Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy

Zinal S Chheda, Gary Kohanbash, Kaori Okada, Naznin Jahan, John Sidney, Matteo Pecoraro, Xinbo Yang, Diego A Carrera, Kira M Downey, Shruti Shrivastav, Shuming Liu, Yi Lin, Chetana Lagisetti, Pavlina Chuntova, Payal B Watchmaker, Sabine Mueller, Ian F Pollack, Raja Rajalingam, Angel M Carcaboso, Matthias Mann, Alessandro Sette, K Christopher Garcia, Yafei Hou, Hideho Okada, Zinal S Chheda, Gary Kohanbash, Kaori Okada, Naznin Jahan, John Sidney, Matteo Pecoraro, Xinbo Yang, Diego A Carrera, Kira M Downey, Shruti Shrivastav, Shuming Liu, Yi Lin, Chetana Lagisetti, Pavlina Chuntova, Payal B Watchmaker, Sabine Mueller, Ian F Pollack, Raja Rajalingam, Angel M Carcaboso, Matthias Mann, Alessandro Sette, K Christopher Garcia, Yafei Hou, Hideho Okada

Abstract

The median overall survival for children with diffuse intrinsic pontine glioma (DIPG) is less than one year. The majority of diffuse midline gliomas, including more than 70% of DIPGs, harbor an amino acid substitution from lysine (K) to methionine (M) at position 27 of histone 3 variant 3 (H3.3). From a CD8+ T cell clone established by stimulation of HLA-A2+ CD8+ T cells with synthetic peptide encompassing the H3.3K27M mutation, complementary DNA for T cell receptor (TCR) α- and β-chains were cloned into a retroviral vector. TCR-transduced HLA-A2+ T cells efficiently killed HLA-A2+H3.3K27M+ glioma cells in an antigen- and HLA-specific manner. Adoptive transfer of TCR-transduced T cells significantly suppressed the progression of glioma xenografts in mice. Alanine-scanning assays suggested the absence of known human proteins sharing the key amino acid residues required for recognition by the TCR, suggesting that the TCR could be safely used in patients. These data provide us with a strong basis for developing T cell-based therapy targeting this shared neoepitope.

© 2018 Chheda et al.

Figures

Figure 1.
Figure 1.
H3.3K27M peptide induces T cell responses in DIPG patients and led to isolation of H3.3K27M-specific CTL clones from HLA-A2+ donor PBMCs. (A) Patient-derived and healthy donor-derived PBMCs were stimulated with H3.3WT, H3.3K27M, and flu peptide or without peptide and co-cultured with T2 cells pulsed with H3.3WT, H3.3K27M, and flu peptide or without peptide. Numbers of IFN-γ spots per 5 × 104 T cells generated in each stimulation condition. Counts shown are normalized to no-peptide stimulation control. Bars and error bars represent median and SD, respectively, for IFN-γ spots per 5 × 104 T cells observed in each stimulation condition. n = 3 in each group. Experiment was conducted once. *, P < 0.05; **, P < 0.01 by Student’s t test comparing H3.3WT and H3.3K27M stimulation groups. ns, not significant. (B) HLA-A*02:01+ healthy donor-derived PBMCs were stimulated in vitro with H3.3K27M peptide and evaluated for reactivity against HLA-A*02:01-H3.3K27M-specific tetramer and anti-CD8 mAb using flow cytometry (gated on live lymphocytes). Tetramer+ gate was based on control T cells. Tetramerhigh population (2.42%) represented the high-affinity CTL population. (C) CTL clones were generated by flow-sorting followed by limiting dilution cloning of tetramerhigh CD8+ T cells. The clone 1H5 was chosen for further investigation based on its excellent tetramer reactivity. The dot plot demonstrates that the clone 1H5 retains the reactivity levels to the HLA-A*0201/H3.3K27M-specific tetramer. (D) Bar graph representing IFN-γ production from the T cell clone 1H5 measured by ELISA after co-culture with T2 cells pulsed with H3.3K27M peptide at titrating concentrations or with H3.3WT peptide (5 µg/ml). Data represent two independent experiments with similar results. Bars and error bars represent median and SD, respectively, for IFN-γ production levels. **, P < 0.01 using Student’s t tests comparing 5 µg/ml H3.3WT and H3.3K27M peptide concentrations.
Figure 2.
Figure 2.
The H3.3K27M peptide is detectable by LC-MS/MS in the HLA class I immunopeptidome of glioma cells bearing the H3.3K27M mutation. HLA class I peptides were biochemically purified from U87H3.3K27M glioma cells and analyzed by LC-MS/MS with a synthetic heavy version of the H3.3K27M peptide as the reference. (A) U87H3.3K27M HLA class I immunopeptidome shows two coeluting isotope patterns corresponding to the target m/z and mass difference of the oxidized forms of the heavy and the endogenous H3.3K27M peptides. (B) Fragmentation spectrum of the heavy peak, showing identification of the oxidized heavy H3.3K27M peptide. (C) Zoom-in of the light isotope pattern shows m/z values and distances between peaks as expected from the endogenous H3.3K27M peptide. This experiment was conducted once.
Figure 3.
Figure 3.
Cloning of cDNA for the H3.3K27M-specific TCR and construction of a retroviral vector for efficient transduction of human T cells. (A) Schema of the TCR retroviral vector design. Synthesized TCR cDNA fragments derived from the CD8+ T cell clone 1H5 were inserted into the NotI/XhoI site of Takara siTCR vector plasmid together with the Kozak sequence, spacer sequence (SP), and P2A sequence. (B) T2 cells loaded with or without H3.3K27M, H3.3WT, or irrelevant influenza matrix M158–66 flu peptide (10 µg/ml) were co-cultured with either control or TCR-transduced Jurkat76CD8+ or Jurkat76CD8− cells in a 1:1 ratio and assessed for IL-2 production by ELISA. Data represent three independent experiments with similar results. Bars and error bars represent median and SD, respectively, for IL-2 levels (n = 3 in each group). *, P < 0.05 by Student’s t test compared with each of the other groups. (C) Human PBMCs were transduced with the retroviral TCR vector, and CD3+ T cells were evaluated for transduction efficiency in CD8+ and CD8− T cell populations by the specific tetramer. Of the 51% of CD8+ T cells, 22.5% were tetramer+, and of 49% of CD4+ T cells, 13.4% were tetramer+. (D) TCR-transduced or mock-transduced primary human CD4+ or CD8+ T cells were co-cultured with T2 cells loaded with H3.3K27M, H3.3WT, or flu peptide in a 1:1 ratio overnight for IFN-γ ELISPOT. Bars and error bars represent median and SD, respectively, for IFN-γ spot counts/5 × 104 T cells in triplicate. **, P < 0.01 by Student’s t test comparing TCR-transduced CD8+ versus TCR-transduced CD4+ T cells, and H3.3WT versus H3.3K27M peptide-stimulated groups for TCR-transduced CD8+ T cells. Data represent two independent experiments with similar results. (E) TCR-transduced or mock-transduced CD8+ T cells were co-cultured with T2 cells pulsed with H3.3WT, H3.3K27M, or flu peptide in a 1:1 ratio, followed by evaluation of CD69 expression. Bars and error bars represent median and SD, respectively, for percentage CD69+ cells among CD8+ T cells (n = 3 in each group). Data represent two independent experiments with similar results. *, P < 0.05 by Student’s t test compared with each of the other groups.
Figure 4.
Figure 4.
Alanine scanning mutagenesis determines key amino acid residues in the H3.3K27M epitope required for TCR recognition. Single alanine mutations (10 in total) were introduced at every amino acid residue within the H3.3K27M epitope (10-mer). Hence, 10 synthetic peptides each containing the specific substitution with alanine (A1–A3 and A5–A10) or valine (A4) were evaluated. In addition, synthetic peptides designed for nonmutated H3.3WT peptide and citrullinated H3.3K27M (Cit H3.3; i.e., the first amino acid of the H3.3K27M epitope was replaced by citrulline) were evaluated. (A) Relative HLA-A*0201 binding affinity of each peptide to that of H3.3K27M was determined by cell-free binding assay. This experiment was performed once. (B) J.RT3-T3.5 cells were transduced with H3.3K27M-specific TCR and evaluated for IL-2 production in response to T2 cells loaded with each peptide (10 µg/ml). Each group was assayed in triplicate. Bars and error bars represent median and SD, respectively, for IL-2 production levels. Data represent two independent experiments with similar results. *, P < 0.05 was calculated using Student’s t test comparing each peptide with the mutant H3.3.
Figure 5.
Figure 5.
Evaluation of TCR avidity to the HLA-A2-peptide complex. (A and B) SPR analysis on the binding of the TCR and H3.3K27M-HLA-A2. (A) H3.3K27M TCR at concentrations of 14, 7, 3.5, 1.75, 0.9, 0.45, 0.23, 0.12, and 0 µM was injected over immobilized H3.3K27M-HLA-A2 (500 resonance units [RU]). (B) Fitted curve for equilibrium binding that resulted in a KD of 2.9 µM. Data represent two independent experiments with similar results. (C) T2 cells (5 × 104/well) loaded with titrating concentrations of the H3.3K27M peptide were co-cultured with TCR-transduced CD4+ or CD8+ T cells derived from three donors (5 × 104/well) and assessed by IFN-γ ELISPOT. The EC50 of the peptide was calculated using nonlinear regression analysis. Each experiment was performed in triplicate, and data represent two independent experiments with similar results. (D) T2 cells were pulsed with 10 µg/ml H3.3WT, H3.3K27M, flu peptide, or no peptide, and HLA-A2negH3.3K27M+HSJD-DIPG019 cells were pulsed with 10 µg/ml H3.3K27M peptide. These cells were co-cultured with CD4+ or CD8+ TCR-transduced T cells and assessed by IFN-γ ELISPOT. Bars and error bars represent median and SD, respectively, for IFN-γ spots. Data represent two independent experiments with similar results. Experiments were performed in triplicate. ***, P < 0.001 by Student’s t test comparing the T2/H3.3K27M group with all the other stimulation conditions for CD8+ T cell groups.
Figure 6.
Figure 6.
TCR-transduced T cells lyse H3.3K27M+HLA-A2+ glioma cells in an HLA-A*0201- and H3.3K27M-dependent manner. (A–E) Cytotoxicity of TCR-transduced T cells was evaluated by LDH cytotoxicity assay. Exogenous, synthetic H3.3K27M peptide (10 µg/ml) was added as a positive control group for TCR reactivity for each cell line. HLA-A2 blocking antibody was added in some experiments to determine HLA-A2–dependent TCR reactivity. (A–C) TCR-transduced or mock-transduced T cells were co-cultured with H3.3K27M+HLA-A*0201+ HSJD-DIPG-017 cells (A), H3.3K27M+HLA-A*0201+HSJD-DIPG-021 cells (B), or control H3.3K27M+HLA-A*0201neg HSJD-DIPG-019 cells (C) at an E/T ratio of 1, 5, and 10 for 24 h. *, P < 0.05; **, P < 0.01 based on Student’s t test comparing TCR-transduced T cells with mock-transduced T cells. Each group was assessed in triplicate. Data represent two independent experiments with similar results. (D) HLA-A*0201-negative HSJD-DIPG-019 cells were stably transduced with lentiviral vector encoding HLA-A*0201 and evaluated for HLA-A2 expression by flow cytometry along with other cell lines. TCR-transduced or mock-transduced T cells were co-cultured with DIPG017, DIPG019, or DIPG-019-HLA-A2+ cells at an E/T ratio of 5, and cytotoxicity was measured by LDH assay. Bars and error bars represent median and SD, respectively, for specific percentage cytotoxicity. *, P < 0.05 based on Student’s t test. Each group was assessed in triplicate, and data represent two independent experiments with similar results. (E) TCR-transduced or control T cells were co-cultured with HLA-A*0201+ U87H3.3K27M cells or U87H3.3WT cells at an E/T ratio of 5. Each group was assessed in triplicate. Data represent three independent experiments with similar results. *, P < 0.05 based on Student’s t test. (F) CFSE-labeled target cells were co-cultured with TCR-transduced or control T cells with or without exogenous peptide at an E/T ratio of 5. After 24-h incubation, cells were stained with 7-AAD. %CFSE+7-AAD+ cells indicated specific percent cytotoxicity. Each group was assessed in triplicate. Data represent two independent experiments with similar results. *, P < 0.05 based on Student’s t test.
Figure 7.
Figure 7.
Adoptive transfer of TCR-transduced T cells but not mock-transduced T cells results in inhibition of intracranial H3.3K27M+ glioma in NSG mice. NSG mice bearing intracranial U87H3.3K27M luciferase+ gliomas received intravenous infusion with PBS, mock-transduced T cells or TCR-transduced T cells. (A) Tumor growth is presented as radiance (107 p/s/cm2/r) using BLI. Arrows indicate days on which mice received treatment. Lines and error bars represent median and SD, respectively, for radiance (107 p/s/cm2/r) using BLI (n = 8 per group). (B) Representative BLI images of mice on days 10 and 32 after tumor inoculation. The background BLI signals were defined based on the levels seen in non–tumor-bearing mice. (C) Preferential accumulation of TCR+ T cells in the tumor site. At the time of intravenous infusion, ∼50% and 30% of the infused CD8+ and CD4+ T cells, respectively, were dextramer+ for TCR. On day 2 after the second intravenous infusion, the percentage of dextramer+ cells among CD8+ T cells and CD4+ T cells was evaluated in the peripheral blood and brain of mice that received TCR-transduced T cells. Bars and error bars represent median and SD, respectively, for percentage of dextramer+ cells among total live CD8+ or CD4+ T cells. Data indicate percentage dextramer+ cells among total live CD8+ or CD4+ T cells (n = 5 per group). The experimental results shown in this figure are representative of two independent experiments with reproducible results. *, P < 0.05; **, P < 0.01 using Student’s t test.

References

    1. Adams J.J., Narayanan S., Birnbaum M.E., Sidhu S.S., Blevins S.J., Gee M.H., Sibener L.V., Baker B.M., Kranz D.M., and Garcia K.C.. 2016. Structural interplay between germline interactions and adaptive recognition determines the bandwidth of TCR-peptide-MHC cross-reactivity. Nat. Immunol. 17:87–94. 10.1038/ni.3310
    1. Aleksic M., Liddy N., Molloy P.E., Pumphrey N., Vuidepot A., Chang K.M., and Jakobsen B.K.. 2012. Different affinity windows for virus and cancer-specific T-cell receptors: Implications for therapeutic strategies. Eur. J. Immunol. 42:3174–3179. 10.1002/eji.201242606
    1. Bakker A.B., Marland G., de Boer A.J., Huijbens R.J., Danen E.H., Adema G.J., and Figdor C.G.. 1995. Generation of antimelanoma cytotoxic T lymphocytes from healthy donors after presentation of melanoma-associated antigen-derived epitopes by dendritic cells in vitro. Cancer Res. 55:5330–5334.
    1. Bassani-Sternberg M., Pletscher-Frankild S., Jensen L.J., and Mann M.. 2015. Mass spectrometry of human leukocyte antigen class I peptidomes reveals strong effects of protein abundance and turnover on antigen presentation. Mol. Cell. Proteomics. 14:658–673. 10.1074/mcp.M114.042812
    1. Birnbaum M.E., Mendoza J.L., Sethi D.K., Dong S., Glanville J., Dobbins J., Ozkan E., Davis M.M., Wucherpfennig K.W., and Garcia K.C.. 2014. Deconstructing the peptide-MHC specificity of T cell recognition. Cell. 157:1073–1087. 10.1016/j.cell.2014.03.047
    1. Brain Tumor Progress Review Group 2000. Report of the Brain Tumor Progress Review Group. National Cancer Institute, National Institute of Neurological Disorders and Stroke, Bethesda, MD: NIH pub. no. 01-4902. Available at:
    1. Cameron B.J., Gerry A.B., Dukes J., Harper J.V., Kannan V., Bianchi F.C., Grand F., Brewer J.E., Gupta M., Plesa G., et al. . 2013. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci. Transl. Med. 5:197ra103 10.1126/scitranslmed.3006034
    1. Cheng Y., and Prusoff W.H.. 1973. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 22:3099–3108. 10.1016/0006-2952(73)90196-2
    1. Chiang J.C., and Ellison D.W.. 2017. Molecular pathology of paediatric central nervous system tumours. J. Pathol. 241:159–172. 10.1002/path.4813
    1. Cole D.K., Pumphrey N.J., Boulter J.M., Sami M., Bell J.I., Gostick E., Price D.A., Gao G.F., Sewell A.K., and Jakobsen B.K.. 2007. Human TCR-binding affinity is governed by MHC class restriction. J. Immunol. 178:5727–5734. 10.4049/jimmunol.178.9.5727
    1. Cunningham B.C., and Wells J.A.. 1989. High-resolution epitope mapping of hGH-receptor interactions by alanine-scanning mutagenesis. Science. 244:1081–1085. 10.1126/science.2471267
    1. Cuthbert G.L., Daujat S., Snowden A.W., Erdjument-Bromage H., Hagiwara T., Yamada M., Schneider R., Gregory P.D., Tempst P., Bannister A.J., and Kouzarides T.. 2004. Histone deimination antagonizes arginine methylation. Cell. 118:545–553. 10.1016/j.cell.2004.08.020
    1. Dionne S.O., Smith M.H., Marincola F.M., and Lake D.F.. 2003. Functional characterization of CTL against gp100 altered peptide ligands. Cancer Immunol. Immunother. 52:199–206.
    1. Eguchi J., Hatano M., Nishimura F., Zhu X., Dusak J.E., Sato H., Pollack I.F., Storkus W.J., and Okada H.. 2006. Identification of interleukin-13 receptor alpha2 peptide analogues capable of inducing improved antiglioma CTL responses. Cancer Res. 66:5883–5891. 10.1158/0008-5472.CAN-06-0363
    1. Garboczi D.N., Hung D.T., and Wiley D.C.. 1992. HLA-A2-peptide complexes: Refolding and crystallization of molecules expressed in Escherichia coli and complexed with single antigenic peptides. Proc. Natl. Acad. Sci. USA. 89:3429–3433. 10.1073/pnas.89.8.3429
    1. García-Peydró M., Paradela A., Albar J.P., and Castro J.A.. 2000. Antagonism of direct alloreactivity of an HLA-B27-specific CTL clone by altered peptide ligands of its natural epitope. J. Immunol. 165:5680–5685. 10.4049/jimmunol.165.10.5680
    1. Genßler S., Burger M.C., Zhang C., Oelsner S., Mildenberger I., Wagner M., Steinbach J.P., and Wels W.S.. 2015. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. OncoImmunology. 5:e1119354 10.1080/2162402X.2015.1119354
    1. Govers C., Sebestyén Z., Coccoris M., Willemsen R.A., and Debets R.. 2010. T cell receptor gene therapy: Strategies for optimizing transgenic TCR pairing. Trends Mol. Med. 16:77–87. 10.1016/j.molmed.2009.12.004
    1. Gulukota K., Sidney J., Sette A., and DeLisi C.. 1997. Two complementary methods for predicting peptides binding major histocompatibility complex molecules. J. Mol. Biol. 267:1258–1267. 10.1006/jmbi.1997.0937
    1. Hatano M., Eguchi J., Tatsumi T., Kuwashima N., Dusak J.E., Kinch M.S., Pollack I.F., Hamilton R.L., Storkus W.J., and Okada H.. 2005. EphA2 as a glioma-associated antigen: A novel target for glioma vaccines. Neoplasia. 7:717–722. 10.1593/neo.05277
    1. Heemskerk M.H., Hoogeboom M., de Paus R.A., Kester M.G., van der Hoorn M.A., Goulmy E., Willemze R., and Falkenburg J.H.. 2003. Redirection of antileukemic reactivity of peripheral T lymphocytes using gene transfer of minor histocompatibility antigen HA-2-specific T-cell receptor complexes expressing a conserved alpha joining region. Blood. 102:3530–3540. 10.1182/blood-2003-05-1524
    1. Johnson L.A., Morgan R.A., Dudley M.E., Cassard L., Yang J.C., Hughes M.S., Kammula U.S., Royal R.E., Sherry R.M., Wunderlich J.R., et al. . 2009. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 114:535–546. 10.1182/blood-2009-03-211714
    1. Johnson L.A., Scholler J., Ohkuri T., Kosaka A., Patel P.R., McGettigan S.E., Nace A.K., Dentchev T., Thekkat P., Loew A., et al. . 2015. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci. Transl. Med. 7:275ra22 10.1126/scitranslmed.aaa4963
    1. Jones C., and Baker S.J.. 2014. Unique genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat. Rev. Cancer. 14:651–661. 10.1038/nrc3811
    1. Kebudi R., and Cakir F.B.. 2013. Management of diffuse pontine gliomas in children: Recent developments. Paediatr. Drugs. 15:351–362. 10.1007/s40272-013-0033-5
    1. Khuong-Quang D.A., Buczkowicz P., Rakopoulos P., Liu X.Y., Fontebasso A.M., Bouffet E., Bartels U., Albrecht S., Schwartzentruber J., Letourneau L., et al. . 2012. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol. 124:439–447. 10.1007/s00401-012-0998-0
    1. Lagerwerf F.M., van de Weert M., Heerma W., and Haverkamp J.. 1996. Identification of oxidized methionine in peptides. Rapid Commun. Mass Spectrom. 10:1905–1910. 10.1002/(SICI)1097-0231(199612)10:15<1905::AID-RCM755>;2-9
    1. Lecoeur H., Février M., Garcia S., Rivière Y., and Gougeon M.L.. 2001. A novel flow cytometric assay for quantitation and multiparametric characterization of cell-mediated cytotoxicity. J. Immunol. Methods. 253:177–187. 10.1016/S0022-1759(01)00359-3
    1. Louis D.N., Perry A., Reifenberger G., von Deimling A., Figarella-Branger D., Cavenee W.K., Ohgaki H., Wiestler O.D., Kleihues P., and Ellison D.W.. 2016. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 131:803–820. 10.1007/s00401-016-1545-1
    1. Lupetti R., Pisarra P., Verrecchia A., Farina C., Nicolini G., Anichini A., Bordignon C., Sensi M., Parmiani G., and Traversari C.. 1998. Translation of a retained intron in tyrosinase-related protein (TRP) 2 mRNA generates a new cytotoxic T lymphocyte (CTL)-defined and shared human melanoma antigen not expressed in normal cells of the melanocytic lineage. J. Exp. Med. 188:1005–1016. 10.1084/jem.188.6.1005
    1. Morgan R.A., Yang J.C., Kitano M., Dudley M.E., Laurencot C.M., and Rosenberg S.A.. 2010. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18:843–851. 10.1038/mt.2010.24
    1. Morgan R.A., Chinnasamy N., Abate-Daga D., Gros A., Robbins P.F., Zheng Z., Dudley M.E., Feldman S.A., Yang J.C., Sherry R.M., et al. . 2013. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 36:133–151. 10.1097/CJI.0b013e3182829903
    1. Navai S.A., and Ahmed N.. 2016. Targeting the tumour profile using broad spectrum chimaeric antigen receptor T-cells. Biochem. Soc. Trans. 44:391–396. 10.1042/BST20150266
    1. Ochs K., Ott M., Bunse T., Sahm F., Bunse L., Deumelandt K., Sonner J.K., Keil M., von Deimling A., Wick W., and Platten M.. 2017. K27M-mutant histone-3 as a novel target for glioma immunotherapy. OncoImmunology. 6:e1328340 10.1080/2162402X.2017.1328340
    1. Ohkuri T., Ghosh A., Kosaka A., Zhu J., Ikeura M., David M., Watkins S.C., Sarkar S.N., and Okada H.. 2014. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol. Res. 2:1199–1208. 10.1158/2326-6066.CIR-14-0099
    1. Ohno M., Ohkuri T., Kosaka A., Tanahashi K., June C.H., Natsume A., and Okada H.. 2013. Expression of miR-17-92 enhances anti-tumor activity of T-cells transduced with the anti-EGFRvIII chimeric antigen receptor in mice bearing human GBM xenografts. J. Immunother. Cancer. 1:21 10.1186/2051-1426-1-21
    1. Okada H., Kohanbash G., Zhu X., Kastenhuber E.R., Hoji A., Ueda R., and Fujita M.. 2009. Immunotherapeutic approaches for glioma. Crit. Rev. Immunol. 29:1–42. 10.1615/CritRevImmunol.v29.i1.10
    1. Okamoto S., Mineno J., Ikeda H., Fujiwara H., Yasukawa M., Shiku H., and Kato I.. 2009. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res. 69:9003–9011. 10.1158/0008-5472.CAN-09-1450
    1. Okamoto S., Amaishi Y., Goto Y., Ikeda H., Fujiwara H., Kuzushima K., Yasukawa M., Shiku H., and Mineno J.. 2012. A promising vector for TCR gene therapy: Differential effect of siRNA, 2A peptide, and disulfide bond on the introduced TCR expression. Mol. Ther. Nucleic Acids. 1:e63 10.1038/mtna.2012.52
    1. Okano F., Storkus W.J., Chambers W.H., Pollack I.F., and Okada H.. 2002. Identification of a novel HLA-A*0201-restricted, cytotoxic T lymphocyte epitope in a human glioma-associated antigen, interleukin 13 receptor alpha2 chain. Clin. Cancer Res. 8:2851–2855.
    1. Parkhurst M.R., Yang J.C., Langan R.C., Dudley M.E., Nathan D.A., Feldman S.A., Davis J.L., Morgan R.A., Merino M.J., Sherry R.M., et al. . 2011. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol. Ther. 19:620–626. 10.1038/mt.2010.272
    1. Reardon D.A., Wucherpfennig K.W., Freeman G., Wu C.J., Chiocca E.A., Wen P.Y., Curry W.T. Jr., Mitchell D.A., Fecci P.E., Sampson J.H., and Dranoff G.. 2013. An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev. Vaccines. 12:597–615. 10.1586/erv.13.41
    1. Rosenberg S.A., and Restifo N.P.. 2015. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 348:62–68. 10.1126/science.aaa4967
    1. Rosenberg S.A., Sherry R.M., Morton K.E., Scharfman W.J., Yang J.C., Topalian S.L., Royal R.E., Kammula U., Restifo N.P., Hughes M.S., et al. . 2005. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J. Immunol. 175:6169–6176. 10.4049/jimmunol.175.9.6169
    1. Sasaki K., Zhu X., Vasquez C., Nishimura F., Dusak J.E., Huang J., Fujita M., Wesa A., Potter D.M., Walker P.R., et al. . 2007. Preferential expression of very late antigen-4 on type 1 CTL cells plays a critical role in trafficking into central nervous system tumors. Cancer Res. 67:6451–6458. 10.1158/0008-5472.CAN-06-3280
    1. Schroeder K.M., Hoeman C.M., and Becher O.J.. 2014. Children are not just little adults: Recent advances in understanding of diffuse intrinsic pontine glioma biology. Pediatr. Res. 75:205–209. 10.1038/pr.2013.194
    1. Schumacher T., Bunse L., Pusch S., Sahm F., Wiestler B., Quandt J., Menn O., Osswald M., Oezen I., Ott M., et al. . 2014. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature. 512:324–327. 10.1038/nature13387
    1. Schwartzentruber J., Korshunov A., Liu X.Y., Jones D.T., Pfaff E., Jacob K., Sturm D., Fontebasso A.M., Quang D.A., Tönjes M., et al. . 2012. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 482:226–231. 10.1038/nature10833
    1. Sidney J., Southwood S., Moore C., Oseroff C., Pinilla C., Grey H.M., and Sette A.. 2013. Measurement of MHC/peptide interactions by gel filtration or monoclonal antibody capture. Curr. Protoc. Immunol. Chatper 18:Unit 18.3 10.1002/0471142735.im1803s100
    1. Solomon D.A., Wood M.D., Tihan T., Bollen A.W., Gupta N., Phillips J.J., and Perry A.. 2016. Diffuse midline gliomas with histone H3-K27M mutation: A series of 47 cases assessing the spectrum of morphologic variation and associated genetic alterations. Brain Pathol. 26:569–580. 10.1111/bpa.12336
    1. Thorne A.H., Zanca C., and Furnari F.. 2016. Epidermal growth factor receptor targeting and challenges in glioblastoma. Neuro-oncol. 18:914–918. 10.1093/neuonc/nov319
    1. Tran E., Ahmadzadeh M., Lu Y.C., Gros A., Turcotte S., Robbins P.F., Gartner J.J., Zheng Z., Li Y.F., Ray S., et al. . 2015. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 350:1387–1390. 10.1126/science.aad1253
    1. Tran E., Robbins P.F., Lu Y.C., Prickett T.D., Gartner J.J., Jia L., Pasetto A., Zheng Z., Ray S., Groh E.M., et al. . 2016. T-cell transfer therapy targeting mutant KRAS in cancer. N. Engl. J. Med. 375:2255–2262. 10.1056/NEJMoa1609279
    1. Werfel T., Boeker M., and Kapp A.. 1997. Rapid expression of the CD69 antigen on T cells and natural killer cells upon antigenic stimulation of peripheral blood mononuclear cell suspensions. Allergy. 52:465–469. 10.1111/j.1398-9995.1997.tb01031.x
    1. Wu G., Broniscer A., McEachron T.A., Lu C., Paugh B.S., Becksfort J., Qu C., Ding L., Huether R., Parker M., et al. St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project . 2012. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 44:251–253. 10.1038/ng.1102
    1. Zhang L., and Morgan R.A.. 2012. Genetic engineering with T cell receptors. Adv. Drug Deliv. Rev. 64:756–762. 10.1016/j.addr.2011.11.009
    1. Zhang A.S., Ostrom Q.T., Kruchko C., Rogers L., Peereboom D.M., and Barnholtz-Sloan J.S.. 2017. Complete prevalence of malignant primary brain tumors registry data in the United States compared with other common cancers, 2010. Neuro-oncol. 19:726–735. 10.1093/neuonc/now252
    1. Zhang J.G., Eguchi J., Kruse C.A., Gomez G.G., Fakhrai H., Schroter S., Ma W., Hoa N., Minev B., Delgado C., et al. . 2007. Antigenic profiling of glioma cells to generate allogeneic vaccines or dendritic cell-based therapeutics. Clin. Cancer Res. 13:566–575. 10.1158/1078-0432.CCR-06-1576
    1. Zhu X., Nishimura F., Sasaki K., Fujita M., Dusak J.E., Eguchi J., Fellows-Mayle W., Storkus W.J., Walker P.R., Salazar A.M., and Okada H.. 2007. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J. Transl. Med. 5:10 10.1186/1479-5876-5-10
    1. Zhu X., Fallert-Junecko B.A., Fujita M., Ueda R., Kohanbash G., Kastenhuber E.R., McDonald H.A., Liu Y., Kalinski P., Reinhart T.A., et al. . 2010. Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-alpha and IFN-gamma dependent manners. Cancer Immunol. Immunother. 59:1401–1409. 10.1007/s00262-010-0876-3

Source: PubMed

3
Abonnieren