Alvino DM, McMullen A, Vine S, et al. Elite controllers with low to absent effector CD8+ T cell responses maintain highly functional, broadly directed central memory responses. J Virol. 2012;86(12):6959–6969.
    1. Avettand-Fenoel V, Prazuck T, Hocqueloux L, Melard A, Michau C, Kerdraon R, et al. HIV-DNA in rectal cells is well correlated with HIV-DNA in blood in different groups of patients, including long-term non-progressors. AIDS. 2008;22(14):1880–1882.
    1. Viard J-P, Burgard M, Hubert J-B, Aaron L, Rabian C, Pertuiset N, et al. Impact of 5 years of maximally successful highly active antiretroviral therapy on CD4 cell count and HIV-1 DNA level. AIDS. 2004;18(1):45–49.
    1. Shytaj IL, Nickel G, Arts E, Farrell N, Biffoni M, Pal R, et al. Two-year follow-up of macaques developing intermittent control of the human immunodeficiency virus homolog simian immunodeficiency virus SIVmac251 in the chronic phase of infection. J Virol. 2015;89(15):7521–7535.
    1. Ponnan MS, Thiruvengadam K, Kathirvel S, Shankar J, Rajaraman A, Mathaiyan M, et al. Elevated numbers of HIV-specific poly-functional CD8+ T cells with stem cell-like and follicular homing phenotypes in HIV-exposed seronegative individuals. Front Immunol. 2021 doi: 10.3389/fimmu.2021.638144.
    1. Polyfunctional CD4+ T cell responses in HIV-1-infected viral controllers compared with those in healthy recipients of an adjuvanted polyprotein HIV-1 vaccine. Vaccine. 2013;31(36):3739–46.
    1. Riou C, Burgers WA, Mlisana K, Koup RA, Roederer M, Abdool Karim SS, et al. Differential impact of magnitude, polyfunctional capacity, and specificity of HIV-specific CD8+ T cell responses on HIV set point. J Virol. 2014;88(3):1819.
    1. Ross SH, Cantrell DA. Signaling and function of interleukin-2 in T lymphocytes. Annu Rev Immunol. 2018;36:411–433. doi: 10.1146/annurev-immunol-042617-053352.

Source: PubMed

3
Sottoscrivi