dna hypomethylating agent
Recently Published Documents


TOTAL DOCUMENTS

23
(FIVE YEARS 6)

H-INDEX

7
(FIVE YEARS 0)

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3452-3452
Author(s):  
Sonali P. Barwe ◽  
Erin Lynn Crowgey ◽  
Qinzhuo Zhang ◽  
E. Anders Kolb ◽  
Anilkumar Gopalakrishnapillai

Abstract Background: Down syndrome is known for its leukemia predisposition effects. Children with Down syndrome myeloid leukemia who fail chemotherapy, have a poor outcome. No targeted therapies are available for this population. We have generated and extensively characterized several patient-derived xenograft (PDX) models of Down syndrome AML (Barwe et al., 134:2683, Blood, 2019). We evaluated the efficacy of a combination of DNA hypomethylating agent, azacitidine and Bcl2 inhibitor, venetoclax in PDX model of Down syndrome AML. Although this drug combination is used in the clinic for the treatment of adult AML, their combinatorial mechanism of action is not well known. Methods: NTPL-386 (3 x 10 6 cells) were injected intravenously in NSG-SGM3 mice. Disease progression was monitored by determination of the percentage of human chimerism in peripheral blood by flow cytometry. Mice were randomly assigned to treatment groups (n=5) on day 13 when human CD45+ cells were detectable in blood. Azacitidine (2.5 mg/Kg) was administered intraperitoneally on days 13-19 and venetoclax (100 mg/Kg) was delivered orally on days 13-40 (Fig. 1A). Mice were monitored daily for pre-determined experimental endpoints and were euthanized when any of the endpoints were attained. Kaplan-Meier survival plots were generated. AML cells were harvested from treated mice and lysed, RNA was isolated and sequenced. Pathway analysis was performed to identify the mechanism of action of these two drugs in combination. Results: Azacitidine treatment for one week increased median survival by 8 days compared to untreated mice (Fig. 1B). Venetoclax treatment for 4 weeks was more effective than azacitidine and extended survival by 27 days (P<0.005, compared to untreated). We observed that the combination of venetoclax and azacitidine was much more efficient and prolonged survival by 39 days in this PDX model of refractory Down syndrome AML (P<0.005 and P<0.05 compared to single agent azacitidine or venetoclax respectively). This combination performed better than chemotherapy consisting of cytarabine and daunorubicin, which showed a 11-day improvement in median survival compared to untreated in our previous study (Barwe et al., 134:2683, Blood, 2019). To understand the mechanism of synergism between these two drugs, we conducted transcriptome analysis on cells harvested from mice (n=3 per group) receiving venetoclax and azacitidine, alone or in combination. The log fold changes for each treatment with respect to untreated were calculated and filtered based on FDR>0.05. The genes that were differentially regulated in the combination treatment (Fig. 1C, blue circle in the Venn diagram) were subjected to ToppGene analysis. A heatmap of the differentially regulated genes shows clustering of the replicates from individual mice (Fig. 1D). 'Hematopoietic cell lineage' and 'cytokine signaling in immune system' were the top modulated pathways in the combination treatment. A heatmap of the logFC of the differentially regulated genes from this pathway showed the modulation of these genes in the combination in comparison with individual drug treatment (Fig. 1E). The differentially regulated genes in the combination treatment that showed high variance from the range defined by individual drug treatment belonged to one of three categories - cell surface proteins, cytokines, and stem cell markers. These data indicate that the synergistic downregulation of cytokine signaling is likely responsible for the combinatorial effect of venetoclax and azacitidine. Conclusion: The combination of venetoclax and azacitidine was effective in prolonging survival in a highly refractory Down syndrome AML PDX model (NTPL-386) generated in the laboratory. The top genes differentially regulated by the combination treatment which varied the most in magnitude from the individual drug treatment identified genes that are likely to contribute to the synergism between the two drugs. Figure 1 Figure 1. Disclosures Barwe: Prelude Therapeutics: Research Funding. Gopalakrishnapillai: Geron: Research Funding.


2021 ◽  
Vol 118 (9) ◽  
pp. e2021795118
Author(s):  
Kartik N. Rajagopalan ◽  
Xiao Chen ◽  
Daniel N. Weinberg ◽  
Haifen Chen ◽  
Jacek Majewski ◽  
...  

Hotspot histone H3 mutations have emerged as drivers of oncogenesis in cancers of multiple lineages. Specifically, H3 lysine 36 to methionine (H3K36M) mutations are recurrently identified in chondroblastomas, undifferentiated sarcomas, and head and neck cancers. While the mutation reduces global levels of both H3K36 dimethylation (H3K36me2) and trimethylation (H3K36me3) by dominantly inhibiting their respective specific methyltransferases, the relative contribution of these methylation states to the chromatin and phenotypic changes associated with H3K36M remains unclear. Here, we specifically deplete H3K36me2 or H3K36me3 in mesenchymal cells, using CRISPR-Cas9 to separately knock out the corresponding methyltransferases NSD1/2 or SETD2. By profiling and comparing the epigenomic and transcriptomic landscapes of these cells with cells expressing the H3.3K36M oncohistone, we find that the loss of H3K36me2 could largely recapitulate H3.3K36M’s effect on redistribution of H3K27 trimethylation (H3K27me3) and gene expression. Consistently, knockout of Nsd1/2, but not Setd2, phenocopies the differentiation blockade and hypersensitivity to the DNA-hypomethylating agent induced by H3K36M. Together, our results support a functional divergence between H3K36me2 and H3K36me3 and their nonredundant roles in H3K36M-driven oncogenesis.


2020 ◽  
Vol 25 ◽  
pp. 100321
Author(s):  
Parunya Chaiyawat ◽  
Nutnicha Sirikaew ◽  
Piyaporn Budprom ◽  
Jeerawan Klangjorhor ◽  
Areerak Phanphaisarn ◽  
...  

2019 ◽  
Author(s):  
Caroline Bull ◽  
Graham Mayrhofer ◽  
Michael Fenech

AbstractBackground5-aza-2’-deoxycytidine (5azadC, decitabine) is a DNA hypomethylating agent used in the treatment of myelodysplastic syndromes. Due to cytotoxic side effects dose optimization is essential. This study defines and quantifies the effects of 5azadC on chromosomal stability and telomere length, at clinically relevant dosages.MethodsHuman WIL2-NS cells were maintained in complete medium containing 0, 0.2 or 1.0μM 5azadC for four days, and analysed daily for telomere length (flow cytometry), chromosomal stability (cytokinesis-block micronucleus cytome (CBMN-cyt) assay), and global methylation (%5me-C).ResultsDNA methylation decreased significantly in 1.0 μM 5azadC, relative to control (p<0.0001). Exposure to 1.0μM 5azadC resulted in 170% increase in telomere length (p<0.0001), in parallel with rapid increase in biomarkers of DNA damage; (micronuclei (MN, 6-fold increase), nucleoplasmic bridges (NPB, a 12-fold increase), and nuclear buds (NBud, a 13-fold increase) (all p<0.0001). Fused nuclei (FUS), indicative of mitotic dysfunction, showed a 5- and 13-fold increase in the 0.2μM and 1.0μM conditions, respectively (p = 0.001) after 4 days.ConclusionsThese data show that (i) clinically relevant concentrations of 5azadC are highly genotoxic; (ii) hypomethylation was associated with increased TL and DNA damage; and (iii) longer TL was associated with chromosomal instability. These findings suggest that lower doses of 5azdC may be effective as a hypomethylating agent, while potentially reducing DNA damage and risk for secondary disease.


Epigenetics ◽  
2016 ◽  
Vol 11 (10) ◽  
pp. 709-720 ◽  
Author(s):  
Simone Jueliger ◽  
John Lyons ◽  
Sara Cannito ◽  
Illar Pata ◽  
Pille Pata ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document