Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy

Research output: Contribution to journalJournal articleResearchpeer-review

  • Zinal S Chheda
  • Gary Kohanbash
  • Kaori Okada
  • Naznin Jahan
  • John Sidney
  • Matteo Pecoraro
  • Xinbo Yang
  • Diego A Carrera
  • Kira M Downey
  • Shruti Shrivastav
  • Shuming Liu
  • Yi Lin
  • Chetana Lagisetti
  • Pavlina Chuntova
  • Payal B Watchmaker
  • Sabine Mueller
  • Ian F Pollack
  • Raja Rajalingam
  • Angel M Carcaboso
  • Alessandro Sette
  • K Christopher Garcia
  • Yafei Hou
  • Hideho Okada

The median overall survival for children with diffuse intrinsic pontine glioma (DIPG) is less than one year. The majority of diffuse midline gliomas, including more than 70% of DIPGs, harbor an amino acid substitution from lysine (K) to methionine (M) at position 27 of histone 3 variant 3 (H3.3). From a CD8+ T cell clone established by stimulation of HLA-A2+ CD8+ T cells with synthetic peptide encompassing the H3.3K27M mutation, complementary DNA for T cell receptor (TCR) α- and β-chains were cloned into a retroviral vector. TCR-transduced HLA-A2+ T cells efficiently killed HLA-A2+H3.3K27M+ glioma cells in an antigen- and HLA-specific manner. Adoptive transfer of TCR-transduced T cells significantly suppressed the progression of glioma xenografts in mice. Alanine-scanning assays suggested the absence of known human proteins sharing the key amino acid residues required for recognition by the TCR, suggesting that the TCR could be safely used in patients. These data provide us with a strong basis for developing T cell-based therapy targeting this shared neoepitope.

Original languageEnglish
JournalThe Journal of Experimental Medicine
Volume215
Issue number1
Pages (from-to)141-157
Number of pages17
ISSN0022-1007
DOIs
Publication statusPublished - Jan 2018
Externally publishedYes

    Research areas

  • Journal Article

ID: 186869209