Disease evolution and outcomes in familial AML with germline CEBPA mutations

Research output: Contribution to journalJournal articleResearchpeer-review

  • Kiran Tawana
  • Jun Wang
  • Aline Renneville
  • Csaba Bödör
  • Robert Hills
  • Chey Loveday
  • Aleksandar Savic
  • Frederik W Van Delft
  • Jennifer Treleaven
  • Panayiotis Georgiades
  • Elizabeth Uglow
  • Norio Asou
  • Naokuni Uike
  • Maruša Debeljak
  • Janez Jazbec
  • Philip Ancliff
  • Rosemary Gale
  • Xavier Thomas
  • Valerie Mialou
  • Konstanze Döhner
  • Lars Bullinger
  • Beatrice Mueller
  • Thomas Pabst
  • Matthias Stelljes
  • Brigitte Schlegelberger
  • Eva Wozniak
  • Sameena Iqbal
  • Jessica Okosun
  • Shamzah Araf
  • Felicia B Lauridsen
  • Claus Nerlov
  • Carolyn Owen
  • Inderjeet Dokal
  • John Gribben
  • Matthew Smith
  • Claude Preudhomme
  • Claude Chelala
  • Jamie Cavenagh
  • Jude Fitzgibbon

In-depth molecular investigation of familial leukemia has been limited by the rarity of recognized cases. This study examines the genetic events initiating leukemia and details the clinical progression of disease across multiple families harboring germ-line CEBPA mutations. Clinical data were collected from 10 CEBPA-mutated families, representing 24 members with acute myeloid leukemia (AML). Whole-exome (WES) and deep sequencing were performed to genetically profile tumors and define patterns of clonal evolution. Germline CEBPA mutations clustered within the N-terminal and were highly penetrant, with AML presenting at a median age of 24.5 years (range, 1.75-46 years). In all diagnostic tumors tested (n = 18), double CEBPA mutations (CEBPAdm) were detected, with acquired (somatic) mutations preferentially targeting the C-terminal. Somatic CEBPA mutations were unstable throughout the disease course, with different mutations identified at recurrence. Deep sequencing of diagnostic and relapse paired samples confirmed that relapse-associated CEBPA mutations were absent at diagnosis, suggesting recurrence was triggered by novel, independent clones. Integrated WES and deep sequencing subsequently revealed an entirely new complement of mutations at relapse, verifying the presentation of a de novo leukemic episode. The cumulative incidence of relapse in familial AML was 56% at 10 years (n = 11), and 3 patients experienced ≥3 disease episodes over a period of 17 to 20 years. Durable responses to secondary therapies were observed, with prolonged median survival after relapse (8 years) and long-term overall survival (10-year overall survival, 67%). Our data reveal that familial CEBPA-mutated AML exhibits a unique model of disease progression, associated with favorable long-term outcomes.

Original languageEnglish
JournalBlood
Volume126
Issue number10
Pages (from-to)1214-23
Number of pages10
ISSN0006-4971
DOIs
Publication statusPublished - 3 Sep 2015

ID: 160407329