Transpresentation of interleukin-15 by IL-15/IL-15Rα mRNA-engineered human dendritic cells boosts antitumoral natural killer cell activity

Johan Van den Bergh, Yannick Willemen, Eva Lion, Heleen Van Acker, Hans De Reu, Sébastien Anguille, Herman Goossens, Zwi Berneman, Viggo Van Tendeloo, Evelien Smits, Johan Van den Bergh, Yannick Willemen, Eva Lion, Heleen Van Acker, Hans De Reu, Sébastien Anguille, Herman Goossens, Zwi Berneman, Viggo Van Tendeloo, Evelien Smits

Abstract

In cancer immunotherapy, the use of dendritic cell (DC)-based vaccination strategies can improve overall survival, but until now durable clinical responses remain scarce. To date, DC vaccines are designed primarily to induce effective T-cell responses, ignoring the antitumor activity potential of natural killer (NK) cells. Aiming to further improve current DC vaccination outcome, we engineered monocyte-derived DC to produce interleukin (IL)-15 and/or IL-15 receptor alpha (IL-15Rα) using mRNA electroporation. The addition of IL-15Rα to the protocol, enabling IL-15 transpresentation to neighboring NK cells, resulted in significantly better NK-cell activation compared to IL-15 alone. Next to upregulation of NK-cell membrane activation markers, IL-15 transpresentation resulted in increased NK-cell secretion of IFN-γ, granzyme B and perforin. Moreover, IL-15-transpresenting DC/NK cell cocultures from both healthy donors and acute myeloid leukemia (AML) patients in remission showed markedly enhanced cytotoxic activity against NK cell sensitive and resistant tumor cells. Blocking IL-15 transpresentation abrogated NK cell-mediated cytotoxicity against tumor cells, pointing to a pivotal role of IL-15 transpresentation by IL-15Rα to exert its NK cell-activating effects. In conclusion, we report an attractive approach to improve antitumoral NK-cell activity in DC-based vaccine strategies through the use of IL-15/IL-15Rα mRNA-engineered designer DC.

Keywords: IL-15 receptor α; Immune response; Immunity; Immunology and Microbiology Section; dendritic cells; interleukin-15 transpresentation; mRNA elektroporation; natural killer cells.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

Figures

Figure 1. Interleukin-15 membrane expression and secretion…
Figure 1. Interleukin-15 membrane expression and secretion of IL-15 mRNA electroporated DC
A. Membrane-bound IL-15 expression was determined by flow cytometric staining of mock EP DC (dashed black line), IL-15 EP DC (grey triangles) and IL-15/IL-15Rα EP DC (black squares) 2h, 4h, 8h, 24h, 48h and 72h after electroporation. Expression levels (MFI) were transformed to relative levels compared to those of the corresponding mock EP DC, which were set to one. Data are shown as mean (± SEM) for 3 independent donors. B. IL-15 secretion was quantified using an ELISA on the same EP conditions (mock EP DC, IL-15 EP DC and IL-15/IL-15Rα EP DC) and the same time points after electroporation (2h, 4h, 8h, 24h, 48h and 72h) as shown in figure 1A. Data are shown as mean (± SEM) for 6 independent donors. Statistical comparison was performed between IL-15 EP DC and IL-15/IL-15Rα EP DC at each time point. ns, not significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001, two-way ANOVA with Bonferroni posthoc test. Abbreviations: EP; electroporation, MFI; mean fluorescence intensity, SEM; standard error of the mean.
Figure 2. Phenotypic activation profile of DC-…
Figure 2. Phenotypic activation profile of DC- stimulated NK cells
NK cells were cultured alone (white bars) or in coculture with mock EP DC (light grey bars), IL-15 EP DC (dark grey bars) or IL-15/IL-15Rα EP DC (black bars) in a NK/DC-ratio of 5:1. After 48h, NK-cell phenotype was determined using flow cytometry. Data, shown as dMFI (as compared to isotype controls), are depicted as mean (± SEM) for 3 independent donors. As a positive control, NK cells were stimulated with recombinant human IL-15 (1 ng/mL) for 48h before measuring phenotypic NK-cell activation (striped bars). ns, not significant; *, p < 0.05; **, p < 0.01; ***,p < 0.001, repeated measures one-way ANOVA with Bonferroni posthoc test. Abbreviations: EP; electroporation, dMFI; delta mean fluorescence intensity, SEM; standard error of the mean.
Figure 3. Granzyme B, perforin and IFN-γ…
Figure 3. Granzyme B, perforin and IFN-γ production after NK/DC/tumor-cell coculture
Granzyme B A. and perforin B. secretion are shown for 48h NK/Daudi (ratio 5:1, white bars) and NK/DC/Daudi (ratio 5:1:1, mock EP DC light grey bars, IL-15 EP DC dark grey bars, IL-15/IL-15Rα EP DC black bars) cocultures. Data are shown as mean (± SEM) for 5 (granzyme B) and 8 (perforin) independent donors, respectively. C. Intracellular IFN-γ, measured after 24h NK/DC/Daudi coculture (ratio 5:1:1), is shown for 6 independent donors. As a positive control, NK cells were stimulated with IFN-α (1000U/mL) for 48h and K562 cells for 4h (striped bar) before measuring intracellular IFN-γ. ns, not significant; *,p < 0.05; **,p < 0.01; ***, p < 0.001, repeated measures one-way ANOVA with Bonferroni posthoc test. Abbreviation: SEM; standard error of the mean.
Figure 4. Cytotoxicity of NK/DC cocultures against…
Figure 4. Cytotoxicity of NK/DC cocultures against Daudi cells
The mean killing percentage (± SEM) of Daudi cells is shown for NK/Daudi (ratio 5:1, white bars), DC/Daudi (ratio 1:1, mock EP DC light grey bars, IL-15 EP DC dark grey bars, IL-15/IL-15Rα EP DC black bars) and NK/DC/Daudi (ratio 5:1:1, mock EP DC light grey bars, IL-15 EP DC dark grey bars, IL-15/IL-15Rα EP DC black bars) cocultures based on a 4h flow cytometric cytotoxicity assay following 44h NK-cell and/or DC cocultures. A. In some conditions, a transwell insert (pore size of 4 μM) was added before starting NK/DC cocultures. These data are shown as mean (± SEM) for 5-9 independent donors. B. 1h prior to the addition of NK cells, anti-IL-15 neutralizing IgG or corresponding IgG isotype control antibody (100 μg/4x105 DC) was added to DC. These data are shown as mean (± SEM) for 4-8 independent donors. **, p < 0.01; ***, p < 0.001, one-way ANOVA with Bonferroni posthoc test. Abbreviation: SEM; standard error of the mean.
Figure 5. Validation of the cytotoxic profile…
Figure 5. Validation of the cytotoxic profile of DC-activated NK cells against tumor cells for AML patients in remission
The killing percentage of A. Daudi, B. K562 and C. THP-1 is shown for NK/DC/tumor-cell (ratio 5:1:1) cocultures of three different AML patients in remission based on a 4h flow cytometric cytotoxicity assay following 44h NK/DC cocultures. AML 1 and AML 2 are already a few years in remission, while patient AML 3 was tested shortly after consolidation. *, p < 0.05, one-way ANOVA with Bonferroni posthoc test.

References

    1. Finn OO, Banchereau J. Cancer vaccines: the state of the art. Semin Immunol. 2010;22:103–104.
    1. Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, Gostick E, Vermeulen K, Pieters K, Nijs G, Stein B, Smits EL, Schroyens WA, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A. 2010;107:13824–13829.
    1. Palucka K, Ueno H, Banchereau J. Recent developments in cancer vaccines. J Immunol. 2011;186:1325–1331.
    1. Anguille S, Lion E, Tel J, de Vries IJ, Coudere K, Fromm PD, Van Tendeloo VF, Smits EL, Berneman ZN. Interleukin-15-induced CD56(+) myeloid dendritic cells combine potent tumor antigen presentation with direct tumoricidal potential. PLoS One. 2012;7:e51851.
    1. Frankenberger B, Schendel DJ. Third generation dendritic cell vaccines for tumor immunotherapy. Eur J Cell Biol. 2012;91:53–58.
    1. Subklewe M, Geiger C, Lichtenegger FS, Javorovic M, Kvalheim G, Schendel DJ, Bigalke I. New generation dendritic cell vaccine for immunotherapy of acute myeloid leukemia. Cancer Immunol Immunother. 2014;63:1093–1103.
    1. Van Elssen CH, Oth T, Germeraad WT, Bos GM, Vanderlocht J. Natural killer cells: the secret weapon in dendritic cell vaccination strategies. Clin Cancer Res. 2014;20:1095–1103.
    1. Pampena MB, Levy EM. Natural killer cells as helper cells in dendritic cell cancer vaccines. Front Immunol. 2015;6:13.
    1. Bouwer AL, Saunderson SC, Caldwell FJ, Damani TT, Pelham SJ, Dunn AC, Jack RW, Stoitzner P, McLellan AD. NK cells are required for dendritic cell-based immunotherapy at the time of tumor challenge. J Immunol. 2014;192:2514–2521.
    1. Lion E, Smits EL, Berneman ZN, Van Tendeloo VF. NK cells: key to success of DC-based cancer vaccines? Oncologist. 2012;17:1256–1270.
    1. McLellan AD. A critical role for natural killer cells in dendritic cell-based anticancer immunotherapy. Oncoimmunology. 2014;3:e28582.
    1. Croce M, Orengo AM, Azzarone B, Ferrini S. Immunotherapeutic applications of IL-15. Immunotherapy. 2012;4:957–969.
    1. Jakobisiak M, Golab J, Lasek W. Interleukin 15 as a promising candidate for tumor immunotherapy. Cytokine Growth Factor Rev. 2011;22:99–108.
    1. Steel JC, Waldmann TA, Morris JC. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol Sci. 2012;33:35–41.
    1. Burkett PR, Koka R, Chien M, Chai S, Boone DL, Ma A. Coordinate expression and trans presentation of interleukin (IL)-15Ralpha and IL-15 supports natural killer cell and memory CD8+ T cell homeostasis. J Exp Med. 2004;200:825–834.
    1. Kobayashi H, Dubois S, Sato N, Sabzevari H, Sakai Y, Waldmann TA, Tagaya Y. Role of trans-cellular IL-15 presentation in the activation of NK cell-mediated killing, which leads to enhanced tumor immunosurveillance. Blood. 2005;105:721–727.
    1. Schluns KS, Stoklasek T, Lefrancois L. The roles of interleukin-15 receptor alpha: trans-presentation, receptor component, or both? Int J Biochem Cell Biol. 2005;37:1567–1571.
    1. Lodolce JP, Burkett PR, Koka RM, Boone DL, Ma A. Regulation of lymphoid homeostasis by interleukin-15. Cytokine Growth Factor Rev. 2002;13:429–439.
    1. Rubinstein MP, Kovar M, Purton JF, Cho JH, Boyman O, Surh CD, Sprent J. Converting IL-15 to a superagonist by binding to soluble IL-15R{alpha} Proc Natl Acad Sci U S A. 2006;103:9166–9171.
    1. Stoklasek TA, Schluns KS, Lefrancois L. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J Immunol. 2006;177:6072–6080.
    1. Van den Bergh JM, Van Tendeloo VF, Smits EL. Interleukin-15: new kid on the block for antitumor combination therapy. Cytokine Growth Factor Rev. 2015;26:15–24.
    1. Cheever MA. Twelve immunotherapy drugs that could cure cancers. Immunol Rev. 2008;222:357–368.
    1. Berger C, Berger M, Hackman RC, Gough M, Elliott C, Jensen MC, Riddell SR. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood. 2009;114:2417–2426.
    1. Waldmann TA, Lugli E, Roederer M, Perera LP, Smedley JV, Macallister RP, Goldman CK, Bryant BR, Decker JM, Fleisher TA, Lane HC, Sneller MC, Kurlander RJ, et al. Safety (toxicity), pharmacokinetics, immunogenicity, and impact on elements of the normal immune system of recombinant human IL-15 in rhesus macaques. Blood. 2011;117:4787–4795.
    1. Waldmann T CK. First-in-Humans Study of New Immunotherapy Agent. NCI Cancer Bulletin. 2012
    1. Van Tendeloo VF, Ponsaerts P, Berneman ZN. mRNA-based gene transfer as a tool for gene and cell therapy. Curr Opin Mol Ther. 2007;9:423–431.
    1. Bergamaschi C, Rosati M, Jalah R, Valentin A, Kulkarni V, Alicea C, Zhang GM, Patel V, Felber BK, Pavlakis GN. Intracellular interaction of interleukin-15 with its receptor alpha during production leads to mutual stabilization and increased bioactivity. J Biol Chem. 2008;283:4189–4199.
    1. Dubois S, Patel HJ, Zhang M, Waldmann TA, Muller JR. Preassociation of IL-15 with IL-15R alpha-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J Immunol. 2008;180:2099–2106.
    1. Steel JC, Ramlogan CA, Yu P, Sakai Y, Forni G, Waldmann TA, Morris JC. Interleukin-15 and its receptor augment dendritic cell vaccination against the neu oncogene through the induction of antibodies partially independent of CD4 help. Cancer Res. 2010;70:1072–1081.
    1. Duitman EH, Orinska Z, Bulanova E, Paus R, Bulfone-Paus S. How a cytokine is chaperoned through the secretory pathway by complexing with its own receptor: lessons from interleukin-15 (IL-15)/IL-15 receptor alpha. Mol Cell Biol. 2008;28:4851–4861.
    1. Vanderlocht J, Van Elssen CH, Senden-Gijsbers BL, Meek B, Cloosen S, Libon C, Bos GM, Germeraad WT. Increased tumor-specific CD8+ T cell induction by dendritic cells matured with a clinical grade TLR-agonist in combination with IFN-gamma. Int J Immunopathol Pharmacol. 2010;23:35–50.
    1. Lucas M, Schachterle W, Oberle K, Aichele P, Diefenbach A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity. 2007;26:503–517.
    1. Castillo EF, Stonier SW, Frasca L, Schluns KS. Dendritic cells support the in vivo development and maintenance of NK cells via IL-15 trans-presentation. J Immunol. 2009;183:4948–4956.
    1. Vujanovic L, Szymkowski DE, Alber S, Watkins SC, Vujanovic NL, Butterfield LH. Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15. Blood. 2010;116:575–583.
    1. Boudreau JE, Stephenson KB, Wang F, Ashkar AA, Mossman KL, Lenz LL, Rosenthal KL, Bramson JL, Lichty BD, Wan Y. IL-15 and type I interferon are required for activation of tumoricidal NK cells by virus-infected dendritic cells. Cancer Res. 2011;71:2497–2506.
    1. Anguille S, Lion E, Van den Bergh J, Van Acker HH, Willemen Y, Smits EL, Van Tendeloo VF, Berneman ZN. Interleukin-15 dendritic cells as vaccine candidates for cancer immunotherapy. Hum Vaccin Immunother. 2013;9:1956–1961.
    1. Zhang Y, Tian S, Liu Z, Zhang J, Zhang M, Bosenberg MW, Kedl RM, Waldmann TA, Storkus WJ, Falo LD, Jr, You Z. Dendritic cell-derived interleukin-15 is crucial for therapeutic cancer vaccine potency. Oncoimmunology. 2014;3:e959321.
    1. Anguille S, Van Acker HH, Van den Bergh J, Willemen Y, Goossens H, Van Tendeloo VF, Smits EL, Berneman ZN, Lion E. Interleukin-15 Dendritic Cells Harness NK Cell Cytotoxic Effector Function in a Contact- and IL-15-Dependent Manner. PLoS One. 2015;10:e0123340.
    1. Mortier E, Woo T, Advincula R, Gozalo S, Ma A. IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J Exp Med. 2008;205:1213–1225.
    1. Elpek KG, Rubinstein MP, Bellemare-Pelletier A, Goldrath AW, Turley SJ. Mature natural killer cells with phenotypic and functional alterations accumulate upon sustained stimulation with IL-15/IL-15Ralpha complexes. Proc Natl Acad Sci U S A. 2010;107:21647–21652.
    1. Anguille S, Lion E, Smits E, Berneman ZN, van Tendeloo VF. Dendritic cell vaccine therapy for acute myeloid leukemia: questions and answers. Hum Vaccin. 2011;7:579–584.
    1. Lion E, Anguille S, Berneman ZN, Smits EL, Van Tendeloo VF. Poly(I:C) enhances the susceptibility of leukemic cells to NK cell cytotoxicity and phagocytosis by DC. PLoS One. 2011;6:e20952.
    1. Anguille S, Willemen Y, Lion E, Smits EL, Berneman ZN. Dendritic cell vaccination in acute myeloid leukemia. Cytotherapy. 2012;14:647–656.
    1. Lichtenegger FS, Krupka C, Kohnke T, Subklewe M. Immunotherapy for Acute Myeloid Leukemia. Semin Hematol. 2015;52:207–214.
    1. Childs RW, Berg M. Bringing natural killer cells to the clinic: ex vivo manipulation. Hematology Am Soc Hematol Educ Program. 2013;2013:234–246.
    1. Guo Y, Luan L, Rabacal W, Bohannon JK, Fensterheim BA, Hernandez A, Sherwood ER. IL-15 Superagonist-Mediated Immunotoxicity: Role of NK Cells and IFN-gamma. J Immunol. 2015;195:2353.
    1. Lu J, Giuntoli RL, 2nd, Omiya R, Kobayashi H, Kennedy R, Celis E. Interleukin 15 promotes antigen-independent in vitro expansion and long-term survival of antitumor cytotoxic T lymphocytes. Clin Cancer Res. 2002;8:3877–3884.
    1. Kutzler MA, Robinson TM, Chattergoon MA, Choo DK, Choo AY, Choe PY, Ramanathan MP, Parkinson R, Kudchodkar S, Tamura Y, Sidhu M, Roopchand V, Kim JJ, et al. Coimmunization with an optimized IL-15 plasmid results in enhanced function and longevity of CD8 T cells that are partially independent of CD4 T cell help. J Immunol. 2005;175:112–123.
    1. Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, Tureci O, Sahin U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–4017.
    1. Willemen Y, Van den Bergh JM, Lion E, Anguille S, Roelandts VA, Van Acker HH, Heynderickx SD, Stein BM, Peeters M, Figdor CG, Van Tendeloo VF, de Vries IJ, Adema GJ, et al. Engineering monocyte-derived dendritic cells to secrete interferon-alpha enhances their ability to promote adaptive and innate anti-tumor immune effector functions. Cancer Immunol Immunother. 2015;64:831–842.
    1. Van den Bergh JM, Guerti K, Willemen Y, Lion E, Cools N, Goossens H, Vorsters A, Van Tendeloo VF, Anguille S, Van Damme P, Smits EL. HPV vaccine stimulates cytotoxic activity of killer dendritic cells and natural killer cells against HPV-positive tumour cells. J Cell Mol Med. 2014;18:1372–1380.

Source: PubMed

3
購読する