Malignant gliomas are aggressive tumors that often recur within the resection margin after treatment. In addition to the development of targeted therapies against neoplastic cells, treatments aimed at tumor stroma are being considered to enhance conventional therapies. Tumor-associated inflammatory cells, such as macrophages and neutrophils, comprise a significant component of the glioma stroma and actively participate in angiogenesis, invasion and metastasis. These myeloid-derived cells express pattern recognition receptors such as the receptor for advanced glycation endproducts (RAGE) that constantly monitor the tumor micro- environment (TME). Engagement of RAGE by its ligands results in activation of multiple downstream pathways that regulate cell proliferation, survival, differentiation, migration, phagocytosis and autophagy. During the previous funding cycle, we demonstrated that upregulation of a common glioma RAGE ligand, S100B, promoted macrophage recruitment and altered their conversion into tumor-promoting cells. Furthermore, we showed that genetic ablation of RAGE in TME prolonged survival of glioma-bearing mice by attenuating tumor- associated inflammation and angiogenesis. These studies also revealed significant variability in the expression of other RAGE ligands in animal glioma models. The objective of this competing renewal is to evaluate the role of the RAGE pathway on TME remodeling and tumor progression in gliomas. Our central hypothesis is that gliomas release RAGE ligands that contribute to the polarization of inflammatory cells, and promote tumor growth and invasion. To test this, we propose the following experiments.
In Aim 1 we will measure RAGE ligands in human glioma tumor samples in order to determine their physiological concentrations in the TME.
Aim 2 will characterize changes in tumor inflammation after inhibition of RAGE ligands. Finally in Aim 3, we will determine the effect of RAGE activation on glioma progression and optimize the antitumor activity of targeting the RAGE axis. These studies will provide the first insights into the effect of the RAGE pathway and surgical trauma on glioma recurrence. This critically needed understanding of the mechanism of immune evasion in gliomas will be valuable in optimizing antiglioma therapies.

Public Health Relevance

Our goal is to evaluate the role of the receptor for advanced glycation endproducts (RAGE) on glioblastoma progression. Our data suggest that proteins that activate RAGE alter the activity of inflammatory cells thereby promoting tumor growth and invasion. By blocking the interaction of RAGE with these proteins, we hope to improve treatment responses for glioblastoma, which currently has no cure.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA155769-06A1
Application #
9312100
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Woodhouse, Elizabeth
Project Start
2011-09-07
Project End
2022-03-31
Budget Start
2017-04-01
Budget End
2018-03-31
Support Year
6
Fiscal Year
2017
Total Cost
$410,877
Indirect Cost
$173,376
Name
Beckman Research Institute/City of Hope
Department
Type
Research Institutes
DUNS #
027176833
City
Duarte
State
CA
Country
United States
Zip Code
91010
Gao, Hang; Zhang, Ian Y; Zhang, Leying et al. (2018) S100B suppression alters polarization of infiltrating myeloid-derived cells in gliomas and inhibits tumor growth. Cancer Lett 439:91-100
Keu, Khun Visith; Witney, Timothy H; Yaghoubi, Shahriar et al. (2017) Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Sci Transl Med 9:
Mirzaei, Hamid R; Rodriguez, Analiz; Shepphird, Jennifer et al. (2017) Chimeric Antigen Receptors T Cell Therapy in Solid Tumor: Challenges and Clinical Applications. Front Immunol 8:1850
Ouyang, Mao; White, Ethan E; Ren, Hui et al. (2016) Metronomic Doses of Temozolomide Enhance the Efficacy of Carbon Nanotube CpG Immunotherapy in an Invasive Glioma Model. PLoS One 11:e0148139
Brown, Christine E; Alizadeh, Darya; Starr, Renate et al. (2016) Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med 375:2561-9
White, Ethan E; Pai, Alex; Weng, Yiming et al. (2015) Functionalized iron oxide nanoparticles for controlling the movement of immune cells. Nanoscale 7:7780-9
Brown, Christine E; Badie, Behnam; Barish, Michael E et al. (2015) Bioactivity and Safety of IL13R?2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin Cancer Res 21:4062-72
Carvalho da Fonseca, Anna Carolina; Wang, Huaqing; Fan, Haitao et al. (2014) Increased expression of stress inducible protein 1 in glioma-associated microglia/macrophages. J Neuroimmunol 274:71-7
Herrmann, Andreas; Cherryholmes, Gregory; Schroeder, Anne et al. (2014) TLR9 is critical for glioma stem cell maintenance and targeting. Cancer Res 74:5218-28
Chen, Xuebo; Zhang, Leying; Zhang, Ian Y et al. (2014) RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res 74:7285-7297

Showing the most recent 10 out of 12 publications