Abstract

The highly invasive property of glioblastoma (GBM) cells and genetic heterogeneity are largely responsible for tumor recurrence after the current standard‐of‐care treatment and thus a direct cause of death. Previously, we have shown that intracranial interferon‐beta (IFN‐β) gene therapy by locally administered adeno‐associated viral vectors (AAV) successfully treats noninvasive orthotopic glioblastoma models. Here, we extend these findings by testing this approach in invasive human GBM xenograft and syngeneic mouse models. First, we show that a single intracranial injection of AAV encoding human IFN‐β eliminates invasive human GBM8 tumors and promotes long‐term survival. Next, we screened five AAV‐IFN‐β vectors with different promoters to drive safe expression of mouse IFN‐β in the brain in the context of syngeneic GL261 tumors. Two AAV‐IFN‐β vectors were excluded due to safety concerns, but therapeutic studies with the other three vectors showed extensive tumor cell death, activation of microglia surrounding the tumors, and a 56% increase in median survival of the animals treated with AAV/P2‐Int‐mIFN‐β vector. We also assessed the therapeutic effect of combining AAV‐IFN‐β therapy with temozolomide (TMZ). As TMZ affects DNA replication, an event that is crucial for second‐strand DNA synthesis of single‐stranded AAV vectors before active transcription, we tested two TMZ treatment regimens. Treatment with TMZ prior to AAV‐IFN‐β abrogated any benefit from the latter, while the reverse order of treatment doubled the median survival compared to controls. These studies demonstrate the therapeutic potential of intracranial AAV‐IFN‐β therapy in a highly migratory GBM model as well as in a syngeneic mouse model and that combination with TMZ is likely to enhance its antitumor potency.

Highlights

  • Glioblastoma is the most common primary brain tumor (Preusser et al, 2011) as well as the most aggressive form (WHO grade IV) of glial cell tumors (Louis et al, 2007)

  • We tested the therapeutic efficacy of intracranial administration of 3 9 1010 gc AAVrh8/CBA-human IFN-b (hIFN-b) in GBM8 tumor-bearing mice at 2, 3, or 4 weeks after tumor implantation (Fig. 1B)

  • Several studies from our group (Maguire et al, 2008; Meijer et al, 2009) and others (Denbo et al, 2011; Streck et al, 2006) have demonstrated the potential of IFN-b gene therapy for glioblastoma. None of these studies has tested this approach in an orthotopic glioblastoma model that is truly invasive in nature

Read more

Summary

Introduction

Glioblastoma is the most common primary brain tumor (Preusser et al, 2011) as well as the most aggressive form (WHO grade IV) of glial cell tumors (Louis et al, 2007). The current standard-of-care treatment involves surgical resection, followed by radioand chemotherapy with temozolomide (TMZ). One of the primary causes for recurrence is the inability to surgically remove all tumor cells due to their ability to infiltrate normal tissue and migrate long distances. GBM tumors develop exceptional resistance to radiation and temozolomide. As a result of these insidious properties of GBM, the median survival of patients with the current standard-of-care treatment is only 15–17 months from the time of diagnosis (Preusser et al, 2015; Stupp et al, 2009). Development of new therapies for GBM remains a high priority

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.