Single DermaVir immunization: dose-dependent expansion of precursor/memory T cells against all HIV antigens in HIV-1 infected individuals

Julianna Lisziewicz, Nyasha Bakare, Sandra A Calarota, Dénes Bánhegyi, János Szlávik, Eszter Ujhelyi, Enikő R Tőke, Levente Molnár, Zsolt Lisziewicz, Brigitte Autran, Franco Lori, Julianna Lisziewicz, Nyasha Bakare, Sandra A Calarota, Dénes Bánhegyi, János Szlávik, Eszter Ujhelyi, Enikő R Tőke, Levente Molnár, Zsolt Lisziewicz, Brigitte Autran, Franco Lori

Abstract

Background: The GIHU004 study was designed to evaluate the safety and immunogenicity of three doses of DermaVir immunization in HIV-infected subjects on fully suppressive combination antiretroviral therapy (cART).

Methodology/principal findings: This first-in-human dose escalation study was conducted with three topical DermaVir doses targeted to epidermal Langerhans cells to express fifteen HIV antigens in draining lymph nodes: 0.1 mg DNA targeted to two, 0.4 mg and 0.8 mg DNA targeted to four lymph nodes. Particularly, in the medium dose cohort 0.1 mg DNA was targeted per draining lymph node via ∼8 million Langerhans cells located in 80 cm(2) epidermis area. The 28-days study with 48-week safety follow-up evaluated HIV-specific T cell responses against Gag p17, Gag p24 and Gag p15, Tat and Rev antigens. DermaVir-associated side effects were mild, transient and not dose-dependent. Boosting of HIV-specific effector CD4(+) and CD8(+) T cells expressing IFN-gamma and IL-2 was detected against several antigens in every subject of the medium dose cohort. The striking result was the dose-dependent expansion of HIV-specific precursor/memory T cells with high proliferation capacity. In low, medium and high dose cohorts this HIV-specific T cell population increased by 325-, 136,202 and 50,759 counts after 4 weeks, and by 3,899, 9,878 and 18,382 counts after one year, respectively, compared to baseline.

Conclusions/significance: Single immunization with the DermaVir candidate therapeutic vaccine was safe and immunogenic in HIV-infected individuals. Based on the potent induction of Gag, Tat and Rev-specific memory T cells, especially in the medium dose cohort, we speculate that DermaVir boost T cell responses specific to all the 15 HIV antigens expressed from the single DNA. For durable immune reactivity repeated DermaVir immunization might be required since the frequency of DermaVir-boosted HIV-specific memory T cells decreased during the 48-week follow up.

Trial registration: ClinicalTrial.gov NCT00712530.

Conflict of interest statement

Competing Interests: Study sponsored by Commercial funder Genetic Immunity, LLC; JL, NB, ZL and FL are employees of or hold shares in Genetic Immunity Inc. There are several patent applications to protect DermaVir product and technology - JL, FL, ET from Genetic Immunity could benefit. DermaVir is a immunotherapeutic product candidate in development by Genetic Immunity. There are no other patents, products in development or marketed products to declare. This does not alter the authors’ adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. DermaVir immunization.
Figure 1. DermaVir immunization.
(a) DermaVir administration area on the skin surface under 80 cm2 large DermaPrep patches. The immunization starts with a standardized skin preparation to interrupt the stratum corneum and activate Langerhans cells causing a mild and transient erythema. (b) Cross-sectional view of the epidermis with approximately 8 million Langerhans cells estimated under one patch. (c) DermaVir pathogen-like nanomedicine, a formulation to facilitate cellular entry, nuclear transport and expression of the plasmid DNA-encoded antigens. The nanomedicine consists of the “core” that is a condensed plasmid DNA and a mannosylated polyethylenimine “envelope” , . (d) Needle-free, topical, Langerhans cell-targeting vaccine administration with the DermaPrep device. The special semi-occlusive patch is kept on the skin for three hours. (e) Activated epidermal Langerhans cells take up DermaVir, mature to antigen presenting dendritic cells and migrate to the draining lymph nodes. (f) In the lymph node dendritic cells express the DNA-encoded antigens: 15 HIV proteins that assemble into a complex VLP+ . These cells present several hundreds of epitopes to naïve T cells and prime HIV-specific precursor/memory T cells. (g) HIV-specific precursor/memory T cells with high proliferative capacity circulate in the body to kill HIV-infected cells.
Figure 2. Kinetics of the HIV-specific PHPC…
Figure 2. Kinetics of the HIV-specific PHPC responses.
Long-lived HIV-specific precursor/memory T cells with high proliferative capacity prior and after a single DermaVir immunization in each HIV-infected individual on fully suppressive cART. 1st raw: Low dose; 2nd raw: Medium Dose; 3rd raw: high dose DermaVir immunizations. Frequency of PHPC responses against Tat, Rev and the three Gag antigens are shown prior (Day 0) and after DermaVir immunization (day 7, day 14, day 28, and week 48). Every sample was assayed in duplicate or triplicate, and data are shown as PHPC counts/106 PBMC.

References

    1. Losina E, Schackman BR, Sadownik SN, Gebo KA, Walensky RP, et al. Racial and sex disparities in life expectancy losses among HIV-infected persons in the United States: impact of risk behavior, late nitiation, and early discontinuation of antiretroviral therapy. Clin Infect Dis. 2009;49:1570–1578.
    1. Haase AT. Perils at mucosal front lines for HIV and SIV and their hosts. Nat Rev Immunol. 2005;5:783–792.
    1. Shen L, Siliciano RF. Viral reservoirs, residual viremia, and the potential of highly active antiretroviral therapy to eradicate HIV infection. J Allergy Clin Immun. 2008;122:22–28.
    1. Hatano H, Hayes TL, Dahl V, Sinclair E, Lee TH, et al. A randomized, controlled trial of raltegravir intensification in antiretroviral-treated, HIV-infected patients with a suboptimal CD4+ T cell response. J Infect Dis. 2011;203(7):894–897.
    1. Dinosoa JB, Kima SY, Wiegandc AM, Palmerc SE, Ganged 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. Gandhi RT, Zheng L, Bosch RZ, Chan ES, Margolis DM, et al. The effect of Raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med. 2011;7:1–11.
    1. McMahon D, Jones J, Wiegand A, Gange SJ, Kearney M, et al. Short-course Raltegravir intensification does not reduce persistent low-level viremia in patients with HIV-1 suppression during receipt of combination antiretroviral therapy. Clin Infect Dis. 2010;50:912–919.
    1. Chapuis AG, Casper C, Kuntz S, Zhu J, Tjernlund A, et al. HIV-specific CD8+ T cells from HIV+ individuals receiving HAART can be expanded ex vivo to augment systemic and mucosal immunity in vivo. Blood. 2011;117:5391–5402.
    1. Somogyi E, Xu J, Gudics A, Toth J, Kovács A, et al. A plasmid DNA immunogen expressing fifteen protein antigens and complex virus-like particles (VLP+) mimicking naturally occurring HIV. Vaccine. 2011;29:744–753.
    1. Toke ER, Lorincz O, Somogyi E, Lisziewicz J. Rational development of a stable liquid formulation for nanomedicine products. Int J Pharm. 2010;392:261–267.
    1. Lorincz O, Toke ER, Somogyi E, Horkay F, Chandran PL, et al. Nanomedicine: Nanotechnology, Biology and Medicine; 2011. Structure and biological activity of pathogen-like synthetic nanomedicines. doi: .
    1. Lisziewicz J, Gabrilovich DI, Varga G, Xu J, Greenberg PD, et al. Induction of potent human immunodeficiency virus type 1-specific T-cell-restricted immunity by genetically modified dendritic cells. J Virol. 2001;75:7621–7628.
    1. Lisziewicz J, Trocio J, Whitman L, Varga G, Xu J, et al. DermaVir: a novel topical vaccine for HIV/AIDS. J Invest Dermatol. 2005;124:160–169.
    1. Cristillo AD, Lisziewicz J, He L, Lori F, Galmin L, et al. HIV-1 prophylactic vaccine comprised of topical DermaVir prime and protein boost elicits cellular immune responses and controls pathogenic R5 SHIV162P3. Virology. 2007;366:197–211.
    1. Lisziewicz J, Trocio J, Xu J, Whitman L, Ryder A, et al. Control of viral rebound through therapeutic immunization with DermaVir. AIDS. 2005;19:35–43.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation and maintenance. Annu Rev Immunol 22:7. 2004;45–763
    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.
    1. Jansen CA, De Cuyper IM, Hooibrink B, van der Bij AK, van Baarle D, et al. Prognostic value of HIV-1 Gag-specific CD4+ T-cell responses for progression to AIDS analyzed in a prospective cohort study. Blood. 2006;107:1427–1433.
    1. Schellens IMM, Borghans JAM, Jansen CA, De Cuyper IM, Geskus RB, et al. Abundance of early functional HIV-specific CD8+ T cells does not predict AIDS-free survival time. PLoSOne. 2008;3:1–8.
    1. Keating SM, Bejon P, Berthoud T, Vuola JM, Todryk S, et al. Durable human memory T cells quantifiable by Cultured Enzyme-Linked Immunospot Assays are induced by heterologous prime boost immunization and correlate with protection against malaria. J Immunol. 2005;175:5675–5680.
    1. Calarota SA, Foli A, Maserati R, Baldanti F, Paolucci S, et al. HIV-1-specific T cell precursors with high proliferative capacity correlate with low viremia and high CD4 counts in untreated individuals. J Immunol. 2008;180:5907–5915.
    1. Flanagan KL, Lee EAM, Gravenor MB, Reece WHH, Urban BC, et al. Unique T cell effector functions elicited by plasmodium falciparum epitopes in malaria-exposed africans tested by three T cell assays. J Immunol. 2001;167:4729–4737.
    1. Godkin AJ, Thomas HC, Openshaw PJ. Evolution of epitope-specific memory CD4 T cells after clearance of Hepatitis C virus. J Immunol. 2002;169:2210–2214.
    1. Matzinger P. The danger model: a renewed sense of self. Science. 2002;296:301–305.
    1. Holzmann S, Tripp CH, Schmuth M, Janke K, Franz K, et al. A Model System Using Tape Stripping for Characterization of Langerhans Cell-Precursors In Vivo. J Invest Dermatol 122: 1165. 2004;–1174
    1. Ondondo BO, Yang H, Dong T, di Gleria K, Suttill A, et al. Immunisation with recombinant modified vaccinia virus Ankara expressing HIV-1 gag in HIV-1-infected subjects stimulates broad functional CD4+ T cell responses. Eur J Immunol. 2006;36:2585–2594.
    1. Notermans DW, de Wolf F, Oudshoorn P, Cuijpers HT, Pirillo M, et al. Evaluation of a second-generation nucleic acid sequence-based amplification assay for quantification of HIV type 1 RNA and the use of ultrasensitive protocol adaptations. AIDS Res Hum Retroviruses. 2000;16:1507–1517.
    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;75:6508–6516.
    1. Koup RA, Safrit JT, Cao Y, Andrews CA, MLeod G, 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;68:4650–4655.
    1. Natz E, Lisziewicz J. Rational design of formulated DNA vaccine: The DermaVir approach. In: Thalhamer J, Weiss R, Scheiblhofer S, editors. Gene vaccines. SpringerWienNew York; 2011. pp. 127–143.
    1. Kitahata MM, Gange SJ, Abraham AG, Merriman B, Saag MS, et al. Effect of early versus deferred antiretroviral therapy for HIV on survival. N Engl J Med. 2009;360:1815–1826.
    1. Thompson MA, Aberg JA, Cahn P, Montaner JSG, Rizzardini G, et al. Antiretroviral Treatment of Adult HIV Infection: 2010 Recommendations of the International AIDS Society–USA Panel. JAMA. 2010;304:321–333.
    1. Ladell K, Hellerstein MK, Cesar D, Busch R, Boban D, et al. Central memory CD8+ T cells appear to have a shorter lifespan and reduced abundance as a function of HIV disease progression. J Immunol. 2008;180:7907–7918.

Source: PubMed

3
Předplatit