FGL2 promotes tumor progression in the CNS by suppressing CD103+ dendritic cell differentiation

Jun Yan, Qingnan Zhao, Konrad Gabrusiewicz, Ling-Yuan Kong, Xueqing Xia, Jian Wang, Martina Ott, Jingda Xu, R Eric Davis, Longfei Huo, Ganesh Rao, Shao-Cong Sun, Stephanie S Watowich, Amy B Heimberger, Shulin Li, Jun Yan, Qingnan Zhao, Konrad Gabrusiewicz, Ling-Yuan Kong, Xueqing Xia, Jian Wang, Martina Ott, Jingda Xu, R Eric Davis, Longfei Huo, Ganesh Rao, Shao-Cong Sun, Stephanie S Watowich, Amy B Heimberger, Shulin Li

Abstract

Few studies implicate immunoregulatory gene expression in tumor cells in arbitrating brain tumor progression. Here we show that fibrinogen-like protein 2 (FGL2) is highly expressed in glioma stem cells and primary glioblastoma (GBM) cells. FGL2 knockout in tumor cells did not affect tumor-cell proliferation in vitro or tumor progression in immunodeficient mice but completely impaired GBM progression in immune-competent mice. This impairment was reversed in mice with a defect in dendritic cells (DCs) or CD103+ DC differentiation in the brain and in tumor-draining lymph nodes. The presence of FGL2 in tumor cells inhibited granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced CD103+ DC differentiation by suppressing NF-κB, STAT1/5, and p38 activation. These findings are relevant to GBM patients because a low level of FGL2 expression with concurrent high GM-CSF expression is associated with higher CD8B expression and longer survival. These data provide a rationale for therapeutic inhibition of FGL2 in brain tumors.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
FGL2 is expressed by glioma cells. a FGL2 protein levels were detected by western blot in human peripheral blood monocytes (PBMCs) from healthy donors, normal endothelial cells (HMVECs), normal neurons (HCN-1A), and four glioma stem cell lines (GSC7-2, GSC11, GSC20, and GSC28). Western blots shown are representative, and quantities are summarized as ratio changes from three independent experiments. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as a protein-loading control. b Percentage of FGL2+ cells in cultured human GSCs. c Representative flow cytometry plots of FGL2 expression on CD45+, CD31+, or GFAP+ cells from tumors of patients with newly diagnosed GBM. Data (n = 5) are presented as the mean ± S.D. and analyzed by one-way ANOVA. d Sections of clinical GBM samples were double stained for co-expression of FGL2 (red) and GFAP (green), or of FGL2 (red) and CD45 (green), or of FGL2 (red) and CD31 (green). Nuclei were counterstained with DAPI (blue). White arrows point to co-localization. Scale bars, 50 μm. Data are presented as the mean ± S.D., and representative images are shown (n = 5). A one-way ANOVA was used to calculate the two-sided P values. Significant results were presented as **P < 0.01, ***P < 0.001
Fig. 2
Fig. 2
FGL2 knockout in tumor cells abolishes tumor progression. a Results of DNA fragment deletion in FGL2 exon 1 in individual clones were validated by gene sequencing. Paired gRNAs were designed to excise exon 1 at the mouse FGL2 locus. Individual clones isolated from cells transfected with gRNAs were assayed for deletions and inversions by RT-PCR. b Expression level of FGL2 in three tumor-cell lines, control (Ctrl) and FGL2-knockout (KO) or knockdown (KD) tumor cells, was detected by western blotting. The western blots shown represent three independent experiments. c Survival curve of wild-type (WT) immunocompetent C57BL/6 mice implanted with GL261-Ctrl or GL261-FGL2KO tumor cells (5 × 104 cells per mouse; n = 9 per group). d Survival curve of FGL2−/− mice implanted with GL261-Ctrl or GL261-FGL2KO tumor cells (5 × 104 cells per mouse; n = 5/group). e Survival curve of WT immunocompetent C57BL/6 mice implanted with Lewis lung cancer (LLC)-Ctrl or LLC-FGL2KO tumor cells (5 × 104 cells per mouse; n = 4 per group). f Survival curve of WT immunocompetent BALB/C mice implanted with mouse astrocytoma (DBT)-Ctrl cells or DBT-FGL2KD tumor cells (5 × 104 cells per mouse; n = 4 per group). g Bioluminescence imaging showing DBT-Ctrl and DBT-FGL2KD tumors at day 1 and day 7 after tumor-cell implantation. All data are representative of at least two independent experiments. The survival curves were analyzed by Kaplan–Meier analysis and the log-rank test was used to compare overall survival between groups
Fig. 3
Fig. 3
CD8+ T cell-dependent clearance of FGL2KO tumor. a Survival analysis of immunodeficient NSG mice implanted with GL261-Ctrl or GL261-FGL2KO tumor cells (5 × 104 cells per mouse; n = 6 per group). bd Survival analysis of WT immunocompetent C57BL/6 mice implanted with GL261-FGL2KO tumor cells and treated with anti-CD4 (GK1.5), anti-NK1.1 (PK136), or anti-CD8 (2.43) depletion antibody or IgG as control (Ctrl) (n = 6 per group). e Survival analysis of WT mice and CD8−/− mice implanted with GL261-FGL2KO tumor cells (n = 6–8 per group). f H&E-stained histopathological sections of brain tissues from representative mice from each group shown in e. Scale bar = 1 cm. g Survival analysis of WT mice and CD8−/− mice implanted with LLC-FGL2KO tumor cells (n = 6 per group). h Cytolytic activity of CD8+ T cells from tumor-bearing mice against GL261 tumor cells. Lymphocytes from tumor-draining lymph nodes (TDLNs) or spleens of mice implanted with FGL2KO tumor cells caused significantly more tumor lysis than those from mice implanted with Ctrl tumor cells (n = 3 mice per group). Two-way ANOVA was used to calculate the two-sided P values. E:T, the ratio of effector cells to target cells. *P < 0.05, **P < 0.01, ***P < 0.001 compared with GL261-Ctrl tumor-bearing mice. i Tumor rechallenge by subcutaneous implantation of GL261-Ctrl cells at days 45, 90, and 135 days after primary challenge of GL261-FGL2KO by intracerebral implantation in WT mice (n = 7). Subcutaneous growth of GL261-Ctrl tumors in age-matched naive WT mice challenged with GL261-Ctrl cells at the same times but without primary challenge is also shown. j The experiment shown in i was repeated in FGL2−/− mice (n = 6). All data are representative of at least two independent experiments. The survival curves were analyzed by Kaplan–Meier analysis and the log-rank test was used to compare overall survival between groups
Fig. 4
Fig. 4
Batf3-dependent CD103+ dendritic cells are required for CD8+ T cells priming. a Brain-infiltrating leukocytes (BIL) and tumor-draining lymph nodes (TDLNs) of Ctrl or FGL2KO tumor-bearing wild-type (WT) mice and FGL2KO tumor-bearing Batf3−/− mice were analyzed for CD103+/CD8a+ dendritic cell (DCs) populations on day 7 post tumor implantation. Data are presented as the mean ± S.D. and were analyzed by one-way ANOVA (n = 5–7 per group). bFGL2KO tumor progression in CD11c-DTR mice treated with diphtheria toxin (DT) or PBS. FGL2KO tumor cells were implanted subcutaneously into CD11c-DTR mice on day 1 after injection of DT or PBS. Data are presented as the mean ± S.D. (n = 8 per group). c Survival analysis of Batf3−/− mice implanted with GL261-FGL2KO tumor cells (n = 7 per group). d FACS assay of CD69 on naive OT-I CD8+ T cells co-cultured with DCs isolated from tumors at a ratio of 1:2. Data are presented as the mean ± S.D. (n = 3 mice per group) and were analyzed by t-test. e Abundance and proliferation of CFSE-labeled OT-I cells in brains and TDLNs of GL261-Ctrl-OVA or GL261-FGL2KO-OVA tumor-bearing mice. CFSE-labeled OT-I cells were adoptively transferred to tumor-bearing CD45.1 mice on day 2 after tumor implantation. Brains and TDLNs were collected 5 days later for analyzing the presence and proliferation of the CD45.2+CD8+ T cell population. Data are presented as the mean ± S.D. (n = 5-6 per group) and were analyzed by t-test. f Representative cytometric analysis of CD8+ T cell priming (CD8+T-bet+Eomes−) in BIL and TDLNs at 7 days after tumor implantation. Data are presented as the mean ± S.D. (n = 5 per group) and were analyzed by t-test. g Survival analysis of WT mice implanted with GL261-FGL2KO tumor cells treated with FTY720. Mice were treated with 1 mg/kg FTY720 1 h before tumor-cell implantation and were maintained with drinking water containing 2 μg/mL FTY720 for the duration of the experiment (n = 6 per group). All data are representative of at least two independent experiments. The survival curves were analyzed by Kaplan–Meier analysis and the log-rank test was used to compare overall survival between groups. Significant results were presented as *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 5
Fig. 5
FGL2 suppresses GM-CSF-dependent CD103 induction on cDCs in vitro. a Expression of CD103 on CD11c+B220− DCs cultured with conditioned medium (CM) for 5 or 15 days. Bone marrow cells were isolated from C57BL/6 mice and then cultured with Ctrl-CM (CM from Ctrl tumor cells) or FGL2KO-CM (CM from FGL2KO tumor cells) for 5 days or 15 days. Non-adherent cells were recovered, stained, and analyzed by FACS to determine the expression levels of CD103, Clec9A, and MHCII on the CD11c+B220− DCs. b OT-I T cell activation by OVA-pulsed CM-cultured bone marrow DCs. Non-adherent bone marrow DCs were recovered from CM-cultured bone marrow DCs at 15 days. The bone marrow DCs (2 × 104) were incubated with soluble OVA protein (100 μg/mL) for 2 h and then washed twice before co-culturing with the CFSE-labeled OT-I cells (5 × 104). OT-I activation was measured by CD69 expression level, which was determined by flow cytometry. c Protein levels of GM-CSF and FLT3L in medium conditioned by tumor cells were detected by ELISA. Data were summarized from five independent experiments and were analyzed by paired t-test. d Representative FACS assay of CD103 expression on CD11c+B220− DCs cultured with FGL2KO-CM in the presence of neutralization antibody anti-GM-CSF (5 μg/mL) or anti-FLT3L (5 μg/mL) for 5 days. Data were summarized from four independent experiments and were analyzed by paired t-test. **P < 0.01 comparing with IgG. e Representative FACS assay of CD103 expression on wild-type (WT) and FGL2KO bone marrow (BM) cells cultured with rmGM-CSF (10 ng/mL) for 7 days with or without rFGL2 (200 ng/mL). All plots are representative of three independent experiments
Fig. 6
Fig. 6
FGL2 suppresses NF-κBp65 and STAT1/5 signaling for CD103 induction. a, Expression levels of key proteins in the TAK1-NF-κB/p38 cascades and JAK2/STAT1/5 signaling were examined by western blotting in bone marrow cells cultured with conditioned medium (CM) from control tumor cells (Ctrl), FGL2KO tumor cells, Ctrl-CM + IgG, Ctrl-CM + anti-FGL2, FGL2KO-CM + IgG, or FGL2KO-CM + anti-GM-CSF for 3 days. Data were summarized as ratio changes from at least three independent experiments. The t-test was used to calculate the two-sided P values. Significant results were presented as *P < 0.05, **P < 0.01, ***P < 0.001. b Representative FACS analysis of CD103 expression on CD11c+B220- dendritic cells (DCs), which were pretreated for 1 h with IκBα inhibitor Bay 11-7085 (Bay), NF-κB inhibitor 6-amino-4-(4-phenoxyphenylethylamino) quinazoline (QNZ), JAK2 inhibitor JSI-124 (JSI), or p38 inhibitor PH797804 (PH) in bone marrow cells cultured with CM for 5 days. Plots are representative of three independent experiments. c Schematic illustration of cellular and molecular events underlying FGL2-regulated GBM progression. Tumor cells secreted GM-CSF and FGL2 simultaneously. GM-CSF-induced CD103+ DCs development was blocked in the presence of FGL2, so that CD8+ T cells were not primed and activated because of small CD103+ DC populations. Less CD103+ DC differentiation and subsequent lack of CD8+ T cell priming and activation resulted in GBM progression. For molecular signaling, TRAF6/TAK1/NF-κB/p38 signal and JAK2/STAT1/5 were activated in response to GM-CSF, contributing to CD103 induction. These signaling cascades were blocked by FGL2, thereby suppressing CD103 induction on DCs
Fig. 7
Fig. 7
Expression level of FGL2 and GM-CSF predicts outcome in GBM patients. Data on survival of GBM patients and gene mRNA expression data (n = 401 patients) were downloaded and retrieved from the TCGA data portal ([http://www.cbioportal.org/public-portal/] Accessed between January 1, 2018, and March 15, 2018). The expression levels for both genes were categorized as high or low using median values as the cutoffs. ad Correlations analysis of mRNA expression levels among FGL2, GM-CSF, CD8B, and IFNG in GBM tumors analyzed using the Pearson correlation coefficient. R = Pearson correlation coefficient. e Kaplan–Meier survival analysis for OS of GBM patients in the TCGA dataset grouped by expression levels of FGL2 and GM-CSF mRNA. Patients’ tumors were classified as FGL2highGM-CSFlow, FGL2highGM-CSFhigh, FGL2lowGM-CSFlow, or FGL2lowGM-CSFhigh. The log-rank test was used to compare overall survival among groups. f Kaplan–Meier survival analysis for OS of GBM patients in the FGL2lowGM-CSFhigh group vs. all others. The log-rank test was used to compare overall survival between groups. g Kaplan–Meier survival analysis for OS of GBM patients in the FGL2lowCD8BhighIFNGhigh group vs. those in the FGL2highCD8BlowIFNGlow group. The log-rank test was used to compare overall survival between groups. The statistical analyses were conducted using the R software package

References

    1. Prasetyanti PR, Medema JP. Intra-tumor heterogeneity from a cancer stem cell perspective. Mol. Cancer. 2017;16:41. doi: 10.1186/s12943-017-0600-4.
    1. Singh SK, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401. doi: 10.1038/nature03128.
    1. Wainwright DA, et al. IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival. Clin. Cancer Res. 2012;18:6110–6121. doi: 10.1158/1078-0432.CCR-12-2130.
    1. Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. 2016;37:855–865. doi: 10.1016/j.it.2016.09.006.
    1. Roberts EW, et al. Critical role for CD103 + /CD141 + dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30:324–336. doi: 10.1016/j.ccell.2016.06.003.
    1. Marazzi S, et al. Characterization of human fibroleukin, a fibrinogen-like protein secreted by T lymphocytes. J. Immunol. 1998;161:138–147.
    1. McGilvray ID, et al. Murine hepatitis virus strain 3 induces the macrophage prothrombinase fgl-2 through p38 mitogen-activated protein kinase activation. J. Biol. Chem. 1999;273:32222–32229. doi: 10.1074/jbc.273.48.32222.
    1. Yan, J. et al. FGL2 as a multimodality regulator of tumor-mediated immune suppression and therapeutic target in gliomas. J. Natl Cancer Inst.107, djv137 (2015).
    1. Chan CWY, et al. Soluble fibrinogen-like protein 2/fibroleukin exhibits immunosuppressive properties: suppressing T cell proliferation and inhibiting maturation of bone marrow-derived dendritic cells. J. Immunol. 2003;170:4036–4044. doi: 10.4049/jimmunol.170.8.4036.
    1. Rabizadeh E, et al. The cell-membrane prothrombinase, fibrinogen-like protein 2, promotes angiogenesis and tumor development. Thromb. Res. 2015;136:118–124. doi: 10.1016/j.thromres.2014.11.023.
    1. Fidler IJ, Balasubramanian K, Lin Q, Kim SW, Kim SJ. The brain microenvironment and cancer metastasis. Mol. Cells. 2010;30:93–98. doi: 10.1007/s10059-010-0133-9.
    1. Preusser M, et al. Recent advances in the biology and treatment of brain metastases of non-small cell lung cancer: summary of a multidisciplinary roundtable discussion. ESMO Open. 2018;3:e000262. doi: 10.1136/esmoopen-2017-000262.
    1. Meng B, et al. Structural and functional analyses of human tryptophan 2,3-dioxygenase. Proteins Struct. Funct. Bioinformatics. 2014;82:3210–3216. doi: 10.1002/prot.24653.
    1. Shalev I, et al. The role of FGL2 in the pathogenesis and treatment of hepatitis C virus infection. Rambam Maimonides Med. J. 2010;1:e0004. doi: 10.5041/RMMJ.10004.
    1. Kaech SM, Ahmed R. Memory CD8 + T cell differentiation: initial antigen encounter triggers a developmental program in naive cells. Nat. Immunol. 2001;2:415–422. doi: 10.1038/87720.
    1. Hildner K, et al. Batf3 deficiency reveals a critical role for CD8a + dendritic cells in cytotoxic T cell immunity. Science. 2008;322:1097–1100. doi: 10.1126/science.1164206.
    1. Ferris ST, et al. A minor subset of Batf3-dependent antigen-presenting cells in islets of langerhans is essential for the development of autoimmune diabetes. Immunity. 2014;41:657–669. doi: 10.1016/j.immuni.2014.09.012.
    1. Spranger S, Dai D, Horton B, Gajewski TF. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31:711–723.e4. doi: 10.1016/j.ccell.2017.04.003.
    1. Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26:638–652. doi: 10.1016/j.ccell.2014.09.007.
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–235. doi: 10.1038/nature14404.
    1. Lazarevic V, Glimcher LH, Lord GM. T-bet: A bridge between innate and adaptive immunity. Nat. Rev. Immunol. 2013;13:777–789. doi: 10.1038/nri3536.
    1. Salmon H, et al. Expansion and activation of CD103 + dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44:924–938. doi: 10.1016/j.immuni.2016.03.012.
    1. Mempel TR, Henrickson SE, Von Andrian UH. T-cell priming by dendritic cells in lymph nodes occurs in three distinct phases. Nature. 2004;427:154–159. doi: 10.1038/nature02238.
    1. Liang F, et al. Vaccine priming is restricted to draining lymph nodes & controlled by adjuvant-mediated antigen uptake. Sci. Transl. Med. 2017;9:eaal2094. doi: 10.1126/scitranslmed.aal2094.
    1. Idzko M, et al. Local application of FTY720 to the lung abrogates experimental asthma by altering dendritic cell function. J. Clin. Invest. 2006;116:2935–2944. doi: 10.1172/JCI28295.
    1. Zhang JG, et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity. 2012;36:646–657. doi: 10.1016/j.immuni.2012.03.009.
    1. Poulin LF, et al. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood. 2012;119:6052–6062. doi: 10.1182/blood-2012-01-406967.
    1. Mayer CT, et al. Selective and ef fi cient generation of functional Batf3-dependent CD103 1 dendritic cells from mouse bone marrow. Blood. 2015;124:3081–3092. doi: 10.1182/blood-2013-12-545772.
    1. Belz GT, Nutt SL. Transcriptional programming of the dendritic cell network. Nat. Rev. Immunol. 2012;12:101–113. doi: 10.1038/nri3149.
    1. Van De Laar L, Coffer PJ, Woltman AM. Regulation of dendritic cell development by GM-CSF: Molecular control and implications for immune homeostasis and therapy. Blood. 2012;119:3383–3393. doi: 10.1182/blood-2011-11-370130.
    1. Perng, P. & Lim, M. Immunosuppressive mechanisms of malignant gliomas: parallels at non-CNS sites. Front. Oncol. 5, 153 (2015).
    1. Hussain SF. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro. Oncol. 2006;8:261–279. doi: 10.1215/15228517-2006-008.
    1. Gabrusiewicz K, et al. Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype. JCI Insight. 2016;1:e85841. doi: 10.1172/jci.insight.85841.
    1. Raychaudhuri B, et al. Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro. Oncol. 2011;13:591–599. doi: 10.1093/neuonc/nor042.
    1. Matyszak MK, et al. Microglia induce myelin basic protein-specific T cell anergy or T cell activation, according to their state of activation. Eur. J. Immunol. 1999;29:3063–3076. doi: 10.1002/(SICI)1521-4141(199910)29:10<3063::AID-IMMU3063>;2-G.
    1. Quail DF, Joyce JA. The microenvironmental landscape of brain tumors. Cancer Cell. 2017;31:326–341. doi: 10.1016/j.ccell.2017.02.009.
    1. Zhai H, Heppner FL, Tsirka SE. Microglia/macrophages promote glioma progression. Glia. 2011;59:472–485. doi: 10.1002/glia.21117.
    1. Candolfi M, et al. Plasmacytoid dendritic cells in the tumor microenvironment: immune targets for glioma therapeutics. Neoplasia. 2012;14:757–IN26. doi: 10.1593/neo.12794.
    1. Le Bon A, et al. Cross-priming of CD8 + T cells stimulated by virus-induced type I interferon. Nat. Immunol. 2003;4:1009–1015. doi: 10.1038/ni978.
    1. Tamoutounour S, et al. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin. Immunity. 2013;39:925–938. doi: 10.1016/j.immuni.2013.10.004.
    1. Ricard C, et al. Phenotypic dynamics of microglial and monocyte-derived cells in glioblastoma-bearing mice. Sci. Rep. 2016;6:26381. doi: 10.1038/srep26381.
    1. Sánchez-Paulete AR, et al. Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy. Ann. Oncol. 2017;28:xii44–xii55. doi: 10.1093/annonc/mdx237.
    1. Haniffa M, et al. Human tissues contain CD141 hi cross-presenting dendritic cells with functional homology to mouse CD103 + nonlymphoid dendritic cells. Immunity. 2012;37:60–73. doi: 10.1016/j.immuni.2012.04.012.
    1. Quintana E, et al. DNGR-1 + dendritic cells are located in meningeal membrane and choroid plexus of the noninjured brain. Glia. 2015;63:2231–2248. doi: 10.1002/glia.22889.
    1. Lau-Kilby AW, et al. Interleukin-2 inhibits FMS-like tyrosine kinase 3 receptor ligand (flt3L)-dependent development and function of conventional and plasmacytoid dendritic cells. Proc. Natl Acad. Sci. USA. 2011;108:2408–2413. doi: 10.1073/pnas.1009738108.
    1. Zhan Y, Xu Y, Lew AM. The regulation of the development and function of dendritic cell subsets by GM-CSF: More than a hematopoietic growth factor. Mol. Immunol. 2012;52:30–37. doi: 10.1016/j.molimm.2012.04.009.
    1. Helft J, et al. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c + MHCII + macrophages and dendritic cells. Immunity. 2015;42:1197–1211. doi: 10.1016/j.immuni.2015.05.018.
    1. Greter M, et al. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity. 2012;36:1031–1046. doi: 10.1016/j.immuni.2012.03.027.
    1. Yan WL, Shen KY, Tien CY, Chen YA, Liu SJ. Recent progress in GM-CSF-based cancer immunotherapy. Immunotherapy. 2017;9:347–360. doi: 10.2217/imt-2016-0141.
    1. Parmiani G, et al. Opposite immune functions of GM-CSF administered as vaccine adjuvant in cancer patients. Ann. Oncol. 2007;18:226–232. doi: 10.1093/annonc/mdl158.
    1. Hong, I. S. Stimulatory versus suppressive effects of GM-CSF on tumor progression in multiple cancer types. Exp. Mol. Med. 48, e242 (2016).
    1. Creelan BC, et al. Phase II trial of a GM-CSF-producing and CD40L-expressing bystander cell line combined with an allogeneic tumor cell-based vaccine for refractory lung adenocarcinoma. J. Immunother. 2013;36:442–450. doi: 10.1097/CJI.0b013e3182a80237.
    1. Faries MB, Hsueh EC, Ye X, Hoban M, Morton DL. Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine. Clin. Cancer Res. 2009;15:7029–7035. doi: 10.1158/1078-0432.CCR-09-1540.
    1. Westermann J, Hecker AC, Flörcken A, Dörken B, Pezzutto A. Granulocyte macrophage-colony stimulating factor plus interleukin-2 plus??-interferon plus 5-fluorouracil in the treatment of metastatic renal cell cancer: Induction of CD80/86+T cells indicates adverse outcome. J. Immunother. 2009;32:667–675. doi: 10.1097/CJI.0b013e3181a950e5.
    1. Revoltella RP, Menicagli M, Campani D. Granulocyte-macrophage colony-stimulating factor as an autocrine survival-growth factor in human gliomas. Cytokine. 2012;57:347–359. doi: 10.1016/j.cyto.2011.11.016.
    1. Choi JK, Kim KH, Park H, Park SR, Choi BH. Granulocyte macrophage-colony stimulating factor shows anti-apoptotic activity in neural progenitor cells via JAK/STAT5-Bcl-2 pathway. Apoptosis. 2011;16:127–134. doi: 10.1007/s10495-010-0552-2.
    1. Li HS, et al. The signal transducers STAT5 and STAT3 control expression of Id2 and E2-2 during dendritic cell development. Blood. 2012;120:4363–4373. doi: 10.1182/blood-2012-07-441311.
    1. Louis DN, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114:97–109. doi: 10.1007/s00401-007-0243-4.
    1. Shalev, I. et al. Targeted deletion of fgl2 leads to impaired regulatory T cell activity and development of autoimmune glomerulonephritis. J. Immunol. 180, 249–260 (2008).
    1. Latha, K. et al. The role of fibrinogen-like protein 2 on immunosuppression and malignant progression in glioma. J. Natl Cancer Inst. djy107 (2018).
    1. Wei J, et al. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res. 2013;73:3913–3926. doi: 10.1158/0008-5472.CAN-12-4318.
    1. Jost, S. C., Collins, L., Travers, S., Piwnica-Worms, D. & Garbow, J. R. Measuring brain tumor growth: combined bioluminescence imaging-magnetic resonance imaging strategy. Mol. Imaging. 8, 245–253 (2009)
    1. Sokolovska A, et al. Activation of caspase-1 by the NLRP3 inflammasome regulates the NADPH oxidase NOX2 to control phagosome function. Nat. Immunol. 2013;14:543–553. doi: 10.1038/ni.2595.
    1. Blaskovich MA, et al. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003;63:1270–1279.
    1. Senger, K. et al. The kinase TPL2 activates ERK and p38 signaling to promote neutrophilic inflammation. Sci. Signal. 10, eaah4273 (2017).
    1. Sathe P, et al. The acquisition of antigen cross-presentation function by newly formed dendritic cells. J. Immunol. 2011;186:5184–5192. doi: 10.4049/jimmunol.1002683.
    1. Jin D, et al. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res. 2010;70:2245–2255. doi: 10.1158/0008-5472.CAN-09-3109.
    1. Mitchell TC, et al. Immunological adjuvants promote activated T cell survival via induction of Bcl-3. Nat. Immunol. 2001;2:397–402. doi: 10.1038/87692.
    1. Koenen P, et al. Mutually exclusive regulation of T cell survival by IL-7R and antigen receptor-induced signals. Nat. Commun. 2013;4:1735. doi: 10.1038/ncomms2719.
    1. Caruana I, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 2015;21:524–529. doi: 10.1038/nm.3833.
    1. LaFrance-Corey, R. G. & Howe, C. L. Isolation of brain-infiltrating leukocytes. J. Vis. Exp. 10.3791/2747 (2011).
    1. Verhaak RGW, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110. doi: 10.1016/j.ccr.2009.12.020.
    1. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385.

Source: PubMed

3
구독하다