Haematologica (Jun 2024)

Genome-scale clustered regularly interspaced short palindromic repeats screen identifies nucleotide metabolism as an actionable therapeutic vulnerability in diffuse large B-cell lymphoma

  • Nicholas Davies,
  • Tegan Francis,
  • Ceri Oldreive,
  • Maria Azam,
  • Jordan Wilson,
  • Philip J. Byrd,
  • Megan Burley,
  • Archana Sharma-Oates,
  • Peter Keane,
  • Sael Alatawi,
  • Martin R. Higgs,
  • Zbigniew Rudzki,
  • Maha Ibrahim,
  • Tracey Perry,
  • Angelo Agathaggelou,
  • Anne-Marie Hewitt,
  • Edward Smith,
  • Constanze Bonifer,
  • Mark O’Connor,
  • Josep V. Forment,
  • Paul G. Murray,
  • Eanna Fennell,
  • Gemma Kelly,
  • Catherine Chang,
  • Grant S. Stewart,
  • Tatjana Stankovic,
  • Marwan Kwok,
  • Alexander Malcolm Taylor

DOI
https://doi.org/10.3324/haematol.2023.284404
Journal volume & issue
Vol. 999, no. 1

Abstract

Read online

Diffuse large B-cell lymphoma (DLBCL) is the most common malignancy that develops in patients with ataxia-telangiectasia, a cancer-predisposing inherited syndrome characterized by inactivating germline ATM mutations. ATM is also frequently mutated in sporadic DLBCL. To investigate lymphomagenic mechanisms and lymphoma-specific dependencies underlying defective ATM, we applied ribonucleic acid (RNA)-seq and genome-scale loss-offunction clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens to systematically interrogate B-cell lymphomas arising in a novel murine model (Atm-/-nu-/-) with constitutional Atm loss, thymic aplasia but residual T-cell populations. Atm-/-nu-/-lymphomas, which phenotypically resemble either activated B-cell-like or germinal center Bcell-like DLBCL, harbor a complex karyotype, and are characterized by MYC pathway activation. In Atm-/-nu-/-lymphomas, we discovered nucleotide biosynthesis as a MYCdependent cellular vulnerability that can be targeted through the synergistic nucleotidedepleting actions of mycophenolate mofetil (MMF) and the WEE1 inhibitor, adavosertib (AZD1775). The latter is mediated through a synthetically lethal interaction between RRM2 suppression and MYC dysregulation that results in replication stress overload in Atm-/-nu-/-lymphoma cells. Validation in cell line models of human DLBCL confirmed the broad applicability of nucleotide depletion as a therapeutic strategy for MYC-driven DLBCL independent of ATM mutation status. Our findings extend current understanding of lymphomagenic mechanisms underpinning ATM loss and highlight nucleotide metabolism as a targetable therapeutic vulnerability in MYC-driven DLBCL.