4.7 Article

Asxl1 loss cooperates with oncogenic Nras in mice to reprogram the immune microenvironment and drive leukemic transformation

Journal

BLOOD
Volume 139, Issue 7, Pages 1066-1079

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2021012519

Keywords

-

Categories

Funding

  1. National Institutes of Health, National Cancer Institute [R01 CA236356, P01 CA108671, R37 CA234021, R01 CA152108, P30 CA014520]

Ask authors/readers for more resources

Mutations in ASXL1 and NRAS genes are associated with poor prognosis in CMML. These mutations lead to dysregulation of MEK/ERK signaling, overexpression of immune checkpoint ligands, and exhaustion of T cells. Combination of targeted therapy and immunotherapy can improve these abnormalities and prolong the survival of AML patients.
Mutations in chromatin regulator ASXL1 are frequently identified in myeloid malignancies, in particular similar to 40% of patients with chronic myelomonocytic leukemia (CMML). ASXL1 mutations are associated with poor prognosis in CMML and significantly co-occur with NRAS mutations. Here, we show that concurrent ASXL1 and NRAS mutations defined a population of CMML patients who had shorter leukemia-free survival than those with ASXL1 mutation only. Corroborating this human data, Asxl1(-/-) accelerated CMML progression and promoted CMML transformation to acute myeloid leukemia (AML) in Nras(G12D/+) mice. Nras(G12D/+);Asxl1(-/-) (NA) leukemia cells displayed hyperactivation of MEK/ERK signaling, increased global levels of H3K27ac, upregulation of Flt3. Moreover, we find that NA-AML cells overexpressed all the major inhibitory immune checkpoint ligands: programmed death-ligand 1 (PD-L1)/PD-L2, CD155, and CD80/CD86. Among them, overexpression of PD-L1 and CD86 correlated with upregulation of AP-1 transcription factors (TFs) in NA-AML cells. An AP-1 inhibitor or short hairpin RNAs against AP-1 TF Jun decreased PD-L1 and CD86 expression in NA-AML cells. Once NA-AML cells were transplanted into syngeneic recipients, NA-derived T cells were not detectable. Host derived wild-type T cells overexpressed programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) receptors, leading to a predominant exhausted T-cell phenotype. Combined inhibition of MEK and BET resulted in downregulation of Flt3 and AP-1 expression, partial restoration of the immune microenvironment, enhancement of CD8 T-cell cytotoxicity, and prolonged survival in NA-AML mice. Our study suggests that combined targeted therapy and immunotherapy may be beneficial for treating secondary AML with concurrent ASXL1 and NRAS mutations.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available