期刊论文详细信息
Journal for ImmunoTherapy of Cancer
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
Hideho Okada3  Atsushi Natsume2  Carl H June4  Kuniaki Tanahashi2  Akemi Kosaka1  Takayuki Ohkuri1  Masasuke Ohno1 
[1] Department of Neurological Surgery, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA;Department of Neurosurgery, Nagoya University School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan;Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA;Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
关键词: Adoptive immunotherapy;    Glioblastoma;    Chimeric antigen receptor;    miR-17-92;    microRNA;   
Others  :  814871
DOI  :  10.1186/2051-1426-1-21
 received in 2013-06-18, accepted in 2013-12-05,  发布年份 2013
PDF
【 摘 要 】

Background

Expression of miR-17-92 enhances T-cell survival and interferon (IFN)-γ production. We previously reported that miR-17-92 is down-regulated in T-cells derived from glioblastoma (GBM) patients. We hypothesized that transgene-derived co-expression of miR17-92 and chimeric antigen receptor (CAR) in T-cells would improve the efficacy of adoptive transfer therapy against GBM.

Methods

We constructed novel lentiviral vectors for miR-17-92 (FG12-EF1a-miR-17/92) and a CAR consisting of an epidermal growth factor receptor variant III (EGFRvIII)-specific, single-chain variable fragment (scFv) coupled to the T-cell receptor CD3ζ chain signaling module and co-stimulatory motifs of CD137 (4-1BB) and CD28 in tandem (pELNS-3C10-CAR). Human T-cells were transduced with these lentiviral vectors, and their anti-tumor effects were evaluated both in vitro and in vivo.

Results

CAR-transduced T-cells (CAR-T-cells) exhibited potent, antigen-specific, cytotoxic activity against U87 GBM cells that stably express EGFRvIII (U87-EGFRvIII) and, when co-transduced with miR-17-92, exhibited improved survival in the presence of temozolomide (TMZ) compared with CAR-T-cells without miR-17-92 co-transduction. In mice bearing intracranial U87-EGFRvIII xenografts, CAR-T-cells with or without transgene-derived miR-17-92 expression demonstrated similar levels of therapeutic effect without demonstrating any uncontrolled growth of CAR-T-cells. However, when these mice were re-challenged with U87-EGFRvIII cells in their brains, mice receiving co-transduced CAR-T-cells exhibited improved protection compared with mice treated with CAR-T-cells without miR-17-92 co-transduction.

Conclusion

These results warrant the development of novel CAR-T-cell strategies that incorporate miR-17-92 to improve therapeutic potency, especially in patients with GBM.

【 授权许可】

   
2013 Ohno et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140710050855368.pdf 1110KB PDF download
Figure 5. 25KB Image download
Figure 4. 45KB Image download
Figure 3. 85KB Image download
Figure 2. 117KB Image download
Figure 1. 32KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

【 参考文献 】
  • [1]Okada H, et al.: Immunotherapeutic approaches for glioma. Crit Rev Immunol 2009, 29:1-42.
  • [2]Aguilar LK, Arvizu M, Aguilar-Cordova E, Chiocca EA: The spectrum of vaccine therapies for patients with glioblastoma multiforme. Curr Treat Options Oncol 2012, 13:437-450.
  • [3]Ruzevick J, Jackson C, Phallen J, Lim M: Clinical trials with immunotherapy for high-grade glioma. Neurosurg Clin N Am 2012, 23:459-470.
  • [4]Okada H, et al.: Induction of CD8+ T-Cell Responses Against Novel Glioma-Associated Antigen Peptides and Clinical Activity by Vaccinations With {alpha}-Type 1 Polarized Dendritic Cells and Polyinosinic-Polycytidylic Acid Stabilized by Lysine and Carboxymethylcellulose in Patients With Recurrent Malignant Glioma. J Clin Oncol 2011, 29:330-336.
  • [5]Kalos M, et al.: T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia. Sci Transl Med 2011, 3:95ra73.
  • [6]Porter DL, Levine BL, Kalos M, Bagg A, June CH: Chimeric Antigen Receptor–Modified T Cells in Chronic Lymphoid Leukemia. N Engl J Med 2011, 365:725-733.
  • [7]Brentjens RJ, et al.: Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011, 118:4817-4828.
  • [8]Kochenderfer JN, et al.: B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012, 119:2709-2720.
  • [9]Facoetti A, et al.: Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors. Clin Cancer Res 2005, 11:8304-8311.
  • [10]Yeung JT, et al.: LOH in the HLA Class I region at 6p21 is Associated with Shorter Survival in Newly Diagnosed Adult Glioblastoma. Clin Cancer Res 2013. Epub ahead of print
  • [11]Pule MA, et al.: A Chimeric T Cell Antigen Receptor that Augments Cytokine Release and Supports Clonal Expansion of Primary Human T Cells. Mol Ther 2005, 12:933-941.
  • [12]Carpenito C, et al.: Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci 2009, 106:3360-3365.
  • [13]Imai C, et al.: Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 2004, 18:676-684.
  • [14]Moeller M, et al.: A functional role for CD28 costimulation in tumor recognition by single-chain receptor-modified T cells. Cancer Gene Ther 2004, 11:371-379.
  • [15]Morgan RA, et al.: Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2. Mol Ther 2010, 18:843-851.
  • [16]Hatanpaa KJ, Burma S, Zhao D, Habib AA: Epidermal growth factor receptor in glioma: signal transduction, neuropathology, imaging, and radioresistance. Neoplasia 2010, 12:675-684.
  • [17]Mukasa A, et al.: Mutant EGFR is required for maintainenece of glioma growth in vivo, and its ablation leads to escape from receptor dependence. Proc Natl Acad Sci U S A 2010, 107:2616-2621.
  • [18]Heimberger AB, Suki D, Yang D, Shi W, Aldape K: The natural history of EGFR and EGFRvIII in glioblastoma patients. J Transl Med 2005, 3:38. BioMed Central Full Text
  • [19]Heimberger AB, et al.: Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res 2005, 11:1462-1466.
  • [20]Zhu X, et al.: Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-α and IFN-γ dependent manners. Cancer Immunol Immunother 2010, 59:1401-1409.
  • [21]Fujita M, et al.: Effective Immunotherapy against Murine Gliomas Using Type 1 Polarizing Dendritic Cells–Significant Roles of CXCL10. Cancer Res 2009, 69:1587-1595.
  • [22]Nishimura F, et al.: Adoptive transfer of Type 1 CTL mediates effective anti-central nervous system tumor response: critical roles of IFN-inducible protein-10. Cancer Res 2006, 66:4478-4487.
  • [23]Ueda R, et al.: Dicer-regulated microRNAs 222 and 339 promote resistance of cancer cells to cytotoxic T-lymphocytes by down-regulation of ICAM-1. Proc Natl Acad Sci U S A 2009, 106:10746-10751.
  • [24]Kohanbash G, Okada H: MicroRNAs and STAT interplay. Semin Cancer Biol 2012, 22:70-75.
  • [25]Xiao C, et al.: Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat Immunol 2008, 9:405-414.
  • [26]Sasaki K, et al.: miR-17-92 expression in differentiated T cells - Implications for cancer immunotherapy. J Transl Med 2010, 8:17. URL: http://www.translational-medicine.com/content/8/1/17 webcite BioMed Central Full Text
  • [27]Stupp R, et al.: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005, 352:987-996.
  • [28]Salaun B, et al.: Differentiation associated regulation of microRNA expression in vivo in human CD8+ T cell subsets. J Transl Med 2011, 9:44. BioMed Central Full Text
  • [29]Wu T, et al.: Temporal expression of microRNA cluster miR-17-92 regulates effector and memory CD8+ T-cell differentiation. Proc Natl Acad Sci U S A 2012, 109:9965-9970.
  • [30]Jiang S, et al.: Molecular dissection of the miR-17-92 cluster's critical dual roles in promoting Th1 responses and preventing inducible Treg differentiation. Blood 2011, 118:5487-5497.
  • [31]Olive V, Li Q, He L: mir-17-92: a polycistronic oncomir with pleiotropic functions. Immunol Rev 2013, 253:158-166.
  • [32]Molitoris JK, McColl KS, Distelhorst CW: Glucocorticoid-mediated repression of the oncogenic microRNA cluster miR-17 92 contributes to the induction of Bim and initiation of apoptosis. Mol Endocrinol 2011, 25:409-420.
  • [33]van Haaften G, Agami R: Tumorigenicity of the miR-17-92 cluster distilled. Genes Dev 2010, 24:1-4.
  • [34]Zhao Y, et al.: Multiple Injections of Electroporated Autologous T Cells Expressing a Chimeric Antigen Receptor Mediate Regression of Human Disseminated Tumor. Cancer Res 2010, 70:9053-9061.
  • [35]Morgan RA, et al.: Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 2012, 23:1043-1053.
  • [36]Sampson JH, et al.: Immunologic Escape After Prolonged Progression-Free Survival With Epidermal Growth Factor Receptor Variant III Peptide Vaccination in Patients With Newly Diagnosed Glioblastoma. J Clin Oncol 2010, 28:4722-4729.
  • [37]Kahlon KS, et al.: Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res 2004, 64:9160-9166.
  • [38]Kong S, et al.: Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells. Clin Cancer Res 2012, 18:5949-5960.
  • [39]Ahmed N, et al.: HER2-Specific T Cells Target Primary Glioblastoma Stem Cells and Induce Regression of Autologous Experimental Tumors. Clin Cancer Res 2010, 16:474-485.
  • [40]Chow KK, et al.: T Cells Redirected to EphA2 for the Immunotherapy of Glioblastoma. Mol Ther 2012, 21:629-637.
  • [41]Schulte A, et al.: Glioblastoma stem-like cell lines with either maintenance or loss of high-level EGFR amplification, generated via modulation of ligand concentration. Clin Cancer Res 2012, 18:1901-1913.
  • [42]Zhang H, et al.: 4-1BB is superior to CD28 costimulation for generating CD8+ cytotoxic lymphocytes for adoptive immunotherapy. J Immunol 2007, 179:4910-4918.
  • [43]Litterman AJ, et al.: Profound impairment of adaptive immune responses by alkylating chemotherapy. J Immunol 2013, 190:6259-6268.
  • [44]Sanchez-Perez LA, et al.: Myeloablative temozolomide enhances CD8(+) T-cell responses to vaccine and is required for efficacy against brain tumors in mice. PLoS One 2013, 8:e59082.
  • [45]Hammond LA, et al.: Phase I and pharmacokinetic study of temozolomide on a daily-for-5-days schedule in patients with advanced solid malignancies. J Clin Oncol 1999, 17:2604-2613.
  • [46]Sasaki K, et al.: 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 2007, 67:6451-6458.
  • [47]Zhu X, et al.: 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 2007, 5:10. BioMed Central Full Text
  • [48]Okamoto S, et al.: Monoclonal antibody against the fusion junction of a deletion-mutant epidermal growth factor receptor. Br J Cancer 1996, 73:1366-1372.
  • [49]Nakayashiki N, et al.: Production of a single-chain variable fragment antibody recognizing type III mutant epidermal growth factor receptor. Jpn J Cancer Res 2000, 91:1035-1043.
  文献评价指标  
  下载次数:47次 浏览次数:36次