Nature Communications (Sep 2024)

CRISPR/Cas9 editing of NKG2A improves the efficacy of primary CD33-directed chimeric antigen receptor natural killer cells

  • Tobias Bexte,
  • Nawid Albinger,
  • Ahmad Al Ajami,
  • Philipp Wendel,
  • Leon Buchinger,
  • Alec Gessner,
  • Jamal Alzubi,
  • Vinzenz Särchen,
  • Meike Vogler,
  • Hadeer Mohamed Rasheed,
  • Beate Anahita Jung,
  • Sebastian Wolf,
  • Raj Bhayadia,
  • Thomas Oellerich,
  • Jan-Henning Klusmann,
  • Olaf Penack,
  • Nina Möker,
  • Toni Cathomen,
  • Michael A. Rieger,
  • Katharina Imkeller,
  • Evelyn Ullrich

DOI
https://doi.org/10.1038/s41467-024-52388-1
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 19

Abstract

Read online

Abstract Chimeric antigen receptor (CAR)-modified natural killer (NK) cells show antileukemic activity against acute myeloid leukemia (AML) in vivo. However, NK cell-mediated tumor killing is often impaired by the interaction between human leukocyte antigen (HLA)-E and the inhibitory receptor, NKG2A. Here, we describe a strategy that overcomes CAR-NK cell inhibition mediated by the HLA-E-NKG2A immune checkpoint. We generate CD33-specific, AML-targeted CAR-NK cells (CAR33) combined with CRISPR/Cas9-based gene disruption of the NKG2A-encoding KLRC1 gene. Using single-cell multi-omics analyses, we identified transcriptional features of activation and maturation in CAR33-KLRC1 ko-NK cells, which are preserved following exposure to AML cells. Moreover, CAR33-KLRC1 ko-NK cells demonstrate potent antileukemic killing activity against AML cell lines and primary blasts in vitro and in vivo. We thus conclude that NKG2A-deficient CAR-NK cells have the potential to bypass immune suppression in AML.