Blood Advances (Oct 2019)

Pediatric ALL relapses after allo-SCT show high individuality, clonal dynamics, selective pressure, and druggable targets

  • Jessica I. Hoell,
  • Sebastian Ginzel,
  • Michaela Kuhlen,
  • Andreas Kloetgen,
  • Michael Gombert,
  • Ute Fischer,
  • Daniel Hein,
  • Salih Demir,
  • Martin Stanulla,
  • Martin Schrappe,
  • Udo zur Stadt,
  • Peter Bader,
  • Florian Babor,
  • Friedhelm Schuster,
  • Brigitte Strahm,
  • Julia Alten,
  • Anja Moericke,
  • Gabriele Escherich,
  • Arend von Stackelberg,
  • Ralf Thiele,
  • Alice C. McHardy,
  • Christina Peters,
  • Beat Bornhauser,
  • Jean-Pierre Bourquin,
  • Stefan Krause,
  • Juergen Enczmann,
  • Lüder Hinrich Meyer,
  • Cornelia Eckert,
  • Arndt Borkhardt,
  • Roland Meisel

Journal volume & issue
Vol. 3, no. 20
pp. 3143 – 3156

Abstract

Read online

Abstract: Survival of patients with pediatric acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem cell transplantation (allo-SCT) is mainly compromised by leukemia relapse, carrying dismal prognosis. As novel individualized therapeutic approaches are urgently needed, we performed whole-exome sequencing of leukemic blasts of 10 children with post–allo-SCT relapses with the aim of thoroughly characterizing the mutational landscape and identifying druggable mutations. We found that post–allo-SCT ALL relapses display highly diverse and mostly patient-individual genetic lesions. Moreover, mutational cluster analysis showed substantial clonal dynamics during leukemia progression from initial diagnosis to relapse after allo-SCT. Only very few alterations stayed constant over time. This dynamic clonality was exemplified by the detection of thiopurine resistance-mediating mutations in the nucleotidase NT5C2 in 3 patients' first relapses, which disappeared in the post–allo-SCT relapses on relief of selective pressure of maintenance chemotherapy. Moreover, we identified TP53 mutations in 4 of 10 patients after allo-SCT, reflecting acquired chemoresistance associated with selective pressure of prior antineoplastic treatment. Finally, in 9 of 10 children's post–allo-SCT relapse, we found alterations in genes for which targeted therapies with novel agents are readily available. We could show efficient targeting of leukemic blasts by APR-246 in 2 patients carrying TP53 mutations. Our findings shed light on the genetic basis of post–allo-SCT relapse and may pave the way for unraveling novel therapeutic strategies in this challenging situation.