Journal for ImmunoTherapy of Cancer (Feb 2023)

Immunotherapy reverses glioma-driven dysfunction of immune system homeostasis

  • Bayli DiVita Dean,
  • Tyler Wildes,
  • Joseph Dean,
  • Oleg Yegorov,
  • Changlin Yang,
  • David Shin,
  • Connor Francis,
  • John W Figg,
  • Mathew Sebastian,
  • Laura Falceto Font,
  • Dan Jin,
  • Alexandra Reid,
  • Ginger Moore,
  • Brandon Fernandez,
  • Brandon Wummer,
  • Carmelle Kuizon,
  • Duane Mitchell,
  • Catherine T Flores

DOI
https://doi.org/10.1136/jitc-2022-004805
Journal volume & issue
Vol. 11, no. 2

Abstract

Read online

Background Glioma-induced immune dysregulation of the hematopoietic system has been described in a limited number of studies. In this study, our group further demonstrates that gliomas interrupt the cellular differentiation programming and outcomes of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. HSPCs from glioma-bearing mice are reprogrammed and driven towards expansion of myeloid lineage precursors and myeloid-derived suppressor cells (MDSCs) in secondary lymphoid organs. However, we found this expansion is reversed by immunotherapy. Adoptive cellular therapy (ACT) has been demonstrably efficacious in multiple preclinical models of central nervous system (CNS) malignancies, and here we describe how glioma-induced dysfunction is reversed by this immunotherapeutic platform.Methods The impact of orthotopic KR158B-luc glioma on HSPCs was evaluated in an unbiased fashion using single cell RNAseq (scRNAseq) of lineage− cells and phenotypically using flow cytometry. Mature myeloid cell frequencies and function were also evaluated using flow cytometry. Finally, ACT containing total body irradiation, tumor RNA-pulsed dendritic cells, tumor-reactive T cells and HSPCs isolated from glioma-bearing or non-tumor-bearing mice were used to evaluate cell fate differentiation and survival.Results Using scRNAseq, we observed an altered HSPC landscape in glioma-bearing versus non-tumor-bearing mice . In addition, an expansion of myeloid lineage subsets, including granulocyte macrophage precursors (GMPs) and MDSCs, were observed in glioma-bearing mice relative to non-tumor-bearing controls. Furthermore, MDSCs from glioma-bearing mice demonstrated increased suppressive capacity toward tumor-specific T cells as compared with MDSCs from non-tumor-bearing hosts. Interestingly, treatment with ACT overcame these suppressive properties. When HSPCs from glioma-bearing mice were transferred in the context of ACT, we observed significant survival benefit and long-term cures in orthotopic glioma models compared with mice treated with ACT using non-glioma-bearing HSPCs.