Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection

Lorna Leal, Alberto C Guardo, Sara Morón-López, Maria Salgado, Beatriz Mothe, Carlo Heirman, Pieter Pannus, Guido Vanham, Henk Jan van den Ham, Rob Gruters, Arno Andeweg, Sonja Van Meirvenne, Judit Pich, Joan Albert Arnaiz, Josep M Gatell, Christian Brander, Kris Thielemans, Javier Martínez-Picado, Montserrat Plana, Felipe García, iHIVARNA consortium, Lorna Leal, Alberto C Guardo, Sara Morón-López, Maria Salgado, Beatriz Mothe, Carlo Heirman, Pieter Pannus, Guido Vanham, Henk Jan van den Ham, Rob Gruters, Arno Andeweg, Sonja Van Meirvenne, Judit Pich, Joan Albert Arnaiz, Josep M Gatell, Christian Brander, Kris Thielemans, Javier Martínez-Picado, Montserrat Plana, Felipe García, iHIVARNA consortium

Abstract

Objective: The efficacy of therapeutic vaccines against HIV-1 infection has been modest. New inerts to redirect responses to vulnerable sites are urgently needed to improve these results.

Design: We performed the first-in-human clinical trial with naked mRNA (iHIVARNA) combining a dendritic cell activation strategy (TriMix:CD40L+CD70+caTLR4 RNA) with a novel HIV immunogen sequences (HTI immunogen).

Methods: A dose escalation, phase I clinical trial was performed in 21 chronic HIV-1-infected patients under ART who received three intranodal doses of mRNA (weeks 0, 2 and 4) as follow: TriMix-100 g, TriMix-300 g, TriMix-300 g with HTI-300 g, TriMix-300 g with HTI-600 g, TriMix-300 g with HTI-900 g. Primary end-point was safety and secondary-exploratory end-points were immunogenicity, changes in viral reservoir and transcriptome.

Results: Overall, the vaccine was secure and well tolerated. There were 31 grade 1/2 and 1 grade 3 adverse events, mostly unrelated to the vaccination. Patients who received the highest dose showed a moderate increase in T-cell responses spanning HTI sequence at week 8. In addition, the proportion of responders receiving any dose of HTI increased from 31% at w0 to 80% postvaccination. The intervention had no impact on caHIV-DNA levels, however, caHIV-RNA expression and usVL were transiently increased at weeks 5 and 6 in the highest dose of iHIVARNA, and these changes were positively correlated with HIV-1-specific-induced immune responses.

Conclusion: This phase I dose-escalating trial showed that iHIVARNA administration was safe and well tolerated, induced moderate HIV-specific T-cell responses and transiently increased different viral replication readouts. These data support further exploration of iHIVARNA in a phase II study. CLINICALTRIALS.

Gov identifier: NCT02413645.

Figures

Fig. 1
Fig. 1
(a) Dose escalation flow chart. (b) Assignation of cohorts.
Fig. 2
Fig. 2
pETheRNA mRNA vector.
Fig. 3
Fig. 3
Changes in the magnitude of total HIV-1-specific immune responses against IN and OUT peptide pools as measured by ELISPOT (at week 0, 4, 6, 8 and 24).
Fig. 4
Fig. 4
(a) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 cell-associated total DNA. (b) Impact of TriMix (Groups 1 and 2) or the different doses of iHIVARNA (Groups 3–5) in HIV-1 cell-associated RNA. (c) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 ultrasensitive plasma RNA
Fig. 4 (Continued)
Fig. 4 (Continued)
(a) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 cell-associated total DNA. (b) Impact of TriMix (Groups 1 and 2) or the different doses of iHIVARNA (Groups 3–5) in HIV-1 cell-associated RNA. (c) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 ultrasensitive plasma RNA
Fig. 4 (Continued)
Fig. 4 (Continued)
(a) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 cell-associated total DNA. (b) Impact of TriMix (Groups 1 and 2) or the different doses of iHIVARNA (Groups 3–5) in HIV-1 cell-associated RNA. (c) Impact of the highest doses of iHIVARNA (Groups 4 and 5) in HIV-1 ultrasensitive plasma RNA
Fig. 5
Fig. 5
Reactome gene set analysis.

References

    1. Palella FJ, Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338:853–860.
    1. Lima VD, Lourenço L, Yip B, Hogg RS, Phillips P, Montaner JSG. AIDS incidence and AIDS-related mortality in British Columbia, Canada, between 1981 and 2013: a retrospective study. Lancet HIV 2015; 2:e92–e97.
    1. Marrazzo JM, del Rio C, Holtgrave DR, Cohen MS, Kalichman SC, Mayer KH, et al. International Antiviral Society-USA Panel. HIV prevention in clinical care settings: 2014 recommendations of the International Antiviral Society-USA Panel. JAMA 2014; 312:390–409.
    1. Gardner EM, McLees MP, Steiner JF, Del Rio C, Burman WJ. The spectrum of engagement in HIV care and its relevance to test-and-treat strategies for prevention of HIV infection. Clin Infect Dis 2011; 52:793–800.
    1. Campbell C, Ambrosioni J, Miro JM, Esteve A, Casabona J, Navarro G, et al. The continuum of HIV care in Catalonia. AIDS Care 2015; 27:1449–1454.
    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; 9:727–728.
    1. Beyrer C, Pozniak A. HIV drug resistance — an emerging threat to epidemic control. New Engl J Med 2017; 377:1605–1607.
    1. Deeks SG, Autran B, Berkhout B, Benkirane M, Cairns S, Chomont N, et al. Towards an HIV cure: a global scientific strategy. Nat Rev Immunol 2012; 12:607–614.
    1. Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P, et al. International AIDS Society global scientific strategy: towards an HIV cure 2016. Nat Med 2016; 22:839–850.
    1. Leal L, Lucero C, Gatell JM, Gallart T, Plana M, Garcia F. New challenges in therapeutic vaccines against HIV infection. Expert Rev Vaccines 2017; 16:587–600.
    1. Garcia F, Climent N, Guardo AC, Gil C, Leon 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; 5:166ra2.
    1. Papagno L, Alter G, Assoumou L, Murphy RL, Garcia F, Clotet B, et al. ORVACS Study Group. Comprehensive analysis of virus-specific T-cells provides clues for the failure of therapeutic immunization with ALVAC-HIV vaccine. Aids 2011; 25:27–36.
    1. Mothe B, Llano A, Ibarrondo J, Daniels M, Miranda C, Zamarreno J, et al. Definition of the viral targets of protective HIV-1-specific T cell responses. J Transl Med 2011; 9:208.
    1. Petsch B, Schnee M, Vogel AB, Lange E, Hoffmann B, Voss D, et al. Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection. Nat Biotechnol 2012; 30:1210–1216.
    1. Weide B, Carralot JP, Reese A, Scheel B, Eigentler TK, Hoerr I, et al. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J Immunother 2008; 31:180–188.
    1. Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, et al. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother 2009; 32:498–507.
    1. Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, nonrandomised, prospective, first-in-human phase 1 clinical trial. Lancet 2017; 390:
    1. Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, et al. Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther 2011; 19:990–999.
    1. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017; 547:222–226.
    1. Van Lint S, Goyvaerts C, Maenhout S, Goethals L, Disy A, Benteyn D, et al. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res 2012; 72:1661–1671.
    1. Guardo AC, Joe PT, Miralles L, Bargallo ME, Mothe B, Krasniqi A, et al. iHIVARNA. Preclinical evaluation of an mRNA HIV vaccine combining rationally selected antigenic sequences and adjuvant signals (HTI-TriMix). AIDS 2017; 31:321–332.
    1. Mothe B, Hu X, Llano A, Rosati M, Olvera A, Kulkarni V, et al. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J Transl Med 2015; 13:60.
    1. Martinez-Bonet M, Puertas MC, Fortuny C, Ouchi D, Mellado MJ, Rojo P, et al. Establishment and replenishment of the viral reservoir in perinatally HIV-1-infected children initiating very early antiretroviral therapy. Clin Infect Dis 2015; 61:1169–1178.
    1. Moron-Lopez S, Puertas MC, Galvez C, Navarro J, Carrasco A, Esteve M, et al. Sensitive quantification of the HIV-1 reservoir in gut-associated lymphoid tissue. PLoS One 2017; 12:e0175899.
    1. Fabregat A, Sidiropoulos K, Viteri G, Forner O, Marin-Garcia P, Arnau V, et al. Reactome pathway analysis: a high-performance in-memory approach. BMC Bioinformatics 2017; 18:142.
    1. Milacic M, Haw R, Rothfels K, Wu G, Croft D, Hermjakob H, et al. Annotating cancer variants and anticancer therapeutics in reactome. Cancers 2012; 4:1180–1211.
    1. Youn H, Chung JK. Modified mRNA as an alternative to plasmid DNA (pDNA) for transcript replacement and vaccination therapy. Expert Opin Biol Ther 2015; 15:1337–1348.
    1. Deng K, Pertea M, Rongvaux A, Wang L, Durand CM, Ghiaur G, et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 2015; 517:381–385.
    1. Mothe B, Manzardo C, Coll P, Cobarsi P, Sanchez A, Escrig R, et al. Safety and immunogenicity of ChAd.HIVconsv and MVA.HIVconsv therapeutic vaccines in a cohort of early treated HIV—1 infected individuals. 8th IAS Conference on HIV Pathogenesis, Treatment & Prevention Vancouver, Canada. 2015
    1. Im EJ, Hong JP, Roshorm Y, Bridgeman A, Letourneau S, Liljestrom P, et al. Protective efficacy of serially up-ranked subdominant CD8+ T cell epitopes against virus challenges. PLoS Pathog 2011; 7:e1002041.
    1. Borthwick N, Ahmed T, Ondondo B, Hayes P, Rose A, Ebrahimsa U, et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther 2014; 22:464–475.
    1. Kulkarni V, Valentin A, Rosati M, Alicea C, Singh AK, Jalah R, et al. Altered response hierarchy and increased T-cell breadth upon HIV-1 conserved element DNA vaccination in macaques. Plos One 2014; 9:e86254.
    1. Letourneau S, Im EJ, Mashishi T, Brereton C, Bridgeman A, Yang H, et al. Design and preclinical evaluation of a universal HIV-1 vaccine. PLoS One 2007; 2:e984.
    1. Streeck H, Frahm N, Walker BD. The role of IFN-gamma Elispot assay in HIV vaccine research. Nat Protoc 2009; 4:461–469.
    1. Schmittel A, Keilholz U, Bauer S, Kuhne U, Stevanovic S, Thiel E, et al. Application of the IFN-gamma ELISPOT assay to quantify T cell responses against proteins. J Immunol Methods 2001; 247:17–24.
    1. Flynn BJ, Kastenmuller K, Wille-Reece U, Tomaras GD, Alam M, Lindsay RW, et al. Immunization with HIV Gag targeted to dendritic cells followed by recombinant New York vaccinia virus induces robust T-cell immunity in nonhuman primates. Proc Natl Acad Sci U S A 2011; 108:7131–7136.
    1. Emery S, Kelleher AD, Workman C, Puls RL, Bloch M, Baker D, et al. Influence of IFNgamma co-expression on the safety and antiviral efficacy of recombinant fowlpox virus HIV therapeutic vaccines following interruption of antiretroviral therapy. Hum Vaccin 2007; 3:260–267.
    1. Emery S, Workman C, Puls RL, Bloch M, Baker D, Bodsworth N, et al. Randomized, placebo-controlled, phase I/IIa evaluation of the safety and immunogenicity of fowlpox virus expressing HIV gag-pol and interferon-gamma in HIV-1 infected subjects. Hum Vaccin 2005; 1:232–238.

Source: PubMed

3
Tilaa