Clinical and Translational Medicine (Feb 2021)

Dissecting the role of TP53 alterations in del(11q) chronic lymphocytic leukemia

  • Miguel Quijada‐Álamo,
  • Claudia Pérez‐Carretero,
  • María Hernández‐Sánchez,
  • Ana‐Eugenia Rodríguez‐Vicente,
  • Ana‐Belén Herrero,
  • Jesús‐María Hernández‐Sánchez,
  • Marta Martín‐Izquierdo,
  • Sandra Santos‐Mínguez,
  • Mónica del Rey,
  • Teresa González,
  • Araceli Rubio‐Martínez,
  • Alfonso García de Coca,
  • Julio Dávila‐Valls,
  • José‐Ángel Hernández‐Rivas,
  • Helen Parker,
  • Jonathan C. Strefford,
  • Rocío Benito,
  • José‐Luis Ordóñez,
  • Jesús‐María Hernández‐Rivas

DOI
https://doi.org/10.1002/ctm2.304
Journal volume & issue
Vol. 11, no. 2
pp. n/a – n/a

Abstract

Read online

Abstract Background Several genetic alterations have been identified as driver events in chronic lymphocytic leukemia (CLL) pathogenesis and oncogenic evolution. Concurrent driver alterations usually coexist within the same tumoral clone, but how the cooperation of multiple genomic abnormalities contributes to disease progression remains poorly understood. Specifically, the biological and clinical consequences of concurrent high‐risk alterations such as del(11q)/ATM‐mutations and del(17p)/TP53‐mutations have not been established. Methods We integrated next‐generation sequencing (NGS) and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 techniques to characterize the in vitro and in vivo effects of concurrent monoallelic or biallelic ATM and/or TP53 alterations in CLL prognosis, clonal evolution, and therapy response. Results Targeted sequencing analysis of the co‐occurrence of high‐risk alterations in 271 CLLs revealed that biallelic inactivation of both ATM and TP53 was mutually exclusive, whereas monoallelic del(11q) and TP53 alterations significantly co‐occurred in a subset of CLL patients with a highly adverse clinical outcome. We determined the biological effects of combined del(11q), ATM and/or TP53 mutations in CRISPR/Cas9‐edited CLL cell lines. Our results showed that the combination of monoallelic del(11q) and TP53 mutations in CLL cells led to a clonal advantage in vitro and in in vivo clonal competition experiments, whereas CLL cells harboring biallelic ATM and TP53 loss failed to compete in in vivo xenotransplants. Furthermore, we demonstrated that CLL cell lines harboring del(11q) and TP53 mutations show only partial responses to B cell receptor signaling inhibitors, but may potentially benefit from ATR inhibition. Conclusions Our work highlights that combined monoallelic del(11q) and TP53 alterations coordinately contribute to clonal advantage and shorter overall survival in CLL.

Keywords