Frontiers in Immunology (Dec 2023)

Antigen presentation deficiency, mesenchymal differentiation, and resistance to immunotherapy in the murine syngeneic CT2A tumor model

  • J. Bryan Iorgulescu,
  • J. Bryan Iorgulescu,
  • J. Bryan Iorgulescu,
  • Neil Ruthen,
  • Ryuhjin Ahn,
  • Ryuhjin Ahn,
  • Eleni Panagioti,
  • Prafulla C. Gokhale,
  • Martha Neagu,
  • Maria C. Speranza,
  • Benjamin K. Eschle,
  • Kara M. Soroko,
  • Raziye Piranlioglu,
  • Meenal Datta,
  • Meenal Datta,
  • Shanmugarajan Krishnan,
  • Kathleen B. Yates,
  • Kathleen B. Yates,
  • Gregory J. Baker,
  • Gregory J. Baker,
  • Rakesh K. Jain,
  • Mario L. Suvà,
  • Mario L. Suvà,
  • Mario L. Suvà,
  • Donna Neuberg,
  • Forest M. White,
  • Forest M. White,
  • E. Antonio Chiocca,
  • Gordon J. Freeman,
  • Arlene H. Sharpe,
  • Arlene H. Sharpe,
  • Arlene H. Sharpe,
  • Catherine J. Wu,
  • Catherine J. Wu,
  • David A. Reardon

DOI
https://doi.org/10.3389/fimmu.2023.1297932
Journal volume & issue
Vol. 14

Abstract

Read online

BackgroundThe GL261 and CT2A syngeneic tumor lines are frequently used as immunocompetent orthotopic mouse models of human glioblastoma (huGBM) but demonstrate distinct differences in their responses to immunotherapy.MethodsTo decipher the cell-intrinsic mechanisms that drive immunotherapy resistance in CT2A-luc and to define the aspects of human cancer biology that these lines can best model, we systematically compared their characteristics using whole exome and transcriptome sequencing, and protein analysis through immunohistochemistry, Western blot, flow cytometry, immunopeptidomics, and phosphopeptidomics.ResultsThe transcriptional profiles of GL261-luc2 and CT2A-luc tumors resembled those of some huGBMs, despite neither line sharing the essential genetic or histologic features of huGBM. Both models exhibited striking hypermutation, with clonal hotspot mutations in RAS genes (Kras p.G12C in GL261-luc2 and Nras p.Q61L in CT2A-luc). CT2A-luc distinctly displayed mesenchymal differentiation, upregulated angiogenesis, and multiple defects in antigen presentation machinery (e.g. Tap1 p.Y488C and Psmb8 p.A275P mutations) and interferon response pathways (e.g. copy number losses of loci including IFN genes and reduced phosphorylation of JAK/STAT pathway members). The defect in MHC class I expression could be overcome in CT2A-luc by interferon-γ treatment, which may underlie the modest efficacy of some immunotherapy combinations. Additionally, CT2A-luc demonstrated substantial baseline secretion of the CCL-2, CCL-5, and CCL-22 chemokines, which play important roles as myeloid chemoattractants.ConclusionAlthough the clinical contexts that can be modeled by GL261 and CT2A for huGBM are limited, CT2A may be an informative model of immunotherapy resistance due to its deficits in antigen presentation machinery and interferon response pathways.

Keywords