Modulation of p38 MAPK signaling enhances dendritic cell activation of human CD4+ Th17 responses to ovarian tumor antigen

Martin J Cannon, Hannah E Goyne, Pamela J B Stone, Laura J Macdonald, Lindsey E James, Everardo Cobos, Maurizio Chiriva-Internati, Martin J Cannon, Hannah E Goyne, Pamela J B Stone, Laura J Macdonald, Lindsey E James, Everardo Cobos, Maurizio Chiriva-Internati

Abstract

The recent finding that Th17 infiltration of ovarian tumors positively predicts patient outcomes suggests that Th17 responses play a protective role in ovarian tumor immunity. This observation has led to the question of whether Th17 cells could be induced or expanded to therapeutic advantage by tumor vaccination. In this study, we show that treatment of ovarian tumor antigen-loaded, cytokine-matured human dendritic cells (DC) with a combination of IL-15 and a p38 MAP kinase inhibitor offers potent synergy in antagonism of CD4(+) Treg induction and redirection toward CD4(+) Th17 responses that correlate with strong CD8(+) cytotoxic T lymphocyte (CTL) activation. Ovarian tumor antigen-specific CD4(+) T cells secrete high levels of IL-17 and show reduced expression of CTLA-4, PD-1, and Foxp3 following activation with IL-15/p38 inhibitor-treated DC. We further show that modulation of p38 MAPK signaling in DC is associated with reduced expression of B7-H1 (PD-L1), loss of indoleamine 2,3-dioxygenase activity, and increased phosphorylation of ERK 1/2 MAPK. These observations may allow the development of innovative DC vaccination strategies to boost Th17 immunity in ovarian cancer patients.

Conflict of interest statement

Conflict of interest Dr. Cannon is founder of DCV Technologies Inc, a biotechnology company dedicated to the clinical development of dendritic cell vaccines for the treatment of cancer. Dr. Chiriva- Internati is founder of Kiromic Inc, a biotechnology company that seeks to develop therapeutic cancer vaccines. The other authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
Functional analysis of hepsin 48–84 tumor antigen-specific CD4+ T cell lines derived by stimulation with cytokine-matured DC, IL-15-treated cytokine-matured DC, p38i-treated cytokine-matured DC or IL-15/p38i-treated, cytokine-matured DC. a CD4+ T cell expression of TNFα, IFNγ and Foxp3 following stimulation with antigen-loaded autologous LCL. CD4+ T cells cocultured with control LCL did not express TNFα or IFNγ (not shown). Representative results from two healthy subjects are shown (from a total of ten experiments). b CD8+ CTL responses against antigen-loaded autologous LCL (filled circle) or control LCL (open circle). c CD4+ T cell IL-17 expression following stimulation with antigen-loaded autologous LCL. Statistically significant differences were observed between IL-17 expression by the IL-15/p38i DC-derived CD4+ T cells and all other CD4+ T cell groups (*P ≤ 0.008). For all groups, CD4+ T cells cocultured with control LCL expressed <15 pg/ml IL-17 (not shown). LCL alone did not express IL-17
Fig. 2
Fig. 2
Costimulatory molecule expression by mature DC following treatment with IL-15 and/or p38 inhibition. a Expression of CD80, CD86, and CD83. b Expression of B7-H1 (PD-L1) and ICOS-L
Fig. 3
Fig. 3
CTLA-4 and PD-1 expression by CD4+ T cell lines derived by DC stimulation following treatment of DC with IL-15 and/or p38 inhibition
Fig. 4
Fig. 4
Phosphorylation of p38, ERK, and NF-κB following treatment of DC with IL-15 and/or p38 inhibition
Fig. 5
Fig. 5
IDOactivity inDC. Kynurenine production by cytokine-matured DC, IL-15-treated cytokine-matured DC, p38i-treated cytokine-matured DC or IL-15/p38i-treated, cytokine-matured DC, incubated with or without tryptophan (the IDO substrate). Statistically significant differences in IDO activity were observed between p38i-treated and untreated DC, and also between cytokine-matured DC and IL-15-treated cytokine-matured DC (*P ≤ 0.001 for all comparisons)
Fig. 6
Fig. 6
Summary of phenotypic and functional changes in mature DC and CD4+ T cells following DC treatment with IL-15 and/or p38 inhibition

References

    1. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–949.
    1. Wolf D, Wolf AM, Rumpold H, Fiegl H, Zeimet AG, Muller-Holzner E, Deibl M, Gastl G, Gunsilius E, Marth C. The expression of the regulatory T cell-specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer. Clin Cancer Res. 2005;11:8326–8331.
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA. 2005;102:18538–18543.
    1. Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, Huang E, Finlayson E, Simeone D, Welling TH, Chang A, Coukos G, Liu R, Zou W. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 2009;114:1141–1149.
    1. Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol. 2007;8:345–350.
    1. Kryczek I, Wei S, Zou L, Altuwaijri S, Szeliga W, Kolls J, Chang A, Zou W. Cutting edge: Th17 and regulatory T cell dynamics and the regulation by IL-2 in the tumor microenvironment. J Immunol. 2007;178:6730–6733.
    1. Munn DH. Th17 cells in ovarian cancer. Blood. 2009;114:1134–1135.
    1. Cannon MJ, Goyne H, Stone PJB, Chiriva-Internati M. Dendritic cell vaccination against ovarian cancer—tipping the Treg/Th17 balance to therapeutic advantage. Exp Opin Biol Ther. 2011;11:1–5.
    1. Jackson AM, Mulcahy LA, Zhu XW, O’Donnell D, Patel PM. Tumour-mediated disruption of dendritic cell function: inhibiting the MEK1/2-p44/42 axis restores IL-12 production and Th1-generation. Int J Cancer. 2008;123:623–632.
    1. Xie J, Qian J, Yang J, Wang S, Freeman ME, 3rd, Yi Q. Critical roles of Raf/MEK/ERK and PI3 K/AKT signaling and inactivation of p38 MAP kinase in the differentiation and survival of monocyte-derived immature dendritic cells. Exp Hematol. 2005;33:564–572.
    1. Wang S, Hong S, Yang J, Qian J, Zhang X, Shpall E, Kwak LW, Yi Q. Optimizing immunotherapy in multiple myeloma: restoring the function of patients’ monocyte-derived dendritic cells by inhibiting p38 or activating MEK/ERK MAPK and neutralizing interleukin-6 in progenitor cells. Blood. 2006;108:4071–4077.
    1. Zhao F, Falk C, Osen W, Kato M, Schadendorf D, Umansky V. Activation of p38 mitogen-activated protein kinase drives dendritic cells to become tolerogenic in ret transgenic mice spontaneously developing melanoma. Clin Cancer Res. 2009;15:4382–4390.
    1. Jarnicki AG, Conroy H, Brereton C, Donnelly G, Toomey D, Walsh K, Sweeney C, Leavy O, Fletcher J, Lavelle EC, Dunne P, Mills KH. Attenuating regulatory T cell induction by TLR agonists through inhibition of p38 MAPK signaling in dendritic cells enhances their efficacy as vaccine adjuvants and cancer immunotherapeutics. J Immunol. 2008;180:3797–3806.
    1. Brereton C, Sutton CE, Lalor SJ, Lavelle EC, Mills KH. Inhibition of ERK MAPK suppresses IL-23-and IL-1-driven IL-17 production and attenuates autoimmune disease. J Immunol. 2009;183:1715–1723.
    1. Tanimoto H, Yan Y, Clarke J, Korourian S, Shigemasa K, Parmley TH, Parham GP, O’Brien TJ. Hepsin, a cell surface serine protease identified in hepatoma cells, is overexpressed on ovarian cancer. Cancer Res. 1997;57:2884–2887.
    1. Southwood S, Sidney J, Kondo A, del Guercio M-F, Appella E, Hoffman S, Kubo RT, Chesnut RW, Grey HM, Sette A. Several common HLA-DR types share largely overlapping peptide binding repertoires. J Immunol. 1998;160:3363–3373.
    1. Bondurant KL, Crew MD, Santin AD, O’Brien TJ, Cannon MJ. Definition of an immunodominant region within the ovarian tumor antigen stratum corneum chymotryptic enzyme. Clin Cancer Res. 2005;11:3446–3454.
    1. Braun D, Longman RS, Albert ML. A two-step induction of indoleamine 2,3 dioxygenase (IDO) activity during dendritic-cell maturation. Blood. 2005;106:2375–2381.
    1. Sallusto F, Schaerli P, Loetscher P, Schaniel C, Lenig D, Mackay CR, Qin S, Lanzavecchia A. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur J Immunol. 1998;28:2760–2769.
    1. Randolph GJ, Sanchez-Schmitz G, Angeli V. Factors and signals that govern the migration of dendritic cells via lymphatics: recent advances. Springer Semin Immunol. 2005;26:273–287.
    1. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, Nomura T, Sakagushi S. CTLA-4 control over Foxp3+ regulatory T cell function. Science. 2008;322:271–275.
    1. Zheng Y, Manzotti CN, Burke F, Dussably L, Qureshi O, Walker LSK, Sansom DM. Acquisition of suppressive function by activated human CD4+CD25− T cells is associated with the expression of CTLA-4 not Foxp3. J Immunol. 2008;181:1683–1691.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Ann Rev Immunol. 2008;26:677–704.
    1. Zou W, Machelon V, Coulomb-L’Hermin A, Borvak J, Nome F, Isaeva T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, Pustilnik T, Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid dendritic cells. Nat Med. 2001;7:1339–1346.
    1. Wei S, Kryczek I, Edwards RP, Zou L, Szeliga W, Banerjee M, Cost M, Cheng P, Chang A, Redman B, Herberman RB, Zou W. Interleukin-2 administration alters the CD4+Foxp3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res. 2007;67:7487–7494.
    1. Baeuerle PA. The inducible transcription activator NF-κB—regulation by distinct protein subunits. Biochim Biophys Acta. 1991;1072:63–80.
    1. Verma IM, Stevenson JK, Schwarz EM, van Antwerp D, Miyamoto S. Rel/NF-κB/IκB family: intimate tales of association and dissociation. Genes Dev. 1995;9:2723–2735.
    1. Chung DJ, Rossi M, Romano E, Ghith J, Yuan J, Munn DH, Young JW. Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells. Blood. 2009;114:555–563.
    1. Sharma MD, Hou DY, Liu Y, Koni PA, Metz R, Chandler P, Mellor AL, He Y, Munn DH. Indoleamine 2,3-dioxygenase controls conversion of Foxp3 + Tregs to TH17-like cells in tumor-draining lymph nodes. Blood. 2009;113:6102–6111.
    1. Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, Munn DH, Mellor AL. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol. 2009;183:2475–2483.
    1. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, Honjo T, Fujii S. Programmed cell death 1 ligand 1 and tumorinfiltrating CD8 + T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA. 2007;104:3360–3365.
    1. Kryczek I, Wei S, Zhu G, Myers L, Mottram P, Cheng P, Chen L, Coukos G, Zou W. Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma. Cancer Res. 2007;67:8900–8905.
    1. Okamoto A, Nikaido T, Ochiai K, Takakura S, Saito M, Aoki Y, Ishii N, Yanaihara N, Yamada K, Takikawa O, Kawaguchi R, Isonishi S, Tanaka T, Urashima M. Indoleamine 2,3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clin Cancer Res. 2005;11:6030–6039.
    1. Inaba T, Ino K, Kajiyama H, Yamamoto E, Shibata K, Nawa A, Nagasaka T, Akimoto H, Takikawa O, Kikkawa F. Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma. Gynecol Oncol. 2009;115:185–192.
    1. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213.
    1. Iida T, Iwahashi M, Katsuda K, Nakamori M, Nakamura M, Naka T, Ojima T, Ueda K, Hayata K, Nakamura Y, Yamaue H. Tumor-infiltrating CD4+ Th17 cells produce IL-17 in tumor microenvironment and promote tumor progression in human gastric cancer. Oncol Rep. 2011;25:1271–1277.
    1. Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, Lin Z, Zhu B. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun. 2011;407:348–354.
    1. Murugaiyan G, Saha B. Protumor vs antitumor functions of IL-17. J Immunol. 2009;183:4169–4175.
    1. Kryczek I, Wu K, Zhao E, Wei S, Vatan L, Szeliga W, Huang E, Greenson J, Chang A, Rolinski J, Radwan P, Fang J, Wang G, Zou W. IL-17+ regulatory T cells in the microenvironments of chronic inflammation and cancer. J Immunol. 2011;186:4388–4395.
    1. Middleton GW, Annels NE, Pandha HS. Are we ready to start studies of Th17 cell manipulation as a therapy for cancer? Cancer Immunol Immunother. 2012;61:1–7.
    1. Derhovanessian E, Adams V, Hähnel K, Groeger A, Pandha H, Ward S, Pawelec G. Pretreatment frequency of circulating IL-17+ CD4+ T-cells, but not Tregs, correlates with clinical response to whole-cell vaccination in prostate cancer patients. Int J Cancer. 2009;125:1372–1379.
    1. Sfanos KS, Bruno TC, Maris CH, Xu L, Thoburn CJ, DeMarzo AM, Meeker AK, Isaacs WB, Drake CG. Phenotypic analysis of prostate-infiltrating lymphocytes reveals TH17 and Treg skewing. Clin Cancer Res. 2008;14:3254–3261.
    1. Gnerlich JL, Mitchem JB, Weir JS, Sankpal NV, Kashiwagi H, Belt BA, Porembka MR, Herndon JM, Eberlein TJ, Goedegebuure P, Linehan DC. Induction of Th17 cells in the tumor microenvironment improves survival in a murine model of pancreatic cancer. J Immunol. 2010;185:4063–4071.
    1. Kryczek I, Wei S, Szeliga W, Vatan L, Zou W. Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood. 2009;114:357–359.
    1. Muranski P, Boni A, Antony PA, Cassard L, Irvine KR, Kaiser A, Paulos CM, Palmer DC, Touloukian CE, Ptak K, Gattinoni L, Wrzesinski C, Hinrichs CS, Kerstann KW, Feigenbaum L, Chan C–C, Restifo NP. Tumor-specific Th17-polarized cells eradicate large established melanoma. Blood. 2008;112:362–373.
    1. Martin-Orozco N, Muranski P, Chung Y, Yang XO, Yamazaki T, Lu S, Hwu P, Restifo NP, Overwijk WW, Dong C. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity. 2009;31:787–798.
    1. Kryczek I, Zhao E, Liu Y, Vatan L, Szeliga W, Moyer J, Klimczak A, Lange A, Zou W. Human Th17 cells are long-lived effector memory cells. Sci Transl Med. 2011;3 104ra100.
    1. Wang L, Pino-Lagos K, de Vries VC, Guleria I, Sayegh MH, Noelle RJ. Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells. Proc Natl Acad Sci USA. 2008;105:9331–9336.
    1. Paulos CM, Carpenito C, Plesa G, Suhoski MM, Varela-Rohena A, Golovina TN, Carroll RG, Riley JL, June CH. The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells. Sci Transl Med. 2010;2 55ra78.
    1. Bouguermouh S, Foritn G, Baba N, Rubio M, Sarfati M. CD28 co-stimulation down regulates Th17 development. PLoS ONE. 2009;4:e5087.

Source: PubMed

3
Subscribe