Blood Cancer Journal (Jun 2023)

Integrated multi-omics analyses reveal homology-directed repair pathway as a unique dependency in near-haploid leukemia

  • Yunpeng Liu-Lupo,
  • James Dongjoo Ham,
  • Swarna K. A. Jeewajee,
  • Lan Nguyen,
  • Toni Delorey,
  • Azucena Ramos,
  • David M. Weinstock,
  • Aviv Regev,
  • Michael T. Hemann

DOI
https://doi.org/10.1038/s41408-023-00863-1
Journal volume & issue
Vol. 13, no. 1
pp. 1 – 14

Abstract

Read online

Abstract Whole chromosome losses resulting in near-haploid karyotypes are found in a rare subgroup of treatment-refractory acute lymphoblastic leukemia. To systematically dissect the unique physiology and uncover susceptibilities that can be exploited in near-haploid leukemia, we leveraged single-cell RNA-Seq and computational inference of cell cycle stages to pinpoint key differences between near-haploid and diploid leukemia cells. Combining cell cycle stage-specific differential expression with gene essentiality scores from a genome-wide CRISPR-Cas9-mediated knockout screen, we identified the homologous recombination pathway component RAD51B as an essential gene in near-haploid leukemia. DNA damage analyses revealed significantly increased sensitivity of RAD51-mediated repair to RAD51B loss in the G2/M stage of near-haploid cells, suggesting a unique role of RAD51B in the homologous recombination pathway. Elevated G2/M and G1/S checkpoint signaling was part of a RAD51B signature expression program in response to chemotherapy in a xenograft model of human near-haploid B-ALL, and RAD51B and its associated programs were overexpressed in a large panel of near-haploid B-ALL patients. These data highlight a unique genetic dependency on DNA repair machinery in near-haploid leukemia and demarcate RAD51B as a promising candidate for targeted therapy in this treatment-resistant disease.