Control of acute myeloid leukemia by a trifunctional NKp46-CD16a-NK cell engager targeting CD123

Nat Biotechnol. 2023 Sep;41(9):1296-1306. doi: 10.1038/s41587-022-01626-2. Epub 2023 Jan 12.

Abstract

CD123, the alpha chain of the IL-3 receptor, is an attractive target for acute myeloid leukemia (AML) treatment. However, cytotoxic antibodies or T cell engagers targeting CD123 had insufficient efficacy or safety in clinical trials. We show that expression of CD64, the high-affinity receptor for human IgG, on AML blasts confers resistance to anti-CD123 antibody-dependent cell cytotoxicity (ADCC) in vitro. We engineer a trifunctional natural killer cell engager (NKCE) that targets CD123 on AML blasts and NKp46 and CD16a on NK cells (CD123-NKCE). CD123-NKCE has potent antitumor activity against primary AML blasts regardless of CD64 expression and induces NK cell activation and cytokine secretion only in the presence of AML cells. Its antitumor activity in a mouse CD123+ tumor model exceeds that of the benchmark ADCC-enhanced antibody. In nonhuman primates, it had prolonged pharmacodynamic effects, depleting CD123+ cells for more than 10 days with no signs of toxicity and very low inflammatory cytokine induction over a large dose range. These results support clinical development of CD123-NKCE.

MeSH terms

  • Animals
  • Antibody-Dependent Cell Cytotoxicity
  • Cytokines / metabolism
  • Humans
  • Interleukin-3 Receptor alpha Subunit
  • Killer Cells, Natural
  • Leukemia, Myeloid, Acute* / drug therapy
  • Leukemia, Myeloid, Acute* / metabolism
  • Mice
  • T-Lymphocytes

Substances

  • Cytokines
  • Interleukin-3 Receptor alpha Subunit