Haematologica (Oct 2020)

Plasmacytoid dendritic cells proliferation associated with acute myeloid leukemia: phenotype profile and mutation landscape

  • Loria Zalmaï,
  • Pierre-Julien Viailly,
  • Sabeha Biichle,
  • Meyling Cheok,
  • Lou Soret,
  • Fanny Angelot-Delettre,
  • Tony Petrella,
  • Marie-Agnès Collonge-Rame,
  • Estelle Seilles,
  • Sandrine Geffroy,
  • Eric Deconinck,
  • Etienne Daguindau,
  • Sabrina Bouyer,
  • Elodie Dindinaud,
  • Victor Baunin,
  • Magali Le Garff-Tavernier,
  • Damien Roos-Weil,
  • Orianne Wagner-Ballon,
  • Véronique Salaun,
  • Jean Feuillard,
  • Sophie Brun,
  • Bernard Drenou,
  • Caroline Mayeur-Rousse,
  • Patricia Okamba,
  • Véronique Dorvaux,
  • Michel Tichionni,
  • Johann Rose,
  • Marie Thérèse Rubio,
  • Marie Christine Jacob,
  • Victoria Raggueneau,
  • Claude Preudhomme,
  • Philippe Saas,
  • Christophe Ferrand,
  • Olivier Adotevi,
  • Christophe Roumier,
  • Fabrice Jardin,
  • Francine Garnache-Ottou,
  • Florian Renosi

DOI
https://doi.org/10.3324/haematol.2020.253740
Journal volume & issue
Vol. 106, no. 12

Abstract

Read online

Neoplasms involving plasmacytoid dendritic cells (pDC) include blastic pDC neoplasms (BPDCN) and other pDC proliferations, where pDC are associated with myeloid malignancies: most frequently chronic myelomonocytic leukemia (CMML) but also acute myeloid leukemia (AML), hereafter named pDC-AML. We aimed to determine the reactive or neoplastic origin of pDC in pDC-AML, and their link with the CD34+ blasts, monocytes or conventional DC (cDC) associated in the same sample, by phenotypic and molecular analyses (targeted next-generation sequencing, 70 genes). We compared 15 pDCAML at diagnosis with 21 BPDCN and 11 normal pDC from healthy donors. CD45low CD34+ blasts were found in all cases (10-80% of medullar cells), associated with pDC (4-36%), monocytes in 14 cases (1-10%) and cDC (two cases, 4.8-19%). pDC in pDC-AML harbor a clearly different phenotype from BPDCN: CD4+ CD56– in 100% of cases, most frequently CD303+, CD304+ and CD34+; lower expression of cTCL1 and CD123 with isolated lymphoid markers (CD22/CD7/CD5) in some cases, suggesting a prepDC stage. In all cases, pDC, monocytes and cDC are neoplastic since they harbor the same mutations as CD34+ blasts. RUNX1 is the most commonly mutated gene: detected in all AML with minimal differentiation (M0-AML) but not in the other cases. Despite the low number of cases, the systematic association between M0-AML, RUNX1 mutations and an excess of pDC is puzzling. Further evaluation in a larger cohort is required to confirm RUNX1 mutations in pDC-AML with minimal differentiation and to investigate whether it represents a proliferation of blasts with macrophage and DC progenitor potential.