Molecular Signatures of a TLR4 Agonist-Adjuvanted HIV-1 Vaccine Candidate in Humans

Jenna Anderson, Thorunn A Olafsdottir, Sven Kratochvil, Paul F McKay, Malin Östensson, Josefine Persson, Robin J Shattock, Ali M Harandi, Jenna Anderson, Thorunn A Olafsdottir, Sven Kratochvil, Paul F McKay, Malin Östensson, Josefine Persson, Robin J Shattock, Ali M Harandi

Abstract

Systems biology approaches have recently provided new insights into the mechanisms of action of human vaccines and adjuvants. Here, we investigated early transcriptional signatures induced in whole blood of healthy subjects following vaccination with a recombinant HIV-1 envelope glycoprotein subunit CN54gp140 adjuvanted with the TLR4 agonist glucopyranosyl lipid adjuvant-aqueous formulation (GLA-AF) and correlated signatures to CN54gp140-specific serum antibody responses. Fourteen healthy volunteers aged 18-45 years were immunized intramuscularly three times at 1-month intervals and whole blood samples were collected at baseline, 6 h, and 1, 3, and 7 days post first immunization. Subtle changes in the transcriptomic profiles were observed following immunization, ranging from over 300 differentially expressed genes (DEGs) at day 1 to nearly 100 DEGs at day 7 following immunization. Functional pathway analysis revealed blood transcription modules (BTMs) related to general cell cycle activation, and innate immune cell activation at early time points, as well as BTMs related to T cells and B cell activation at the later time points post-immunization. Diverse CN54gp140-specific serum antibody responses of the subjects enabled their categorization into high or low responders, at early (<1 month) and late (up to 6 months) time points post vaccination. BTM analyses revealed repression of modules enriched in NK cells, and the mitochondrial electron chain, in individuals with high or sustained antigen-specific antibody responses. However, low responders showed an enhancement of BTMs associated with enrichment in myeloid cells and monocytes as well as integrin cell surface interactions. Flow cytometry analysis of peripheral blood mononuclear cells obtained from the subjects revealed an enhanced frequency of CD56dim NK cells in the majority of vaccines 14 days after vaccination as compared with the baseline. These results emphasize the utility of a systems biology approach to enhance our understanding on the mechanisms of action of TLR4 adjuvanted human vaccines.

Trial registration: ClinicalTrials.gov NCT01966900.

Keywords: HIV vaccine; TLR4; adjuvant; glucopyranosyl lipid adjuvant-aqueous formulation; systems vaccinology; transcriptomics.

Figures

Figure 1
Figure 1
Transcriptional changes in gene expression over time following vaccination. Subjects were intramuscularly immunized with CN54gp140 + GLA. In the Venn diagram, (A) the number of unique and shared differentially expressed genes (DEGs) in whole blood at 6 h, 1 day, 3 days, and 7 days after immunization (compared to baseline, 0 h) are shown, while the numbers and magnitudes (y-axis) of upregulated (red dots) or downregulated (blue dots) DEGs at 6 h (B), 1 day (C), 3 days (D), and 7 days (E), are depicted in volcano plots. An overview of all DEGs hierarchically clustered and arranged by time, are shown in a heat map (F), according to the color legend (upregulated DEGs in red, downregulated in blue, log2FC scale).
Figure 2
Figure 2
Enrichment of blood transcription modules (BTMs) regulated upon vaccination. Spider web chart indicating enriched BTMs at 6 h (red), 1 day (yellow), 3 days (blue), and 7 days (green). BTMs are functionally divided into: cell cycle regulation and signaling (A), innate immune cells and activation (B), and B and T cell activation and signaling (C). Only BTMs with FDR q values < 0.05 at at least one of the four time points (those time points are marked with asterisk) are considered. Points inside the dashed blue circle are negative enrichment and points outside the blue circle are positive enrichment.
Figure 3
Figure 3
Molecular signatures associated with early (A) and late (B) humoral responses. Blood transcription modules (BTMs) at early time points (6 h, 1 day, 3 days, and 7 days) associated with IgA (measured at 14 days), IgG (measured at 28 days), and IgM (measured at 28 days) responses in high and low responders (A). BTMs at early time points (6 h, 1 day, 3 days, and 7 days) associating with IgA, IgG, IgG1, IgG2a, IgG3, and IgG4, all measured at 168 days except IgG3 (measured at 84 days) responses in high and low responders (B). Size of the circles indicate level of significance [−log10(FDR)]. Only BTMs with FDR q value < 0.25 are included in the plot. Negative association is indicated by blue color and positive association is indicated by red color.
Figure 4
Figure 4
Frequency of CD56dim NK cell was enhanced in high antibody responders 14 days post vaccination. FACS plots showing NK cell populations of representative low (no IgM 14 days and low IgG 168 days) and high (IgM response 14 days and high IgG 168 days) responder before (D0) and 14 days following vaccination (A). Average frequency of the CD56bright and CD56dim NK cell populations at 0 h and 14 days (B). Log2 fold change of CD56bright and CD56dim NK cell population divided into IgM responders (Y) and non-responders (N) Day 14 following vaccination (C). Log2 fold change of CD56bright and CD56dim NK cell populations divided into early (84–168 days) high and low responders (IgG, IgG1, IgG2, IgG3, IgG4, and IgA) (D).

References

    1. WHO. HIV/AIDS. WHO; (2017). Available from:
    1. Shattock RJ, Warren M, McCormack S, Hankins CA. Turning the tide against HIV. Science (2011) 333(6038):42–3.10.1126/science.1206399
    1. de Bruyn G. Cofactors that may influence vaccine responses. Curr Opin HIV AIDS (2010) 5(5):404–8.10.1097/COH.0b013e32833d1fca
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med (2012) 366(14):1275–86.10.1056/NEJMoa1113425
    1. Cotugno N, De Armas L, Pallikkuth S, Rossi P, Palma P, Pahwa S. Paediatric HIV infection in the ‘omics era: defining transcriptional signatures of viral control and vaccine responses. J Virus Erad (2015) 1:153–8.
    1. Day TA, Kublin JG. Lessons learned from HIV vaccine clinical efficacy trials. Curr HIV Res (2013) 11(6):441–9.10.2174/1570162X113116660051
    1. Katinger D, Jeffs S, Altmann F, Cope A, McKay P, Almond N, et al. CN54gp140: product characteristics, preclinical and clinical use – recombinant glycoprotein for HIV immunization. Retrovirology (2012) 9(2):351.10.1186/1742-4690-9-S2-P351
    1. Curran RM, Donnelly L, Morrow RJ, Fraser C, Andrews G, Cranage M, et al. Vaginal delivery of the recombinant HIV-1 clade-C trimeric gp140 envelope protein CN54gp140 within novel rheologically structured vehicles elicits specific immune responses. Vaccine (2009) 27(48):6791–8.10.1016/j.vaccine.2009.08.088
    1. Pattani A, McKay PF, Garland MJ, Curran RM, Migalska K, Cassidy CM, et al. Microneedle mediated intradermal delivery of adjuvanted recombinant HIV-1 CN54gp140 effectively primes mucosal boost inoculations. J Control Release (2012) 162(3):529–37.10.1016/j.jconrel.2012.07.039
    1. McKay PF, Cope AV, Mann JFS, Joseph S, Esteban M, Tatoud R, et al. Glucopyranosyl lipid A adjuvant significantly enhances HIV specific T and B cell responses elicited by a DNA-MVA-protein vaccine regimen. PLoS One (2014) 9(1):e84707.10.1371/journal.pone.0084707
    1. O’Neill LAJ, Bowie AG. The family of five: TIR-domain-containing adaptors in toll-like receptor signalling. Nat Rev Immunol (2007) 7(5):353–64.10.1038/nri2079
    1. Arias MA, Roey GAV, Tregoning JS, Moutaftsi M, Coler RN, Windish HP, et al. Glucopyranosyl lipid adjuvant (GLA), a synthetic TLR4 agonist, promotes potent systemic and mucosal responses to intranasal immunization with HIVgp140. PLoS One (2012) 7(7):e41144.10.1371/journal.pone.0041144
    1. Lambert SL, Yang C-F, Liu Z, Sweetwood R, Zhao J, Cheng L, et al. Molecular and cellular response profiles induced by the TLR4 agonist-based adjuvant glucopyranosyl lipid A. PLoS One (2012) 7(12):e51618.10.1371/journal.pone.0051618
    1. Li S, Rouphael N, Duraisingham S, Romero-Steiner S, Presnell S, Davis C, et al. Molecular signatures of antibody responses derived from a systems biological study of 5 human vaccines. Nat Immunol (2014) 15(2):195–204.10.1038/ni.2789
    1. Olafsdottir T, Lindqvist M, Harandi AM. Molecular signatures of vaccine adjuvants. Vaccine (2015) 33(40):5302–7.10.1016/j.vaccine.2015.04.099
    1. Olafsdottir TA, Lindqvist M, Nookaew I, Andersen P, Maertzdorf J, Persson J, et al. Comparative systems analyses reveal molecular signatures of clinically tested vaccine adjuvants. Sci Rep (2016) 6:39097.10.1038/srep39097
    1. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol (2009) 10(1):116–25.10.1038/ni.1688
    1. Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, et al. Systems analysis of immunity to influenza vaccination across multiple years and in diverse populations reveals shared molecular signatures. Immunity (2015) 43(6):1186–98.10.1016/j.immuni.2015.11.012
    1. Nakaya HI, Clutterbuck E, Kazmin D, Wang L, Cortese M, Bosinger SE, et al. Systems biology of immunity to MF59-adjuvanted versus nonadjuvanted trivalent seasonal influenza vaccines in early childhood. Proc Natl Acad Sci U S A (2016) 113(7):1853–8.10.1073/pnas.1519690113
    1. Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol (2011) 12(8):786–95.10.1038/ni.2067
    1. Joseph S, Quinn K, Greenwood A, Cope AV, McKay PF, Hayes PJ, et al. A comparative phase I study of combination, homologous subtype-C DNA, MVA, and Env gp140 protein/adjuvant HIV vaccines in two immunization regimes. Front Immunol (2017) 8:149.10.3389/fimmu.2017.00149
    1. Cosgrove CA, Lacey CJ, Cope AV, Bartolf A, Morris G, Yan C, et al. Comparative immunogenicity of HIV-1 gp140 vaccine delivered by parenteral, and mucosal routes in female volunteers; MUCOVAC2, a randomized two centre study. PLoS One (2016) 11(5):e0152038.10.1371/journal.pone.0152038
    1. Kratochvil S, McKay PF, Kopycinski JT, Bishop C, Hayes PJ, Muir L, et al. A phase 1 human immunodeficiency virus vaccine trial for cross-profiling the kinetics of serum and mucosal antibody responses to CN54gp140 modulated by two homologous prime-boost vaccine regimens. Front Immunol (2017) 8:595.10.3389/fimmu.2017.00595/full
    1. R Development Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; (2012). Available from:
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A (2005) 102(43):15545–50.10.1073/pnas.0506580102
    1. Broad Institute. GSEA. Gene Set Enrichment Analysis. v5.0; Available from:
    1. Nakanishi K, Ida M, Suzuki H, Kitano C, Yamamoto A, Mori N, et al. Molecular characterization of a transport vesicle protein neurensin-2, a homologue of neurensin-1, expressed in neural cells. Brain Res (2006) 1081(1):1–8.10.1016/j.brainres.2006.01.085
    1. Chamberland JP, Antonow LT, Santos MD, Ritter B. NECAP2 controls clathrin coat recruitment to early endosomes for fast endocytic recycling. J Cell Sci (2016) 129(13):2625–37.10.1242/jcs.173708
    1. Hou J, Wang S, Jia M, Li D, Liu Y, Li Z, et al. A systems vaccinology approach reveals temporal transcriptomic changes of immune responses to the yellow fever 17D vaccine. J Immunol (2017) 199(4):1476–89.10.4049/jimmunol.1700083
    1. Serena M, Parolini F, Biswas P, Sironi F, Miranda AB, Zoratti E, et al. HIV-1 Env associates with HLA-C free-chains at the cell membrane modulating viral infectivity. Sci Rep (2017) 7:40037.10.1038/srep40037
    1. Vivier E, Anfossi N. Inhibitory NK-cell receptors on T cells: witness of the past, actors of the future. Nat Rev Immunol (2004) 4(3):190–8.10.1038/nri1306
    1. Kaiser BK, Pizarro JC, Kerns J, Strong RK. Structural basis for NKG2A/CD94 recognition of HLA-E. Proc Natl Acad Sci U S A (2008) 105(18):6696–701.10.1073/pnas.0802736105
    1. Kazmin D, Nakaya HI, Lee EK, Johnson MJ, van der Most R, van den Berg RA, et al. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. Proc Natl Acad Sci U S A (2017) 114(9):2425–30.10.1073/pnas.1621489114
    1. Rydyznski C, Daniels KA, Karmele EP, Brooks TR, Mahl SE, Moran MT, et al. Generation of cellular immune memory and B-cell immunity is impaired by natural killer cells. Nat Commun (2015) 6:6375.10.1038/ncomms7375
    1. Cauwelaert ND, Desbien AL, Hudson TE, Pine SO, Reed SG, Coler RN, et al. The TLR4 agonist vaccine adjuvant, GLA-SE, requires canonical and atypical mechanisms of action for T H 1 induction. PLoS One (2016) 11(1):e0146372.10.1371/journal.pone.0146372
    1. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol (2008) 9(5):503–10.10.1038/ni1582
    1. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood (2010) 115(11):2167–76.10.1182/blood-2009-08-238469
    1. Dou Y, Fu B, Sun R, Li W, Hu W, Tian Z, et al. Influenza vaccine induces intracellular immune memory of human NK cells. PLoS One (2015) 10(3):e0121258.10.1371/journal.pone.0121258
    1. Scott-Algara D, Mancini-Bourgine M, Fontaine H, Pol S, Michel M-L. Changes to the natural killer cell repertoire after therapeutic hepatitis B DNA vaccination. PLoS One (2010) 5(1):e8761.10.1371/journal.pone.0008761
    1. Rydyznski CE, Waggoner SN. Boosting vaccine efficacy the natural (killer) way. Trends Immunol (2015) 36(9):536–46.10.1016/j.it.2015.07.004

Source: PubMed

3
Abonner