Abstract

Simple SummaryEpigenetic drugs, such as azacitidine (AZA), hold promise in the treatment of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), however, the mechanisms predicting the patients’ response to AZA is not completely understood. Quiescence of hematopoietic CD34+ progenitors has been proposed as a predictive factor for AZA therapy failure in MDS/AML patients, but the interplay between CD34+ cell cycle status and their metabolic signature in a predisposition to AZA (non)responsiveness remains unclear. Our data on patients with MDS or AML with myelodysplasia-related changes (AML-MRC) suggest that AZA-responders have actively cycling CD34+ cells poised for erythro-myeloid differentiation, with high metabolic activity controlling histone acetylation. Conversely, the patients who progressed early on AZA therapy revealed quiescence signature of their CD34+ cells, with signs of reduced metabolically-controlled acetylation of histones needed for transcription-permissive chromatin configuration. Our study delineates plasma citrate levels and CD34+ cells’ transcriptional signatures associated with cycling status and metabolic characteristics as factors predicting the response to AZA monotherapy in MDS/AML-MRC patients.To better understand the molecular basis of resistance to azacitidine (AZA) therapy in myelodysplastic syndromes (MDS) and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC), we performed RNA sequencing on pre-treatment CD34+ hematopoietic stem/progenitor cells (HSPCs) isolated from 25 MDS/AML-MRC patients of the discovery cohort (10 AZA responders (RD), six stable disease, nine progressive disease (PD) during AZA therapy) and from eight controls. Eleven MDS/AML-MRC samples were also available for analysis of selected metabolites, along with 17 additional samples from an independent validation cohort. Except for two patients, the others did not carry isocitrate dehydrogenase (IDH)1/2 mutations. Transcriptional landscapes of the patients’ HSPCs were comparable to those published previously, including decreased signatures of active cell cycling and DNA damage response in PD compared to RD and controls. In addition, PD-derived HSPCs revealed repressed markers of the tricarboxylic acid cycle, with IDH2 among the top 50 downregulated genes in PD compared to RD. Decreased citrate plasma levels, downregulated expression of the (ATP)-citrate lyase and other transcriptional/metabolic networks indicate metabolism-driven histone modifications in PD HSPCs. Observed histone deacetylation is consistent with transcription-nonpermissive chromatin configuration and quiescence of PD HSPCs. This study highlights the complexity of the molecular network underlying response/resistance to hypomethylating agents.

Highlights

  • Hypomethylating agents (HMAs) like azacitidine (AZA) and decitabine have become a standard treatment option for high-risk myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) patients (including AML with myelodysplasia-related changes (AML-MRC)) [1,2,3,4]

  • To examine whether observed differences in the cycling/non-cycling characteristics of CD34+ hematopoietic stem/progenitor cells (HSPCs) populations of RD vs. PD is reflected in the mechanisms of DNA repair activated in these cells, we evaluated transcripts of DNA repair genes distributed to categories as described [36]

  • We addressed the progenitor cell cycling status and the metabolic pathways that could be correlated with responses to AZA therapy in MDS/acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) patients

Read more

Summary

Introduction

Hypomethylating agents (HMAs) like azacitidine (AZA) and decitabine have become a standard treatment option for high-risk myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) patients (including AML with myelodysplasia-related changes (AML-MRC)) [1,2,3,4]. Assessment of epigenetic mutations and detailed DNA methylation patterns in MDS patients has recently enabled the identification of clinically relevant subtypes of MDS, as well as the selection of those patients who have an increased likelihood of sensitivity or resistance to HMA therapy [11,12,13,14]. Association between these molecular factors and eventual response to HMAs is still incompletely understood and, in some cases, remains controversial [15,16,17,18]. It seems that treatment strategies with HMAs should consider differences in HMA cell metabolism within individual patients’ groups ([19,20]) and may include effective combinations of HMAs with other new drugs with activity against MDS/AML ([21,22,23,24,25,26]; recent clinical trials are reviewed in [27,28])

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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