Impact of composite scaffold degradation rate on neural stem cell persistence in the glioblastoma surgical resection cavity

Mater Sci Eng C Mater Biol Appl. 2020 Jun:111:110846. doi: 10.1016/j.msec.2020.110846. Epub 2020 Mar 13.

Abstract

Tumoricidal neural stem cells (NSCs) are an emerging therapy to combat glioblastoma (GBM). This therapy employs genetically engineered NSCs that secrete tumoricidal agents to seek out and kill tumor foci remaining after GBM surgical resection. Biomaterial scaffolds have previously been utilized to deliver NSCs to the resection cavity. Here, we investigated the impact of scaffold degradation rate on NSC persistence in the brain resection cavity. Composite acetalated dextran (Ace-DEX) gelatin electrospun scaffolds were fabricated with two distinct degradation profiles created by changing the ratio of cyclic to acyclic acetal coverage of Ace-DEX. In vitro, fast degrading scaffolds were fully degraded by one week, whereas slow degrading scaffolds had a half-life of >56 days. The scaffolds also retained distinct degradation profiles in vivo. Two different NSC lines readily adhered to and remained viable on Ace-DEX gelatin scaffolds, in vitro. Therapeutic NSCs secreting tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) had the same TRAIL output as tissue culture treated polystyrene (TCPS) when seeded on both scaffolds. Furthermore, secreted TRAIL was found to be highly potent against the human derived GBM cell line, GBM8, in vitro. Firefly luciferase expressing NSCs were seeded on scaffolds, implanted in a surgical resection cavity and their persistence in the brain was monitored by bioluminescent imaging (BLI). NSC loaded scaffolds were compared to a direct injection (DI) of NSCs in suspension, which is the current clinical approach to NSC therapy for GBM. Fast and slow degrading scaffolds enhanced NSC implantation efficiency 2.87 and 3.08-fold over DI, respectively. Interestingly, scaffold degradation profile did not significantly impact NSC persistence. However, persistence and long-term survival of NSCs was significantly greater for both scaffolds compared to DI, with scaffold implanted NSCs still detected by BLI at day 120 in most mice. Overall, these results highlight the benefit of utilizing a scaffold for application of tumoricidal NSC therapy for GBM.

Keywords: Acetalated dextran; Electrospinning; Gelatin; TRAIL; Tumoricidal neural stem cell therapy.

MeSH terms

  • Acetylation
  • Animals
  • Brain Neoplasms / pathology*
  • Cell Line
  • Cell Survival
  • Cross-Linking Reagents / chemistry
  • Dextrans / chemistry
  • Female
  • Gelatin / chemistry
  • Glioblastoma / pathology*
  • Mice, Nude
  • Neural Stem Cells / pathology*
  • Temperature
  • Tissue Scaffolds / chemistry*

Substances

  • Cross-Linking Reagents
  • Dextrans
  • Gelatin