Abstract

Classical approaches to immunotherapy that show promise in some malignancies have generally been disappointing when applied to high-grade brain tumors such as glioblastoma multiforme (GBM). We recently showed that ex vivo expanded/activated γδ T cells recognize NKG2D ligands expressed on malignant glioma and are cytotoxic to glioma cell lines and primary GBM explants. In addition, γδ T cells extend survival and slow tumor progression when administered to immunodeficient mice with intracranial human glioma xenografts. We now show that temozolomide (TMZ), a principal chemotherapeutic agent used to treat GBM, increases the expression of stress-associated NKG2D ligands on TMZ-resistant glioma cells, potentially rendering them vulnerable to γδ T cell recognition and lysis. TMZ is also highly toxic to γδ T cells, however, and to overcome this cytotoxic effect γδ T cells were genetically modified using a lentiviral vector encoding the DNA repair enzyme O(6)-alkylguanine DNA alkyltransferase (AGT) from the O(6)-methylguanine methyltransferase (MGMT) cDNA, which confers resistance to TMZ. Genetic modification of γδ T cells did not alter their phenotype or their cytotoxicity against GBM target cells. Importantly, gene modified γδ T cells showed greater cytotoxicity to two TMZ resistant GBM cell lines, U373TMZ-R and SNB-19TMZ-R cells, in the presence of TMZ than unmodified cells, suggesting that TMZ exposed more receptors for γδ T cell-targeted lysis. Therefore, TMZ resistant γδ T cells can be generated without impairing their anti-tumor functions in the presence of high concentrations of TMZ. These results provide a mechanistic basis for combining chemotherapy and γδ T cell-based drug resistant cellular immunotherapy to treat GBM.

Highlights

  • Treatment strategies for high-grade primary brain tumors such as glioblastoma multiforme (GBM) have failed to significantly and consistently extended survival despite 50 years of advances in radiotherapy, chemotherapy, and surgical techniques [1]

  • These results indicate that the increase in NKG2D ligand expression in response to TMZ could increase the vulnerability of glioma cells to recognition and lysis by cd T cells within the first 4 to 6 hours following TMZ-based chemotherapy

  • Generation of TMZ-resistant cd T cells To produce expanded/activated cd T cells that retain function when exposed to high concentrations of TMZ chemotherapy, vectors were generated that confer TMZ-resistance based on enforced expression of AGT

Read more

Summary

Introduction

Treatment strategies for high-grade primary brain tumors such as glioblastoma multiforme (GBM) have failed to significantly and consistently extended survival despite 50 years of advances in radiotherapy, chemotherapy, and surgical techniques [1]. The most predominant cell therapy consisted of autologous lymphokine-activated killer (LAK) cells, a combination of NK and T lymphocytes cultured in high doses of IL-2. CTL therapies are based on adaptive immunity (i.e. MHC-restricted, antigen-specific responses) and are dependent upon the dose of T cell clones that recognize various tumor-associated peptide antigens dispersed among various subsets of glioma cells. Infusion or intracranial placement of HLA-mismatched CTL relies on allogeneic recognition of transplantation antigens and is highly dependent on glioma cell MHC Class I expression [10,11]. LAK cell preparations are difficult to consistently manufacture, are short-lived in vivo [12], and are complicated by IL-2 related toxicity once infused or placed in the tumor resection cavity [2,13,14,15,16]

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