Abstract

Tissue hypoxia contributes to solid tumor pathogenesis by activating a series of adaptive programs. We previously showed that hypoxia promotes the preferential expansion and maintenance of CD133 positive human glioma stem cells (GSC) in a hypoxia inducible factor 1 alpha (HIF-1α)-dependent mechanism. Here, we examined the activity of digitoxin (DT), a cardiac glycoside and a putative inhibitor of HIF-1α, on human GSC in vitro and in vivo. During hypoxic conditions (1% O2), we observed the effect of DT on the intracellular level of HIF-1α and the extracellular level of vascular endothelial growth factor (VEGF) in human GSC. We found that DT at clinically achievable concentrations, suppressed HIF-1α accumulation during hypoxic conditions in human GSC and established glioma cell lines. DT treatment also significantly attenuated hypoxia-induced expression of VEGF, a downstream target of HIF-1α. Exposure to DT also reduced hypoxia-induced activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. Furthermore, DT potently inhibited neurosphere formation, and decreased CD133 expression even at concentrations that were not overtly cytotoxic. Lastly, treatment with DT reduced GSC engraftment in an in vivo xenograft model of glioblastoma. Intraperitoneal injections of DT significantly inhibited the growth of established glioblastoma xenografts, and suppressed expression of HIF-1α and carbonic anhydrase (CA9), a surrogate marker of hypoxia. Taken together, these results suggest that DT at clinically achievable concentration functions as an inhibitor of HIF-1α, worthy of further investigations in the therapy of glioblastoma.

Highlights

  • Therapeutic targeting of the cancer stem cell subpopulation has potential for more durable treatment of cancer

  • We previously showed that hypoxia promotes the preferential expansion and maintenance of CD133 positive human glioma stem cells (GSC) in a hypoxia inducible factor 1 alpha (HIF-1α)-dependent mechanism

  • During hypoxic conditions (1% O2), we observed the effect of DT on the intracellular level of HIF-1α and the extracellular level of vascular endothelial growth factor (VEGF) in human GSC

Read more

Summary

Introduction

Therapeutic targeting of the cancer stem cell subpopulation has potential for more durable treatment of cancer. Relatively few specific targets have been identified. HIF was recently shown to regulate the tumorigenic capacity of glioma stem cells (GSC) in hypoxic conditions [1,2,3]. A suggested reason for the highly refractory nature of GB is the presence of GSC, a subpopulation of tumor cells regarded as the source of tumor initiation and relapse. GSC may represent the tumor-initiating cells of GB, and are believed to contribute to the resistance to conventional therapies. Development of novel chemotherapeutic agents and treatment approaches against GB, those that target GSC, are likely to result in more effective and durable response. GSC are enriched in the perivascular niche and areas near necrosis [5], which in turn are associated with reduced oxygen tension, or hypoxia

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call