Abstract

Acute myeloid leukemia (AML) is a very heterogeneous and highly malignant blood cancer. Mutations of the DNA methyltransferase DNMT3A are among the most frequent recurrent genetic lesions in AML. The majority of DNMT3A-mutant AML patients shows fast relapse and poor survival, but also patients with long survival or long-term remission have been reported. Underlying molecular signatures and mechanisms that contribute to these survival differences are only poorly understood and have not been studied in detail so far. We applied hierarchical clustering to somatic gene mutation profiles of 51 DNMT3A-mutant patients from The Cancer Genome Atlas (TCGA) AML cohort revealing two robust patient subgroups with profound differences in survival. We further determined molecular signatures that distinguish both subgroups. Our results suggest that FLT3 and/or NPM1 mutations contribute to survival differences of DNMT3A-mutant patients. We observed an upregulation of genes of the p53, VEGF and DNA replication pathway and a downregulation of genes of the PI3K-Akt pathway in short- compared to long-lived patients. We identified that the majority of measured miRNAs was downregulated in the short-lived group and we found differentially expressed microRNAs between both subgroups that have not been reported for AML so far (miR-153-2, miR-3065, miR-95, miR-6718) suggesting that miRNAs could be important for prognosis. In addition, we learned gene regulatory networks to predict potential major regulators and found several genes and miRNAs with known roles in AML pathogenesis, but also interesting novel candidates involved in the regulation of hematopoiesis, cell cycle, cell differentiation, and immunity that may contribute to the observed survival differences of both subgroups and could therefore be important for prognosis. Moreover, the characteristic gene mutation and expression signatures that distinguished short- from long-lived patients were also predictive for independent DNMT3A-mutant AML patients from other cohorts and could also contribute to further improve the European LeukemiaNet (ELN) prognostic scoring system. Our study represents the first in-depth computational approach to identify molecular factors associated with survival differences of DNMT3A-mutant AML patients and could trigger additional studies to develop robust molecular markers for a better stratification of AML patients with DNMT3A mutations.

Highlights

  • Acute myeloid leukemia (AML) is a very heterogeneous and highly malignant blood cancer

  • Considering the gene mutation data from The Cancer Genome Atlas (TCGA) for all 197 AML patients, we found that 51 of them had a DNA methlytransferase 3A (DNMT3A) mutation

  • The shortlived subgroup was enriched with patients harboring a R882 DNMT3A mutation (n=17, 71%) compared to patients with non-R882 mutations (n=7, 29%), while the long-lived subgroup was composed of 12 patients with R882 (44%) and 15 patients with non-R882 mutations (56%)

Read more

Summary

Introduction

Acute myeloid leukemia (AML) is a very heterogeneous and highly malignant blood cancer. We applied hierarchical clustering to somatic gene mutation profiles of 51 DNMT3A-mutant patients from The Cancer Genome Atlas (TCGA) AML cohort revealing two robust patient subgroups with profound differences in survival. The characteristic gene mutation and expression signatures that distinguished short- from long-lived patients were predictive for independent DNMT3A-mutant AML patients from other cohorts and could contribute to further improve the European LeukemiaNet (ELN) prognostic scoring system. Acute myeloid leukemia (AML) is a highly malignant cancer of myeloid blood cells affecting about one million people globally in 2­ 0151,2 It most frequently occurs in older adults and shows a relatively poor five-year survival rate of about 25%, which is worsening with increasing age of a patient at ­diagnosis[3]. Significant associations of DNMT3A mutations with IDH1/2 mutations, FLT3 internal tandem duplications (ITD) and tyrosine kinase domain mutations (TKD), and NPM1 mutations have been r­ eported[9,28]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.