Montanide, Poly I:C and nanoparticle based vaccines promote differential suppressor and effector cell expansion: a study of induction of CD8 T cells to a minimal Plasmodium berghei epitope

Kirsty L Wilson, Sue D Xiang, Magdalena Plebanski, Kirsty L Wilson, Sue D Xiang, Magdalena Plebanski

Abstract

The development of practical and flexible vaccines to target liver stage malaria parasites would benefit from an ability to induce high levels of CD8 T cells to minimal peptide epitopes. Herein we compare different adjuvant and carrier systems in a murine model for induction of interferon gamma (IFN-γ) producing CD8 T cells to the minimal immuno-dominant peptide epitope from the circumsporozoite protein (CSP) of Plasmodium berghei, pb9 (SYIPSAEKI, referred to as KI). Two pro-inflammatory adjuvants, Montanide and Poly I:C, and a non-classical, non-inflammatory nanoparticle based carrier (polystyrene nanoparticles, PSNPs), were compared side-by-side for their ability to induce potentially protective CD8 T cell responses after two immunizations. KI in Montanide (Montanide + KI) or covalently conjugated to PSNPs (PSNPs-KI) induced such high responses, whereas adjuvanting with Poly I:C or PSNPs without conjugation was ineffective. This result was consistent with an observed induction of an immunosuppressed environment by Poly I:C in the draining lymph node (dLN) 48 h post injection, which was reflected by increased frequencies of myeloid derived suppressor cells (MDSCs) and a proportion of inflammation reactive regulatory T cells (Treg) expressing the tumor necrosis factor receptor 2 (TNFR2), as well as decreased dendritic cell (DC) maturation. The other inflammatory adjuvant, Montanide, also promoted proportional increases in the TNFR2(+) Treg subpopulation, but not MDSCs, in the dLN. By contrast, injection with non-inflammatory PSNPs did not cause these changes. Induction of high CD8 T cell responses, using minimal peptide epitopes, can be achieved by non-inflammatory carrier nanoparticles, which in contrast to some conventional inflammatory adjuvants, do not expand either MDSCs or inflammation reactive Tregs at the site of priming.

Keywords: CD8 peptide; MDSC; Treg; adjuvant; malaria; nanoparticle.

Figures

FIGURE 1
FIGURE 1
Size distribution for PSNPs-KI formulations. SYIPSAEKI peptides were covalently conjugated to PSNPs, and the final sizes were measured by dynamic light scattering instruments (Zetasizer).
FIGURE 2
FIGURE 2
Antigen specific CD8 T cell responses induced by SYIPSAEKI peptide vaccines in combination with PSNPs. Mice (BALB/c) were immunized with SYIPSAEKI peptide mixed with or conjugated to PSNPs intradermally at the base of tail. 14 days after the last immunization, spleen cells were collected and assessed for IFN-γ production by ELISpot assay. (A) KI peptides conjugated to, but not mixed with, PSNPs induced high levels of KI specific CD8 T cell responses after two immunizations (2 weeks apart). Data presented as mean ± SD of SFU/million cells from each group (n = 3 mice/group). (B) Immunogenicity of PSNPs-KI formulation after one immunization (n = 4 mice per group). Data presented as mean ± SD of SFU/million cells (pooled for each group) from the triplicated wells in ELISpot assay. Statistical analysis was performed via ANOVA, ***p ≤ 0.001.
FIGURE 3
FIGURE 3
Induction of SYIPSAEKI specific CD8 T cells by different adjuvants. BALB/c mice were immunized twice, intradermally, 2 weeks apart, with SYIPSAEKI peptides incorporated with respective adjuvants. 14 days after the last immunization, spleen cells were collected and assessed for IFN-γ production by ELISpot assay. Data presented as mean ± SD of SFU/million cells from each group (n = 3 mice/group). Statistical analysis was performed via ANOVA, ***p ≤ 0.001.
FIGURE 4
FIGURE 4
Dendritic cell activation in dLNs after injection with PSNPs, Montanide, and Poly I:C. Mice (BALB/c) were injected once intradermally at the base of tail with the different adjuvants alone. 48 h after injection, mice were sacrificed, local (inguinal) dLNs were harvested and the levels of CD11c+ DCs and various activation markers were assessed by flow cytometry. (A) gating strategy; (B) frequency of GR-1-CD11c+ cells; (C) Mean fluorescent intensity (MFI) of CD80 and CD86 on GR-1-CD11c+ cells; (D) MFI of CD40 and MHCII on GR-1-CD11c+ cells. Data presented as mean ± SD of MFI for each group of treatment (n = 3 mice/group). Statistical analysis was performed via t-tests, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.
FIGURE 5
FIGURE 5
Differential expression of GR-1+ MDSCs and DCs in the local dLN. Mice (BALB/c) were injected once intradermally at the base of the tail with the different adjuvants alone. 48 h after injection, mice were sacrificed, local (inguinal) dLNs were harvested and levels of GR-1+ MDSCs and DCs, and their ratios, were assessed by flow cytometry. (A) gating strategy; (B) ratio of GR-1+ MDSCs: CD11c+ cells; (C) ratio of moMDSCs: CD11c+ cells; (D) ratio of gMDSCs: CD11c+ cells. Data presented as mean ± SD of ratio for each group of treatment (n = 3 mice/group). Statistical analysis was performed via t-tests, *p ≤ 0.05, **p ≤ 0.01.
FIGURE 6
FIGURE 6
Differential expression of CD4+ Treg and effector T cells in the local dLN. Mice (BALB/c) were injected once intradermally at the base of tail with the different adjuvants alone. 48 h after injection, mice were sacrificed, local (inguinal) dLNs were harvested and the levels of CD4+ Treg and effector T cells, and their ratios, were assessed by flow cytometry. (A) gating strategy; (B) ratio of FoxP3+CD25+: FoxP3-CD25- cells; (C) ratio of FoxP3+CD25+TNFR2+: FoxP3+CD25+TNFR2- cells. Data presented as mean ± SD of ratio for each group of treatment (n = 3 mice/group). Statistical analysis was performed via t-tests, *p ≤ 0.05.

References

    1. Arama C., Troye-Blomberg M. (2014). The path of malaria vaccine development: challenges and perspectives. J. Intern. Med. 275 456–466 10.1111/joim.12223
    1. Aucouturier J., Dupuis L., Deville S., Ascarateil S., Ganne V. (2002). Montanide ISA 720 and 51: a new generation of water in oil emulsions as adjuvants for human vaccines. Expert Rev. Vaccines 1 111–118 10.1586/14760584.1.1.111
    1. Bonhoure F., Gaucheron J. (2006). Montanide ISA 51 VG as adjuvant for human vaccines. J. Immunother. 29 647–648.
    1. Chen X., Oppenheim J. J. (2011). Contrasting effects of TNF and anti-TNF on the activation of effector T cells and regulatory T cells in autoimmunity. FEBS Lett. 585 3611–3618 10.1016/j.febslet.2011.04.025
    1. Clarke S. R. (2000). The critical role of CD40/CD40L in the CD4-dependent generation of CD8+ T cell immunity. J. Leukoc. Biol. 67 607–614.
    1. Doolan D. L., Martinez-Alier N. (2006). Immune response to pre-erythrocytic stages of malaria parasites. Curr. Mol. Med. 6 169–185 10.2174/156652406776055249
    1. Fifis T., Gamvrellis A., Crimeen-Irwin B., Pietersz G. A., Li J., Mottram P. L., et al. (2004a). Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J. Immunol. 173 3148–3154 10.4049/jimmunol.173.5.3148
    1. Fifis T., Mottram P., Bogdanoska V., Hanley J., Plebanski M. (2004b). Short peptide sequences containing MHC class I and/or class II epitopes linked to nano-beads induce strong immunity and inhibition of growth of antigen-specific tumour challenge in mice. Vaccine 23 258–266 10.1016/j.vaccine.2004.05.022
    1. Gamvrellis A., Walsh K., Tatarczuch L., Smooker P., Plebanski M., Scheerlinck J. P. (2013). Phenotypic analysis of ovine antigen presenting cells loaded with nanoparticles migrating from the site of vaccination. Methods 60 257–263 10.1016/j.ymeth.2013.02.008
    1. Govindaraj C., Tan P., Walker P., Wei A., Spencer A., Plebanski M. (2014). Reducing TNF receptor 2+ regulatory T cells via the combined action of azacitidine and the HDAC inhibitor, panobinostat for clinical benefit in acute myeloid leukemia patients. Clin. Cancer Res. 20 724–735 10.1158/1078-0432.CCR-13-1576
    1. Henrickson S. E., Mempel T. R., Mazo I. B., Liu B., Artyomov M. N., Zheng H., et al. (2008). In vivo imaging of T cell priming. Sci. Signal. 1 pt2. 10.1126/stke.112pt2
    1. Karlson Tde L., Kong Y. Y., Hardy C. L., Xiang S. D., Plebanski M. (2013). The signalling imprints of nanoparticle uptake by bone marrow derived dendritic cells. Methods 60 275–283 10.1016/j.ymeth.2013.02.009
    1. Kong Y. Y., Fuchsberger M., Xiang S. D., Apostolopoulos V., Plebanski M. (2013). Myeloid derived suppressor cells and their role in diseases. Curr. Med. Chem. 20 1437–1444 10.2174/0929867311320110006
    1. Krzych U., Zarling S., Pichugin A. (2014). Memory T cells maintain protracted protection against malaria. Immunol. Lett. 161 189–195 10.1016/j.imlet.2014.03.011
    1. Longhi M. P., Trumpfheller C., Idoyaga J., Caskey M., Matos I., Kluger C., et al. (2009). Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J. Exp. Med. 206 1589–1602 10.1084/jem.20090247
    1. Maroof A., Beattie L., Kirby A., Coles M., Kaye P. M. (2009). Dendritic cells matured by inflammation induce CD86-dependent priming of naive CD8+ T cells in the absence of their cognate peptide antigen. J. Immunol. 183 7095–7103 10.4049/jimmunol.0901330
    1. Mbow M. L., De Gregorio E., Valiante N. M., Rappuoli R. (2010). New adjuvants for human vaccines. Curr. Opin. Immunol. 22 411–416 10.1016/j.coi.2010.04.004
    1. Mottram P. L., Leong D., Crimeen-Irwin B., Gloster S., Xiang S. D., Meanger J., et al. (2007). Type 1 and 2 immunity following vaccination is influenced by nanoparticle size: formulation of a model vaccine for respiratory syncytial virus. Mol. Pharm. 4 73–84 10.1021/mp060096p
    1. Nordly P., Rose F., Christensen D., Nielsen H. M., Andersen P., Agger E. M., et al. (2011). Immunity by formulation design: induction of high CD8+ T-cell responses by poly(I:C) incorporated into the CAF01 adjuvant via a double emulsion method. J. Control. Release 150 307–317 10.1016/j.jconrel.2010.11.021
    1. Ohkusu-Tsukada K., Ohta S., Kawakami Y., Toda M. (2011). Adjuvant effects of formalin-inactivated HSV through activation of dendritic cells and inactivation of myeloid-derived suppressor cells in cancer immunotherapy. Int. J. Cancer 128 119–131 10.1002/ijc.25319
    1. Perret R., Sierro S. R., Botelho N. K., Corgnac S., Donda A., Romero P. (2013). Adjuvants that improve the ratio of antigen-specific effector to regulatory T cells enhance tumor immunity. Cancer Res. 73 6597–6608 10.1158/0008-5472.CAN-13-0875
    1. Plebanski M., Gilbert S. C., Schneider J., Hannan C. M., Layton G., Blanchard T., et al. (1998). Protection from Plasmodium berghei infection by priming and boosting T cells to a single class I-restricted epitope with recombinant carriers suitable for human use. Eur. J. Immunol. 28 4345–4355 10.1002/(SICI)1521-4141(199812)28:12<4345::AID-IMMU4345>;2-P
    1. Plebanski M., Lopez E., Proudfoot O., Cooke B. M., Itzstein M., Coppel R. L. (2006). Economic and practical challenges to the formulation of vaccines against endemic infectious diseases such as malaria. Methods 40 77–85 10.1016/j.ymeth.2006.05.021
    1. Postels D. G., Birbeck G. L. (2013). Cerebral malaria. Handb. Clin. Neurol. 114 91–102 10.1016/B978-0-444-53490-3.00006-6
    1. Schneider J., Gilbert S. C., Hannan C. M., Degano P., Prieur E., Sheu E. G., et al. (1999). Induction of CD8+ T cells using heterologous prime-boost immunisation strategies. Immunol. Rev. 170 29–38 10.1111/j.1600-065X.1999.tb01326.x
    1. Shen J., Chen X., Wang Z., Zhang G., Chen W. (2014). Downregulation of CD40 expression contributes to the accumulation of myeloid-derived suppressor cells in gastric tumors. Oncol. Lett. 8 775–780 10.3892/ol.2014.2174
    1. Steinman R. M., Hawiger D., Liu K., Bonifaz L., Bonnyay D., Mahnke K., et al. (2003). Dendritic cell function in vivo during the steady state: a role in peripheral tolerance. Ann. N. Y. Acad. Sci. 987 15–25 10.1111/j.1749-6632.2003.tb06029.x
    1. Trumpfheller C., Caskey M., Nchinda G., Longhi M. P., Mizenina O., Huang Y., et al. (2008). The microbial mimic poly IC induces durable and protective CD4+ T cell immunity together with a dendritic cell targeted vaccine. Proc. Natl. Acad. Sci. U.S.A. 105 2574–2579 10.1073/pnas.0711976105
    1. Xiang S. D., Kalkanidis M., Pietersz G. A., Mottram P. L., Crimeen-Irwin B., Ardipradja K., et al. (2006). Methods for nano-particle based vaccine formulation and evaluation of their immunogenicity. Methods 40 20–29 10.1016/j.ymeth.2006.05.018
    1. Xiang S. D., Wilson K., Day S., Fuchsberger M., Plebanski M. (2013). Methods of effective conjugation of antigens to nanoparticles as non-inflammatory vaccine carriers. Methods 60 232–241 10.1016/j.ymeth.2013.03.036

Source: PubMed

3
Sottoscrivi