iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy

Wesley de Jong, Joeri Aerts, Sabine Allard, Christian Brander, Jozefien Buyze, Eric Florence, Eric van Gorp, Guido Vanham, Lorna Leal, Beatriz Mothe, Kris Thielemans, Montse Plana, Félipe Garcia, Rob Gruters, iHIVARNA consortium, F G, José M Gatell, Joan-Albert Arnaiz, M P, L L, Albert Guardo, Maria José Maleno, G V, E F, Pieter Pannus, J B, Leo Heyndrickx, K T, Joeri Aerts, S A, Patrick Tjok, C B, B M, J Martinez-Picado, Alex Olvera, Miriam Rosas, Maria Salgado, Sara Moron, Jose Moltó, Miriam López, R G, Marion Koopmans, W J, Patrick Boers, Rachel Scheuer, Cynthia Lungu, Carlo Heirman, Sonja Van Meirvenne, Anna Graupera, Ángel Honrado, Wesley de Jong, Joeri Aerts, Sabine Allard, Christian Brander, Jozefien Buyze, Eric Florence, Eric van Gorp, Guido Vanham, Lorna Leal, Beatriz Mothe, Kris Thielemans, Montse Plana, Félipe Garcia, Rob Gruters, iHIVARNA consortium, F G, José M Gatell, Joan-Albert Arnaiz, M P, L L, Albert Guardo, Maria José Maleno, G V, E F, Pieter Pannus, J B, Leo Heyndrickx, K T, Joeri Aerts, S A, Patrick Tjok, C B, B M, J Martinez-Picado, Alex Olvera, Miriam Rosas, Maria Salgado, Sara Moron, Jose Moltó, Miriam López, R G, Marion Koopmans, W J, Patrick Boers, Rachel Scheuer, Cynthia Lungu, Carlo Heirman, Sonja Van Meirvenne, Anna Graupera, Ángel Honrado

Abstract

Background: HIV therapeutic vaccination aims to improve the immune responses against HIV in order to control viral replication without the need for combined antiretroviral therapy (cART). iHIVARNA-01 is a novel vaccine combining mRNA delivery and T-cell immunogen (HTI) based on conserved targets of effective antiviral T-cell responses. In addition, it holds adequate stimuli required for activating antigen presenting cells (APC)s and co-activating specific T-cells (TriMix), including human CD40L, constitutively active TLR4 (caTLR4) and CD70. We propose that in-vivo targeting of dendritic cells (DCs) by direct administration of a HIV mRNA encoding these immune modulating proteins might be an attractive alternative to target DCs in vitro.

Methods/design: This is a phase-IIa, randomized, double-blinded, placebo-controlled, multicenter study in chronically HIV-1 infected patients under stable cART. One of the three study arms is randomly allocated to subjects. Three vaccinations with either HIVACAT T-cell immunogen (HTI)-TriMix (iHIVARNA-01), TriMix or water for injection (WFI) (weeks 0, 2 and 4) are administered by intranodal injection in the inguinal region. Two weeks after the last immunization (week 6) cART is stopped for 12 weeks. The two primary endpoints are: (1) safety and tolerability of intranodal iHIVARNA-01 vaccination compared with TriMix or WFI and (2) induced immunogenicity, i.e., increase in the frequency of HIV-specific T-cell responses between baseline, week 6 and 12 weeks after treatment interruption in iHIVARNA-01-treated patients as compared to the control groups, immunized with TriMix-mRNA or WFI measured by an IFNγ ELISPOT assay. Secondary endpoints include the evaluation of time to viral rebound, plasma viral load (pVL) at w18, the proportion of patients with control of viral load, induction of T-cell responses to new HIV epitopes, polyfunctionality of HIV-specific T-cells, CD8+ T-cell in-vitro HIV suppressive capacity, the effect on viral reservoir (measured by proviral DNA and cell-associated RNA), assessment of viral immune escape by mutation and mRNA expression profiles of host immune genes.

Discussion: This trial aims to direct target DC in situ with mRNA encoding HTI and TriMix for co-stimulation. Intranodal injection circumvents laborious DC isolation and handling in the laboratory. The trial extends on the safety results of a phase-I dose-escalating trial. This candidate vaccine could complement or even replace cART for chronic HIV infection and could be applicable to improve the care and cost of HIV infection.

Trial registration: EudraCT 2016-002724-83 (22 September 2016); ClinicalTrials.gov, ID: NCT02888756 . Registered on 23 August 2016.

Keywords: Antigen-presenting cell; Functional cure; HIV-1; Immunotherapy; Lymph node; Reservoir; Therapeutic vaccine; TriMix.

Conflict of interest statement

CB and BM are co-inventors of the HTI immunogen sequence. CB is CSO of, and receives compensation from, AELIX Therapeutics, a Barcelona biotech company developing HTI in the context of different vaccine vectors. BM is a consultant for AELIX Therapeutics. KT is the founder and SCO of eTheRNA and receives compensation from eTheRNA, a Brussels immunotherapy spin-off company of Vrije Universiteit Brussel. All other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Overview of the study period, interventions and assessments
Fig. 2
Fig. 2
Screening, assessments and interventions schedule for iHIVARNA phase IIa. 1 screening at most 8 weeks prior to enrollment at W0. Interval of 4 to 2 weeks is advised and optional consideration period of up to 2 weeks is offered. 2F full visit, L laboratory alone, P phone call. 3 full physical examination vs. vitals/lymph nodes alone. A full physical examination must be performed during follow-up when clinically indicated. 4 applicable to female subjects (reproductive age and not anatomically sterilized). Intranodal injection and treatment interruption is only allowed after a negative pregnancy test. 5 laboratory tests for safety includes hemoglobin, hematocrit, leucocytes, antibodies. Neutrophil count, thrombocytes, creatinine, aspartate aminotransferase (ASAT), alanine transaminase (ALAT), gamma-glutamyl transpeptidase (GGT), alkaline phosphatase (ALP), total bilirubin, amylase (or lipase), creatine kinase (CK, known as creatine phosphokinase), glucose, CD4+ and CD8+ cells and HIV viral load. 6 follow-up of vital signs (RR/pulse) at 5 min and 60 min after injection. 7 cART treatment interruption (ATI) starting from week 6 up to week 18. When there is an undetectable viral load at week 18, ATI is continued until week 30. Schedule according to template available from SPIRIT Statement,© 2013 Chan et al. [12]

References

    1. WHO. HIV/AIDS data and statistics: World Health Organization; 2017. Available from: . Accessed 11 May 2017.
    1. Phillips AN, Eron JJ, Bartlett JA, Rubin M, Johnson J, Price S, et al. HIV-1 RNA levels and the development of clinical disease. North American Lamivudine HIV Working Group. AIDS. 1996;10(8):859–865. doi: 10.1097/00002030-199607000-00009.
    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(6):727–728. doi: 10.1038/nm880.
    1. WHO. Prevent HIV, test and treat all—WHO support for country impact. Progress report 2016: WHO; 2016. . Accessed 11 May 2017.
    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(6):587–600. doi: 10.1080/14760584.2017.1322513.
    1. Barouch DH, Deeks SG. Immunologic strategies for HIV-1 remission and eradication. Science. 2014;345(6193):169–174. doi: 10.1126/science.1255512.
    1. Carcelain G, Autran B. Immune interventions in HIV infection. Immunol Rev. 2013;254(1):355–371. doi: 10.1111/imr.12083.
    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(166):166ra2. doi: 10.1126/scitranslmed.3004682.
    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. doi: 10.1186/s12967-015-0392-5.
    1. Bonehill A, Tuyaerts S, Van Nuffel AM, Heirman C, Bos TJ, Fostier K, et al. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol Ther. 2008;16(6):1170–1180. doi: 10.1038/mt.2008.77.
    1. AETC. HIV Classification: CDC and WHO Staging Systems: AIDS Education & Training Center Program; 2014. [Available from: . Accessed 11 May 2017.
    1. Chan AW, Tetzlaff JM, Gotzsche PC, Altman DG, Mann H, Berlin JA, et al. SPIRIT 2013 explanation and elaboration: guidance for protocols of clinical trials. BMJ. 2013;346:e7586. doi: 10.1136/bmj.e7586.
    1. Calin R, Hamimi C, Lambert-Niclot S, Carcelain G, Bellet J, Assoumou L, et al. Treatment interruption in chronically HIV-infected patients with an ultralow HIV reservoir. AIDS. 2016;30(5):761–769. doi: 10.1097/QAD.0000000000000987.
    1. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science. 2009;323(5919):1304–1307. doi: 10.1126/science.1165706.
    1. Mothe B, Llano A, Ibarrondo J, Zamarreno J, Schiaulini M, Miranda C, et al. CTL responses of high functional avidity and broad variant cross-reactivity are associated with HIV control. PLoS One. 2012;7(1):e29717. doi: 10.1371/journal.pone.0029717.
    1. Van Lint S, Renmans D, Broos K, Goethals L, Maenhout S, Benteyn D, et al. Intratumoral delivery of TriMix mRNA results in T-cell activation by cross-presenting dendritic cells. Cancer Immunol Res. 2016;4(2):146–156. doi: 10.1158/2326-6066.CIR-15-0163.
    1. Van Lint S, Wilgenhof S, Heirman C, Corthals J, Breckpot K, Bonehill A, et al. Optimized dendritic cell-based immunotherapy for melanoma: the TriMix-formula. Cancer Immunol Immunother. 2014;63(9):959–967. doi: 10.1007/s00262-014-1558-3.
    1. Borducchi EN, Cabral C, Stephenson KE, Liu J, Abbink P, Ng'ang'a D, et al. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature. 2016;540(7632):284–287. doi: 10.1038/nature20583.
    1. Allard SD, De Keersmaecker B, de Goede AL, Verschuren EJ, Koetsveld J, Reedijk ML, et al. A phase I/IIa immunotherapy trial of HIV-1-infected patients with Tat, Rev and Nef expressing dendritic cells followed by treatment interruption. Clin Immunol. 2012;142(3):252–268. doi: 10.1016/j.clim.2011.10.010.
    1. Kauffman KJ, Dorkin JR, Yang JH, Heartlein MW, DeRosa F, Mir FF, et al. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano Lett. 2015;15(11):7300–7306. doi: 10.1021/acs.nanolett.5b02497.
    1. Oberli MA, Reichmuth AM, Dorkin JR, Mitchell MJ, Fenton OS, Jaklenec A, et al. Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy. Nano Lett. 2017;17(3):1326–1335. doi: 10.1021/acs.nanolett.6b03329.
    1. Tavernier G, Andries O, Demeester J, Sanders NN, De Smedt SC, Rejman J. mRNA as gene therapeutic: how to control protein expression. J Control Release. 2011;150(3):238–247. doi: 10.1016/j.jconrel.2010.10.020.
    1. Van Lint S, Heirman C, Thielemans K, Breckpot K. mRNA: from a chemical blueprint for protein production to an off-the-shelf therapeutic. Hum Vaccin Immunother. 2013;9(2):265–274. doi: 10.4161/hv.22661.
    1. Ruiz-Riol M, Llano A, Ibarrondo J, Zamarreno J, Yusim K, Bach V, et al. Alternative effector-function profiling identifies broad HIV-specific T-cell responses in highly HIV-exposed individuals who remain uninfected. J Infect Dis. 2015;211(6):936–946. doi: 10.1093/infdis/jiu534.
    1. Dock J, Hultin L, Hultin P, Elliot J, Yang OO, Anton PA, et al. Human immune compartment comparisons: optimization of proliferative assays for blood and gut T lymphocytes. J Immunol Methods. 2017;445:77–87. doi: 10.1016/j.jim.2017.03.014.
    1. Lambotte O, Pollara J, Boufassa F, Moog C, Venet A, Haynes BF, et al. High antibody-dependent cellular cytotoxicity responses are correlated with strong CD8 T cell viral suppressive activity but not with B57 status in HIV-1 elite controllers. PLoS One. 2013;8(9):e74855. doi: 10.1371/journal.pone.0074855.
    1. Van Gulck E, Vlieghe E, Vekemans M, Van Tendeloo VF, 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. doi: 10.1097/QAD.0b013e32834f33e8.
    1. de Goede AL, Andeweg AC, van den Ham HJ, Bijl MA, Zaaraoui-Boutahar F, van IJcken WF, et al. DC immunotherapy in HIV-1 infection induces a major blood transcriptome shift. Vaccine. 2015;33(25):2922–2929. doi: 10.1016/j.vaccine.2015.04.047.
    1. de Goede AL, van Deutekom HW, Vrancken B, Schutten M, Allard SD, van Baalen CA, et al. HIV-1 evolution in patients undergoing immunotherapy with Tat, Rev, and Nef expressing dendritic cells followed by treatment interruption. AIDS. 2013;27(17):2679–2689. doi: 10.1097/01.aids.0000433813.67662.92.

Source: PubMed

3
Se inscrever