Co-delivery of PLGA encapsulated invariant NKT cell agonist with antigenic protein induce strong T cell-mediated antitumor immune responses

Yusuf Dölen, Martin Kreutz, Uzi Gileadi, Jurjen Tel, Angela Vasaturo, Eric A W van Dinther, Maaike A van Hout-Kuijer, Vincenzo Cerundolo, Carl G Figdor, Yusuf Dölen, Martin Kreutz, Uzi Gileadi, Jurjen Tel, Angela Vasaturo, Eric A W van Dinther, Maaike A van Hout-Kuijer, Vincenzo Cerundolo, Carl G Figdor

Abstract

Antitumor immunity can be enhanced by the coordinated release and delivery of antigens and immune-stimulating agents to antigen-presenting cells via biodegradable vaccine carriers. So far, encapsulation of TLR ligands and tumor-associated antigens augmented cytotoxic T cell (CTLs) responses. Here, we compared the efficacy of the invariant NKT (iNKT) cell agonist α-galactosylceramide (α-GalCer) and TLR ligands (R848 and poly I:C) as an adjuvant for the full length ovalbumin (OVA) in PLGA nanoparticles. We observed that OVA+α-GalCer nanoparticles (NP) are superior over OVA+TLR-L NP in generating and stimulating antigen-specific cytotoxic T lymphocytes without the need for CD4+ T cell help. Not only a 4-fold higher induction of antigen-specific T cells was observed, but also a more profound IFN-γ secretion was obtained by the addition α-GalCer. Surprisingly, we observed that mixtures of OVA containing NP with α-GalCer were ineffective, demonstrating that co-encapsulation of both α-GalCer and antigen within the same nanoparticle is essential for the observed T cell responses. Moreover, a single immunization with OVA+α-GalCer NP provided substantial protection from tumor formation and even delayed the growth of already established tumors, which coincided with a prominent and enhanced antigen-specific CD8+ T cell infiltration. The provided evidence on the advantage of antigen and α-GalCer coencapsulation should be considered in the design of future nanoparticle vaccines for therapeutic purposes.

Keywords: CTL response; PLGA; melanoma; nanoparticle; α-GalCer.

Figures

Figure 1.
Figure 1.
In vivo antigen-specific T cell responses induced by nanoparticles. (A) Mice were vaccinated with indicated nanoparticle combinations and transferred with SIINFEKL peptide-loaded syngeneic target cells and irrelevant peptide-loaded control cells. Primary antigen-specific in vivo killing responses (left) and SIINFEKL H-2Kb tetramer-positive cell percentages (right) obtained by OVA and α-GalCer or TLR-L injections were demonstrated. Results of three independent experiments were pooled. n> 6 for each group (B) OT-I T-cell proliferation induced by nanoparticle administration to wild type naive mice, n=5 (left). Comparison of OVA+ α-GalCer nanoparticle induced proliferation of OT-I T cells transferred to either CD1d KO or wild-type mice, n=30 (right). Parenthesis indicates encapsulation, ↔ indicates injection of NP(OVA) and free α-GalCer to different veins consequently. 0,66ug OVA, 1ng α-GalCer, 143ng R848 and 70ng poly I:C was introduced within NPs. Mean values were shown with standard error, ns: no significance, * p < .05, ** p < .01, *** p < .001
Figure 2.
Figure 2.
Cytokine responses obtained by nanoparticle encapsulated α-GalCer and TLR-ligands. (A) IFN-g levels of 48h splenocyte cultures with indicated NPs or free α-GalCer in solution. (B) IL-6 levels of 48h splenocyte cultures with indicated NPs. Splenocytes were isolated from either naive wild-type BL/6 or CD1d KO BL/6 mice (A-B, n>4). (C) IFN-g serum levels measured 24 h after iv. injections of 1ng α-GalCer either in nanoparticles or free. Vaccinations were repeated 2 times on days 7 and 14, empty particles were used as a control, n≥3. Mean values were shown with standard error. (D) IL-2 response curves of DN32D3 NKT cell hybridoma cultured 24h with α-GalCer loaded BMDCs. α-GalCer was serially diluted in DMSO, RMPI medium or PBS 0.05% tween 20 (Vehicle) and cultured with BMDCs at a range of 25ng-0.01ng/mL. One representative graph of three experiments was shown.
Figure 3.
Figure 3.
Antitumor effects of (OVA+α-GalCer) nanoparticle immunization. (A) Mice were immunized on day 0 with indicated vaccines and B16.OVA cells were subcutaneously inoculated on day 7(dotted line). Survival results of two independent experiments were shown. Censored data points were shown with symbols. PBS, n= 7; NP(OVA+TLR-L), n=8 and NP(OVA+ α-GalCer), n=8. (B) Vaccinations were performed on day 0 and tumors were inoculated on day 7 as in A. NP(OVA+ α-GalCer) group was re-challenged with tumor cells on day 50 and 78 (dotted line). NP(OVA+TLR-L) n=6, NP(OVA+ α-GalCer) n=8 . (C) Mice were first subcutaneously inoculated with B16.OVA cells and vaccinations were started individually when each tumor reached ≈30mm3 and repeated three times weekly. NP(OVA), n=7, Mitv=26mm3; NP(OVA+TLR-L), n=13, Mitv=33mm3 ; NP(OVA+α-GalCer), n=19, Mitv=33mm3. Data points of mice culled due to early necrosis were censored. Censored data points were shown with symbols. Mitv: Mean initial tumor volume. *** P < .001.(D) Serum samples were collected 24 h after each vaccination of tumor-bearing mice in (C) and IFN-g levels were determined by ELISA. n=7. Mean results were shown with standard error.
Figure 4.
Figure 4.
Intratumoral T cell infiltration after therapeutic vaccinations. Similar sized tumors were isolated 8 d after first vaccination. Frozen tumor tissues were screened for CTL infiltration by CD8+ (red) and DAPI (Blue) staining by Vectra automated quantitative imaging system. NP(OVA), n=2; NP(OVA+TLR-L), n=4; NP(OVA+α-GalCer), n=2.

References

    1. Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5:140; PMID:24744762;
    1. Andersen BM, Ohlfest JR. Increasing the efficacy of tumor cell vaccines by enhancing cross priming. Cancer Lett 2012; 325:155-64; PMID:22809568;
    1. Brennan PJ, Brigl M, Brenner MB. Invariant natural killer T cells: an innate activation scheme linked to diverse effector functions. Nat Rev Immunol 2013; 13:101-17; PMID:23334244;
    1. Carreno LJ, Kharkwal SS, Porcelli SA. Optimizing NKT cell ligands as vaccine adjuvants. Immunotherapy 2014; 6:309-20; PMID:24762075;
    1. Terabe M, Berzofsky JA. The immunoregulatory role of type I and type II NKT cells in cancer and other diseases. Cancer Immunol Immunother 2014; 63:199-213; PMID:24384834;
    1. Fujii S, Liu K, Smith C, Bonito AJ, Steinman RM. The linkage of innate to adaptive immunity via maturing dendritic cells in vivo requires CD40 ligation in addition to antigen presentation and CD80/86 costimulation. J Exp Med 2004; 199:1607-18; PMID:15197224;
    1. Hermans IF, Silk JD, Gileadi U, Salio M, Mathew B, Ritter G, Schmidt R, Harris AL, Old L, Cerundolo V. NKT cells enhance CD4+ and CD8+ T cell responses to soluble antigen in vivo through direct interaction with dendritic cells. J Immunol 2003; 171:5140-7; PMID:14607913;
    1. Matsuda JL, Mallevaey T, Scott-Browne J, Gapin L. CD1d-restricted iNKT cells, the 'Swiss-Army knife' of the immune system. Curr Opin Immunol 2008; 20:358-68; PMID:18501573;
    1. Fujii S, Shimizu K, Smith C, Bonifaz L, Steinman RM. Activation of natural killer T cells by α-galactosylceramide rapidly induces the full maturation of dendritic cells in vivo and thereby acts as an adjuvant for combined CD4+ and CD8+ T cell immunity to a coadministered protein. J Exp Med 2003; 198:267-79; PMID:12874260;
    1. Gonzalez-Aseguinolaza G, Van Kaer L, Bergmann CC, Wilson JM, Schmieg J, Kronenberg M, Nakayama T, Taniguchi M, Koezuka Y, Tsuji M. Natural killer T cell ligand α-galactosylceramide enhances protective immunity induced by malaria vaccines. J Exp Med 2002; 195:617-24; PMID:11877484;
    1. Silk JD, Hermans IF, Gileadi U, Chong TW, Shepherd D, Salio M, Mathew B, Schmidt RR, Lunt SJ, Williams KJ et al.. Utilizing the adjuvant properties of CD1d-dependent NK T cells in T cell-mediated immunotherapy. J Clin Invest 2004; 114:1800-11; PMID:15599405;
    1. Hayakawa Y, Rovero S, Forni G, Smyth MJ. Alpha-galactosylceramide (KRN7000) suppression of chemical- and oncogene-dependent carcinogenesis. Proc Natl Acad Sci U S A 2003; 100:9464-9; PMID:12867593;
    1. Shimizu K, Goto A, Fukui M, Taniguchi M, Fujii S. Tumor cells loaded with α-galactosylceramide induce innate NKT and NK cell-dependent resistance to tumor implantation in mice. J Immunol 2007; 178:2853-61; PMID:17312129;
    1. Schmieg J, Yang G, Franck RW, Tsuji M. Superior protection against malaria and melanoma metastases by a C-glycoside analogue of the natural killer T cell ligand α-Galactosylceramide. J Exp Med 2003; 198:1631-41; PMID:14657217;
    1. Smyth MJ, Crowe NY, Pellicci DG, Kyparissoudis K, Kelly JM, Takeda K, Yagita H, Godfrey DI. Sequential production of interferon-gamma by NK1.1(+) T cells and natural killer cells is essential for the antimetastatic effect of α-galactosylceramide. Blood 2002; 99:1259-66; PMID:11830474;
    1. Salio M, Silk JD, Jones EY, Cerundolo V. Biology of CD1- and MR1-restricted T cells. Annu Rev Immunol 2014; 32:323-66; PMID:24499274;
    1. Liu Y, Goff RD, Zhou D, Mattner J, Sullivan BA, Khurana A, Cantu C 3rd, Ravkov EV, Ibegbu CC, Altman JD et al.. A modified α-galactosyl ceramide for staining and stimulating natural killer T cells. J Immunol Methods 2006; 312:34-9; PMID:16647712;
    1. Giaccone G, Punt CJ, Ando Y, Ruijter R, Nishi N, Peters M, von Blomberg BM, Scheper RJ, van der Vliet HJ et al.. A phase I study of the natural killer T-cell ligand α-galactosylceramide (KRN7000) in patients with solid tumors. Clin Cancer Res 2002; 8:3702-9; PMID:12473579
    1. Ishikawa A, Motohashi S, Ishikawa E, Fuchida H, Higashino K, Otsuji M, Iizasa T, Nakayama T, Taniguchi M, Fujisawa T. A phase I study of α-galactosylceramide (KRN7000)-pulsed dendritic cells in patients with advanced and recurrent non-small cell lung cancer. Clin Cancer Res 2005; 11:1910-7; PMID:15756017;
    1. Chang DH, Osman K, Connolly J, Kukreja A, Krasovsky J, Pack M, Hutchinson A, Geller M, Liu N, Annable R et al.. Sustained expansion of NKT cells and antigen-specific T cells after injection of α-galactosyl-ceramide loaded mature dendritic cells in cancer patients. J Exp Med 2005; 201:1503-17; PMID:15867097;
    1. Fujii S, Shimizu K, Kronenberg M, Steinman RM. Prolonged IFN-gamma-producing NKT response induced with α-galactosylceramide-loaded DCs. Nat Immunol 2002; 3:867-74; PMID:12154358;
    1. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Preat V. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release 2012; 161:505-22; PMID:22353619;
    1. Bershteyn A, Hanson MC, Crespo MP, Moon JJ, Li AV, Suh H, Irvine DJ. Robust IgG responses to nanograms of antigen using a biomimetic lipid-coated particle vaccine. J Control Release 2012; 157:354-65; PMID:21820024;
    1. Barral P, Polzella P, Bruckbauer A, van Rooijen N, Besra GS, Cerundolo V, Batista FD. CD169(+) macrophages present lipid antigens to mediate early activation of iNKT cells in lymph nodes. Nat Immunol 2010; 11:303-12; PMID:20228797;
    1. Barral P, Sanchez-Nino MD, van Rooijen N, Cerundolo V, Batista FD. The location of splenic NKT cells favours their rapid activation by blood-borne antigen. EMBO J 2012; 31:2378-90; PMID:22505026;
    1. Thapa P, Zhang G, Xia C, Gelbard A, Overwijk WW, Liu C, Hwu P, Chang DZ, Courtney A, Sastry JK et al.. Nanoparticle formulated α-galactosylceramide activates NKT cells without inducing anergy. Vaccine 2009; 27:3484-8; PMID:19200815;
    1. Macho Fernandez E, Chang J, Fontaine J, Bialecki E, Rodriguez F, Werkmeister E, Krieger V, Ehret C, Heurtault B, Fournel S et al.. Activation of invariant Natural Killer T lymphocytes in response to the α-galactosylceramide analogue KRN7000 encapsulated in PLGA-based nanoparticles and microparticles. Int J Pharm 2012; 423:45-54; PMID:21575695;
    1. Macho-Fernandez E, Cruz LJ, Ghinnagow R, Fontaine J, Bialecki E, Frisch B, Trottein F, Faveeuw C. Targeted Delivery of α-Galactosylceramide to CD8alpha+ dendritic cells optimizes type I NKT cell-based antitumor responses. J Immunol 2014; 193:961-9; PMID:24913977;
    1. Iyoda T, Ushida M, Kimura Y, Minamino K, Hayuka A, Yokohata S, Ehara H, Inaba K. Invariant NKT cell anergy is induced by a strong TCR-mediated signal plus co-stimulation. Int Immunol 2010; 22:905-13; PMID:21118907;
    1. Diwan M, Elamanchili P, Lane H, Gainer A, Samuel J. Biodegradable nanoparticle mediated antigen delivery to human cord blood derived dendritic cells for induction of primary T cell responses. J Drug Target 2003; 11:495-507; PMID:15203918;
    1. Zhang Z, Tongchusak S, Mizukami Y, Kang YJ, Ioji T, Touma M, Reinhold B, Keskin DB, Reinherz EL, Sasada T. Induction of anti-tumor cytotoxic T cell responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials 2011; 32:3666-78; PMID:21345488;
    1. Hamdy S, Molavi O, Ma Z, Haddadi A, Alshamsan A, Gobti Z, Elhasi S, Samuel J, Lavasanifar A. Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+ T cell-mediated anti-tumor immunity. Vaccine 2008; 26:5046-57; PMID:18680779;
    1. Cruz LJ, Tacken PJ, Fokkink R, Joosten B, Stuart MC, Albericio F, Torensma R, Figdor CG. Targeted PLGA nano- but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. J Control Release 2010; 144:118-26; PMID:20156497;
    1. Tacken PJ, Zeelenberg IS, Cruz LJ, van Hout-Kuijer MA, van de Glind G, Fokkink RG, Lambeck AJ, Figdor CG. Targeted delivery of TLR ligands to human and mouse dendritic cells strongly enhances adjuvanticity. Blood 2011; 118:6836-44; PMID:21967977;
    1. McKee SJ, Young VL, Clow F, Hayman CM, Baird MA, Hermans IF, Young SL, Ward VK. Virus-like particles and α-galactosylceramide form a self-adjuvanting composite particle that elicits anti-tumor responses. J Control Release 2012; 159:338-45; PMID:22386518;
    1. Gehrmann U, Hiltbrunner S, Georgoudaki AM, Karlsson MC, Naslund TI, Gabrielsson S. Synergistic induction of adaptive antitumor immunity by codelivery of antigen with α-galactosylceramide on exosomes. Cancer Res 2013; 73:3865-76; PMID:23658368;
    1. Schlosser E, Mueller M, Fischer S, Basta S, Busch DH, Gander B, Groettrup M. TLR ligands and antigen need to be coencapsulated into the same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses. Vaccine 2008; 26:1626-37; PMID:18295941;
    1. Nair-Gupta P, Baccarini A, Tung N, Seyffer F, Florey O, Huang Y, Banerjee M, Overholtzer M, Roche PA, Tampé R. TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation. Cell 2014; 158:506-21; PMID:25083866;
    1. Semmling V, Lukacs-Kornek V, Thaiss CA, Quast T, Hochheiser K, Panzer U, Rossjohn J, Perlmutter P, Cao J, Godfrey DI et al.. Alternative cross-priming through CCL17-CCR4-mediated attraction of CTLs toward NKT cell-licensed DCs. Nat Immunol 2010; 11:313-20; PMID:20190758;
    1. Lee YR, Lee YH, Im SA, Kim K, Lee CK. Formulation and characterization of antigen-loaded PLGA Nanoparticles for efficient cross-priming of the antigen. Immune Netw 2011; 11:163-8; PMID:21860609;
    1. Fischer S, Schlosser E, Mueller M, Csaba N, Merkle HP, Groettrup M, Gander B. Concomitant delivery of a CTL-restricted peptide antigen and CpG ODN by PLGA microparticles induces cellular immune response. J Drug Target 2009; 17:652-61; PMID:19622019;
    1. Silva AL, Rosalia RA, Sazak A, Carstens MG, Ossendorp F, Oostendorp J, Jiskoot W. Optimization of encapsulation of a synthetic long peptide in PLGA nanoparticles: low-burst release is crucial for efficient CD8(+) T cell activation. Eur J Pharm Biopharm 2013; 83:338-45; PMID:23201055;

Source: PubMed

3
Prenumerera