Enhanced immunity in a mouse model of malignant glioma is mediated by a therapeutic ketogenic diet

Danielle M Lussier, Eric C Woolf, John L Johnson, Kenneth S Brooks, Joseph N Blattman, Adrienne C Scheck, Danielle M Lussier, Eric C Woolf, John L Johnson, Kenneth S Brooks, Joseph N Blattman, Adrienne C Scheck

Abstract

Background: Glioblastoma multiforme is a highly aggressive brain tumor with a poor prognosis, and advances in treatment have led to only marginal increases in overall survival. We and others have shown previously that the therapeutic ketogenic diet (KD) prolongs survival in mouse models of glioma, explained by both direct tumor growth inhibition and suppression of pro-inflammatory microenvironment conditions. The aim of this study is to assess the effects of the KD on the glioma reactive immune response.

Methods: The GL261-Luc2 intracranial mouse model of glioma was used to investigate the effects of the KD on the tumor-specific immune response. Tumor-infiltrating CD8+ T cells, CD4+ T cells and natural killer (NK) cells were analyzed by flow cytometry. The expression of immune inhibitory receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1) on CD8+ T cells were also analyzed by flow cytometry. Analysis of intracellular cytokine production was used to determine production of IFN, IL-2 and IFN- in tumor-infiltrating CD8+ T and natural killer (NK) cells and IL-10 production by T regulatory cells.

Results: We demonstrate that mice fed the KD had increased tumor-reactive innate and adaptive immune responses, including increased cytokine production and cytolysis via tumor-reactive CD8+ T cells. Additionally, we saw that mice maintained on the KD had increased CD4 infiltration, while T regulatory cell numbers stayed consistent. Lastly, mice fed the KD had a significant reduction in immune inhibitory receptor expression as well as decreased inhibitory ligand expression on glioma cells.

Conclusions: The KD may work in part as an immune adjuvant, boosting tumor-reactive immune responses in the microenvironment by alleviating immune suppression. This evidence suggests that the KD increases tumor-reactive immune responses, and may have implications in combinational treatment approaches.

Keywords: CTLA-4; Glioblastoma; Glioma; Immune inhibitory checkpoints; Immunology; Immunosuppression; Ketogenic diet; Metabolism; Microenvironment; PD-1.

Figures

Fig. 1
Fig. 1
Enhanced survival with the ketogenic diet is mediated in part by CD8 T cells. Kaplan-Meier survival curves for ketogenic diet (KD) versus standard diet (SD) (a), SD versus SD + CD8 depletion (b), SD versus KD + CD8 depletion (c), KD versus KD + CD8 depletion (d). Bioluminescent tumor signals plotted as in vivo photon count versus days post-implantation (e). N = 12 for immune competent mice; N = 5 for CD8 depleted mice; Log-rank (Mantel-Cox) test; p-values indicated on graphs
Fig. 2
Fig. 2
CD4+ T cell infiltration increases in mice fed the KD, without increases in Treg cell numbers. Flow cytometry analysis was performed to assess the cell types infiltrating tumors from mice fed both SD and KD. CD8 T cells (a), CD4 T cells (b) and CD4 + FoxP3+ T regulatory cells (c) were assessed. The ratio of CD8 T cells to T regulatory cells (d) and CD4 to T regulatory cells (e) were determined. The percent of infiltrating NKp46 + CD3- natural killer cells (f) were also assessed. N = 5; student’s two-tailed t-test; ***p < 0.001; ****p < 0.0001
Fig. 3
Fig. 3
The ketogenic diet reduces expression of immune inhibitory receptors and ligands expressed in glioma tumors. Expression of the immune inhibitory receptors, PD-1 (a) and CTLA-4 (b) on infiltrating CD8 T cells isolated from tumors from mice fed each diet were assessed. Expression of the immune inhibitory ligands, CD86 (c) and PD-L1 (d), on GL261-Luc2 tumor tissue was also assessed. N = 5; student’s two tailed t-test; *p < 0.05; **p < 0.01; ****p < 0.0001
Fig. 4
Fig. 4
The ketogenic diet significantly enhances tumor-reactive CD8+ T cell and NK cell activity. Tumor-infiltrating lymphocytes (TILs) isolated from gliomas from mice fed KD versus SD were cultured alone (white bar) or in the presence of GL261-Luc2 tumor cells (black bar) to access activity. Analysis of IFNγ, TNF and IL-2 production in tumor-infiltrating CD8+ T cells was performed (a). Cytotoxic capability of CD8+ T cells isolated from tumors was assessed following exposure to GL261-Luc2 cells (b). IL-10-production in CD4 + FoxP3+ T regulatory cells was also assessed in response to stimulation with GL261-Luc2 cells (c). IFNγ and TNF production in the infiltrating NKp46 + CD3- natural killer cells isolated from tumors were assessed (d). N = 5; student’s two-tailed t-test between the antigen-challenged SD and KD groups only; *p < 0.05; **p < 0.01; ***p < 0.001
Fig. 5
Fig. 5
The ketogenic diet had no effect on T cell activity in an acute and chronic mouse model of LCMV infection. Splenocytes from non-tumor bearing mice infected with LCMV Armstrong or Clone 13 were isolated at day 6 and 30, and stimulated with GP33 or NP396 antigens. IFNγ + TNF + CD8+ cells in mice fed SD versus KD (a). PD-1 + CD8+ expression in mice fed SD versus KD (b). N = 5 in each group

References

    1. Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro Oncol. 2013;15:4–27. doi: 10.1093/neuonc/nos273.
    1. Olar A, Aldape KD. Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol. 2014;232:165–177. doi: 10.1002/path.4282.
    1. Woolf EC, Scheck AC. The ketogenic diet for the treatment of malignant glioma. J Lipid Res. 2015;56:5–10. doi: 10.1194/jlr.R046797.
    1. Neal EG, Chaffe H, Schwartz RH, Lawson MS, Edwards N, Fitzsimmons G, et al. The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial. Lancet Neurol. 2008;7:500–506. doi: 10.1016/S1474-4422(08)70092-9.
    1. Stafford P, Abdelwahab MG, Kim DY, Preul MC, Rho JM, Scheck AC. The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr Metab (Lond) 2010;7:74. doi: 10.1186/1743-7075-7-74.
    1. Abdelwahab MG, Fenton KE, Preul MC, Rho JM, Lynch A, Stafford P, et al. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma. PLoS One. 2012;7:e36197. doi: 10.1371/journal.pone.0036197.
    1. Zhou W, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 2007;4:5. doi: 10.1186/1743-7075-4-5.
    1. Seyfried TN, Flores R, Poff AM, D'Agostino DP, Mukherjee P. Metabolic therapy: a new paradigm for managing malignant brain cancer. Cancer Lett. 2015;356:289–300. doi: 10.1016/j.canlet.2014.07.015.
    1. Woolf EC, Curley KL, Liu Q, Turner GH, Charlton JA, Preul MC, et al. The Ketogenic Diet Alters the Hypoxic Response and Affects Expression of Proteins Associated with Angiogenesis, Invasive Potential and Vascular Permeability in a Mouse Glioma Model. PLoS One. 2015;10:e0130357. doi: 10.1371/journal.pone.0130357.
    1. Wei J, Wu A, Kong LY, Wang Y, Fuller G, Fokt I, et al. Hypoxia potentiates glioma-mediated immunosuppression. PLoS One. 2011;6:e16195. doi: 10.1371/journal.pone.0016195.
    1. Joon YA, Bazar KA, Lee PY. Tumors may modulate host immunity partly through hypoxia-induced sympathetic bias. Med Hypotheses. 2004;63:352–356. doi: 10.1016/j.mehy.2004.02.026.
    1. Bruzzese L, Fromonot J, By Y, Durand-Gorde JM, Condo J, Kipson N, et al. NF-kappaB enhances hypoxia-driven T-cell immunosuppression via upregulation of adenosine A(2A) receptors. Cell Signal. 2014;26:1060–1067. doi: 10.1016/j.cellsig.2014.01.024.
    1. Abdelwahab MG, Sankar T, Preul MC, Scheck AC. Intracranial Implantation with Subsequent In Vivo Bioluminescent Imaging of Murine Gliomas. JOVE. 2011;57:e3403.
    1. Jouanneau E, Poujol D, Gulia S, Le MI, Blay JY, Belin MF, et al. Dendritic cells are essential for priming but inefficient for boosting antitumour immune response in an orthotopic murine glioma model. Cancer Immunol Immunother. 2006;55:254–267. doi: 10.1007/s00262-005-0040-7.
    1. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188:2205–2213. doi: 10.1084/jem.188.12.2205.
    1. Villarete L, Somasundaram T, Ahmed R. Tissue-mediated selection of viral variants: correlation between glycoprotein mutation and growth in neuronal cells. J Virol. 1994;68:7490–7496.
    1. Ampie L, Woolf EC, Dardis C. Immunotherapeutic advancements for glioblastoma. Front Oncol. 2015;5:12. doi: 10.3389/fonc.2015.00012.
    1. Nduom EK, Weller M, Heimberger AB. Immunosuppressive mechanisms in glioblastoma. Neuro Oncol. 2015;17 Suppl 7:vii9–vii14. doi: 10.1093/neuonc/nov151.
    1. Kmiecik J, Poli A, Brons NH, Waha A, Eide GE, Enger PO, et al. Elevated CD3+ and CD8+ tumor-infiltrating immune cells correlate with prolonged survival in glioblastoma patients despite integrated immunosuppressive mechanisms in the tumor microenvironment and at the systemic level. J Neuroimmunol. 2013;264:71–83. doi: 10.1016/j.jneuroim.2013.08.013.
    1. Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol. 2010;11:7–13. doi: 10.1038/ni.1818.
    1. Fecci PE, Mitchell DA, Whitesides JF, Xie W, Friedman AH, Archer GE, et al. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006;66:3294–3302. doi: 10.1158/0008-5472.CAN-05-3773.
    1. Husain Z, Huang Y, Seth P, Sukhatme VP. Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J Immunol. 2013;191:1486–1495. doi: 10.4049/jimmunol.1202702.
    1. Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, et al. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003;63:7462–7467.
    1. Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wohrer A, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015;17:1064–1075. doi: 10.1093/neuonc/nou307.
    1. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13:84–88. doi: 10.1038/nm1517.
    1. Avril T, Saikali S, Vauleon E, Jary A, Hamlat A, De TM, et al. Distinct effects of human glioblastoma immunoregulatory molecules programmed cell death ligand-1 (PDL-1) and indoleamine 2,3-dioxygenase (IDO) on tumour-specific T cell functions. J Neuroimmunol. 2010;225:22–33. doi: 10.1016/j.jneuroim.2010.04.003.
    1. Bloch O, Crane CA, Kaur R, Safaee M, Rutkowski MJ, Parsa AT. Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages. Clin Cancer Res. 2013;19:3165–3175. doi: 10.1158/1078-0432.CCR-12-3314.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm0902-1039c.
    1. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–122. doi: 10.1016/j.immuni.2007.05.016.
    1. Fecci PE, Ochiai H, Mitchell DA, Grossi PM, Sweeney AE, Archer GE, et al. Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T cell compartment without affecting regulatory T-cell function. Clin Cancer Res. 2007;13:2158–2167. doi: 10.1158/1078-0432.CCR-06-2070.
    1. Vom BJ, Vrohlings M, Haller S, Haimovici A, Kulig P, Sledzinska A, et al. Intratumoral IL-12 combined with CTLA-4 blockade elicits T cell-mediated glioma rejection. J Exp Med. 2013;210:2803–2811. doi: 10.1084/jem.20130678.
    1. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 2013;86:343–349. doi: 10.1016/j.ijrobp.2012.12.025.
    1. Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res. 2014;20:5290–5301. doi: 10.1158/1078-0432.CCR-14-0514.
    1. Reardon DA, Gokhale PC, Klein SR, Ligon KL, Rodig SJ, Ramkissoon SH, et al. Glioblastoma Eradication Following Immune Checkpoint Blockade in an Orthotopic, Immunocompetent Model. Cancer Immunol Res. 2016;4:124–135. doi: 10.1158/2326-6066.CIR-15-0151.
    1. Patel MA, Pardoll DM. Concepts of immunotherapy for glioma. J Neurooncol. 2015;123:323–330. doi: 10.1007/s11060-015-1810-5.
    1. Yu P, Fu YX. Tumor-infiltrating T lymphocytes: friends or foes? Lab Invest. 2006;86:231–245. doi: 10.1038/labinvest.3700389.
    1. Hagemann T, Wilson J, Burke F, Kulbe H, Li NF, Pluddemann A, et al. Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J Immunol. 2006;176:5023–5032. doi: 10.4049/jimmunol.176.8.5023.
    1. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588.
    1. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75:163–189. doi: 10.1189/jlb.0603252.
    1. Muller-Hubenthal B, Azemar M, Lorenzen D, Huber M, Freudenberg MA, Galanos C, et al. Tumour Biology: tumour-associated inflammation versus antitumor immunity. Anticancer Res. 2009;29:4795–4805.
    1. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025.
    1. Kohrt HE, Nouri N, Nowels K, Johnson D, Holmes S, Lee PP. Profile of immune cells in axillary lymph nodes predicts disease-free survival in breast cancer. PLoS Med. 2005;2:e284. doi: 10.1371/journal.pmed.0020284.
    1. Procaccini C, Galgani M, De Rosa V, Matarese G. Intracellular metabolic pathways control immune tolerance. Trends Immunol. 2012;33:1–7. doi: 10.1016/j.it.2011.09.002.
    1. Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA, Thompson CB. In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol Cell. 2000;6:683–692. doi: 10.1016/S1097-2765(00)00066-6.
    1. Frauwirth KA, Thompson CB. Regulation of T lymphocyte metabolism. J Immunol. 2004;172:4661–4665. doi: 10.4049/jimmunol.172.8.4661.
    1. Yap M, Brouard S, Pecqueur C, Degauque N. Targeting CD8 T-Cell Metabolism in Transplantation. Front Immunol. 2015;6:547.
    1. Lochner M, Berod L, Sparwasser T. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 2015;36:81–91. doi: 10.1016/j.it.2014.12.005.
    1. Morris AAM. Cerebral ketone body metabolism. J Inherit Metab Dis. 2005;28:109–121. doi: 10.1007/s10545-005-5518-0.
    1. Veech RL, Chance B, Kashiwaya Y, Lardy HA, Cahill GF., Jr Ketone bodies, potential therapeutic uses. IUBMB Life. 2001;51:241–247. doi: 10.1080/152165401753311780.
    1. Molon B, Cali B, Viola A. T Cells and Cancer: How Metabolism Shapes Immunity. Front Immunol. 2016;7:20. doi: 10.3389/fimmu.2016.00020.
    1. Sukumar M, Roychoudhuri R, Restifo NP. Nutrient Competition: A New Axis of Tumor Immunosuppression. Cell. 2015;162:1206–1208. doi: 10.1016/j.cell.2015.08.064.

Source: PubMed

3
S'abonner