Cancer stem cells reside in hypoxic areas and contribute to tumor progression. These cells are particularly difficult to treat because they are inherently resistant to conventional therapy, and their microenvironment reduces the efficacy of radiation and impairs delivery of chemotherapy. Glioblastoma is an incurable primary brain tumor that is characterized by regions of hypoxia. Notch signaling maintains GSCs within the hypoxic niche, but how Notch is dysregulated is unclear. Our preliminary data strongly suggest that hypoxia can regulate membranous Notch levels, thereby regulating receptor availability for ligand binding. We have found that the recently identified hypoxia-regulated protein, Vasorin, is preferentially expressed in GSCs to regulate Notch signaling. Our data suggest that Vasorin regulates GSC self-renewal by inhibiting the lysosomal degradation of Notch1. Thus, we have identified a novel mechanism by which hypoxia directs Notch signaling: regulation of receptor turnover. Our central hypothesis is that Vasorin regulates GSC self-renewal within the hypoxic niche by regulating Notch signaling and that targeting Vasorin may improve GBM therapy. We will test our hypothesis through mechanistic and preclinical studies.
In Aim 1, we will determine the role of Vasorin in mediating GSC properties and tumorigenic potential in vitro and in vivo.
In Aim 2, we will define the role of Vasorin in regulating Notch signaling to promote GSC self-renewal under hypoxic conditions.
In Aim 3, we will assess Vasorin as a prognostic biomarker and provide proof-of-principle that maximizing Notch inhibition by dual targeting of Vasorin and Notch proteolytic processing can improve survival in mouse models of glioblastoma. Importantly, Vasorin does not appear to be expressed in normal embryonic or adult brain. Therefore, targeting Vasorin may have limited normal brain toxicity. These data will reveal an important role for Vasorin in regulating Notch signaling and have significant clinical ramifications. Current therapies using hypoxia modifiers or Notch pathway inhibitors, such as ?-secretase inhibitors, are limited by side effects. If successful, the findings of this study will provide a strong rationale for the development of novel therapeutics against Vasorin.

Public Health Relevance

Cancer stem cells reside in areas of hypoxia to contribute to tumor progression. These cells are particularly difficult to treat because they are inherently resistant to conventional therapy. In addition, their microenvironment reduces the efficacy of radiation and impairs delivery of chemotherapy. If successful, the findings of this study could pave the way for the development of new therapies that can kill off the cancer stem cells within the hypoxic niche.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS094199-05
Application #
9752672
Study Section
Tumor Microenvironment Study Section (TME)
Program Officer
Fountain, Jane W
Project Start
2015-09-30
Project End
2020-07-31
Budget Start
2019-08-01
Budget End
2020-07-31
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Cleveland Clinic Lerner
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
135781701
City
Cleveland
State
OH
Country
United States
Zip Code
44195
Hao, Jing; Yu, Jennifer S (2018) Semaphorin 3C and Its Receptors in Cancer and Cancer Stem-Like Cells. Biomedicines 6:
Man, Jianghong; Yu, Xingjiang; Huang, Haidong et al. (2018) Hypoxic Induction of Vasorin Regulates Notch1 Turnover to Maintain Glioma Stem-like Cells. Cell Stem Cell 22:104-118.e6
Zhou, Wenchao; Chen, Cong; Shi, Yu et al. (2017) Targeting Glioma Stem Cell-Derived Pericytes Disrupts the Blood-Tumor Barrier and Improves Chemotherapeutic Efficacy. Cell Stem Cell 21:591-603.e4