Molecular synergy underlies the co-occurrence patterns and phenotype of NPM1-mutant acute myeloid leukemia

OM Dovey, JL Cooper, A Mupo… - Blood, The Journal …, 2017 - ashpublications.org
OM Dovey, JL Cooper, A Mupo, CS Grove, C Lynn, N Conte, RM Andrews, S Pacharne…
Blood, The Journal of the American Society of Hematology, 2017ashpublications.org
NPM1 mutations define the commonest subgroup of acute myeloid leukemia (AML) and
frequently co-occur with FLT3 internal tandem duplications (ITD) or, less commonly, NRAS
or KRAS mutations. Co-occurrence of mutant NPM1 with FLT3-ITD carries a significantly
worse prognosis than NPM1-RAS combinations. To understand the molecular basis of these
observations, we compare the effects of the 2 combinations on hematopoiesis and
leukemogenesis in knock-in mice. Early effects of these mutations on hematopoiesis show …
Abstract
NPM1 mutations define the commonest subgroup of acute myeloid leukemia (AML) and frequently co-occur with FLT3 internal tandem duplications (ITD) or, less commonly, NRAS or KRAS mutations. Co-occurrence of mutant NPM1 with FLT3-ITD carries a significantly worse prognosis than NPM1-RAS combinations. To understand the molecular basis of these observations, we compare the effects of the 2 combinations on hematopoiesis and leukemogenesis in knock-in mice. Early effects of these mutations on hematopoiesis show that compound Npm1cA/+;NrasG12D/+ or Npm1cA;Flt3ITD share a number of features: Hox gene overexpression, enhanced self-renewal, expansion of hematopoietic progenitors, and myeloid differentiation bias. However, Npm1cA;Flt3ITD mutants displayed significantly higher peripheral leukocyte counts, early depletion of common lymphoid progenitors, and a monocytic bias in comparison with the granulocytic bias in Npm1cA/+;NrasG12D/+ mutants. Underlying this was a striking molecular synergy manifested as a dramatically altered gene expression profile in Npm1cA;Flt3ITD, but not Npm1cA/+;NrasG12D/+, progenitors compared with wild-type. Both double-mutant models developed high-penetrance AML, although latency was significantly longer with Npm1cA/+;NrasG12D/+. During AML evolution, both models acquired additional copies of the mutant Flt3 or Nras alleles, but only Npm1cA/+;NrasG12D/+ mice showed acquisition of other human AML mutations, including IDH1 R132Q. We also find, using primary Cas9-expressing AMLs, that Hoxa genes and selected interactors or downstream targets are required for survival of both types of double-mutant AML. Our results show that molecular complementarity underlies the higher frequency and significantly worse prognosis associated with NPM1c/FLT3-ITD vs NPM1/NRAS-G12D-mutant AML and functionally confirm the role of HOXA genes in NPM1c-driven AML.
ashpublications.org