Abstract B126: A small molecule inhibitor of the RNA m6A writer METTL3 inhibits the development of acute myeloid leukemia (AML) in vivo

Author(s):  
Mark Albertella ◽  
Wesley Blackaby ◽  
Richard Fosbeary ◽  
Alan Hendrick ◽  
Dan Leggate ◽  
...  
Blood ◽  
2016 ◽  
Vol 127 (9) ◽  
pp. 1173-1182 ◽  
Author(s):  
Sagar Uttarkar ◽  
Emilie Dassé ◽  
Anna Coulibaly ◽  
Simone Steinmann ◽  
Anke Jakobs ◽  
...  

Key Points Inhibition of Myb activity by a small molecule blocks proliferation of AML cells and prolongs survival of mice in an in vivo AML model.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3849-3849
Author(s):  
Alisa B. Lee-Sherick ◽  
Kelly Menachof ◽  
Amanda A Hill ◽  
Sean Rinella ◽  
Jing Liu ◽  
...  

Abstract The identification of novel biologically targeted agents for the treatment of acute myeloid leukemia (AML) could potentially augment therapeutic outcome, decrease toxicity to normal tissues, and/or provide a therapy option for those who are not able to tolerate conventional therapy. Previously, Flt-3 and Mer have been identified as potential targets in the treatment of acute myeloid leukemia. Flt-3 internal tandem duplication (ITD) has been described in ∼30-40% of AML patient samples and Mer overexpression has been detected in ∼80-100% of AML patient samples. Here we describe a novel first-in-class small molecule inhibitor that has dual inhibition of these two tyrosine kinases with effective killing of myeloblasts in preclinical evaluation. UNC1666 is an ATP competitive reversible small molecule inhibitor which potentially inhibits Mer (IC50: 0.55 nM) and Flt-3 (IC50: 0.69 nM), preventing phosphorylation of the kinase and resultant downstream signaling. In these studies, UNC1666 was analyzed in AML cell lines which were either known to be Mer-positive (Nomo-1, Kasumi-1 and U937) or positive for the Flt3-ITD (Molm-13 and MV4;11). Short term exposure of these cell lines to UNC1666 resulted in decreased AKT and STAT6 activation in a dose dependent manner compared to equivalent concentration of drug vehicle (DMSO). AML cell lines were treated with nanomolar doses of UNC1666 or equivalent concentration of vehicle for 72 hours, stained with Yo-Pro-1 iodide and propidium iodide, and were assessed for apoptosis by flow cytometry. Compared to AML cells treated with vehicle, treatment of Mer-positive cell lines with UNC1666 resulted in a two to five-fold induction of apoptosis (mean of 20±10% apoptosis after treatment with vehicle versus 66±10% after treatment with UNC1666, p<0.01). Treatment of Flt3-ITD cell lines with UNC1666 resulted in a nine-fold induction of apoptosis (mean of 10±2% apoptosis after treatment with vehicle versus 90±6% after treatment with UNC1666, p<0.001). To further evaluate for sustained effects of UNC1666 on cell growth and survival after drug removal, AML cell lines were treated with UNC1666 for 72 hours, then washed and replated in normal growth conditions to assess for possible rebound growth. Viable cell count was performed on a Cedex analyzer using trypan blue exclusion over the course of six days. Cells treated with UNC1666 continued to have a significant decrease in their ability to proliferate even after the drug was removed (day six viable cell count of 1.9 x 106 cells previously treated with vehicle versus 0.04 x 106 cells previously treated with UNC1666, p<0.01). Furthermore, when these AML cell lines were plated in equal number in soft agar, and then treated with medium containing nanomolar doses of UNC1666 for 10 days, cell lines treated with the dual inhibitor demonstrated decreased colony formation compared to cells treated with medium containing vehicle only (relative colony count of 100 with vehicle treatment versus 34±15 colonies with Mer-positive cell lines and 15±12 with Flt3-ITD cell lines after treatment with UNC1666, p<0.01). In summary, UNC1666 is novel first-in-class small molecule with ability to inhibit kinase activity and activation of both Mer and Flt-3 tyrosine kinases. Inhibition using this molecule effectively prevented downstream activation of several intracellular signaling molecules including AKT and STAT6, induced apoptosis and decreased myeloblast proliferative capacity. These data validate dual Mer/Flt-3 inhibition as a new and attractive potentially clinically applicable therapeutic in the treatment of AML. Disclosures: Liu: University of North Carolina: inventor on a patent application relevant to this work, inventor on a patent application relevant to this work Patents & Royalties. Wang:University of North Carolina at Chapel Hill: This author is an inventor on patent application relevant to this work, This author is an inventor on patent application relevant to this work Patents & Royalties. Frye:University of North Carolina at Chapel Hill: This author is an inventor on a patent application relevant to this work, This author is an inventor on a patent application relevant to this work Patents & Royalties.


Oncotarget ◽  
2015 ◽  
Vol 6 (9) ◽  
pp. 6722-6736 ◽  
Author(s):  
Alisa B. Lee-Sherick ◽  
Weihe Zhang ◽  
Kelly K. Menachof ◽  
Amanda A. Hill ◽  
Sean Rinella ◽  
...  

Blood ◽  
2015 ◽  
Vol 125 (2) ◽  
pp. 296-303 ◽  
Author(s):  
Andrew Kernytsky ◽  
Fang Wang ◽  
Erica Hansen ◽  
Stefanie Schalm ◽  
Kimberly Straley ◽  
...  

Key Points IDH2 R140Q expression in TF-1 cells can induce DNA and histone hypermethylation that mirrors human IDH2 mutant acute myeloid leukemia. The hypermethylation can be reversed on treatment with AGI-6780, an IDH2 mutant-specific small-molecule inhibitor.


Leukemia ◽  
2021 ◽  
Author(s):  
Madeline Niederkorn ◽  
Chiharu Ishikawa ◽  
Kathleen M. Hueneman ◽  
James Bartram ◽  
Emily Stepanchick ◽  
...  

AbstractUbiquitin-specific peptidase 15 (USP15) is a deubiquitinating enzyme implicated in critical cellular and oncogenic processes. We report that USP15 mRNA and protein are overexpressed in human acute myeloid leukemia (AML) as compared to normal hematopoietic progenitor cells. This high expression of USP15 in AML correlates with KEAP1 protein and suppression of NRF2. Knockdown or deletion of USP15 in human and mouse AML models significantly impairs leukemic progenitor function and viability and de-represses an antioxidant response through the KEAP1-NRF2 axis. Inhibition of USP15 and subsequent activation of NRF2 leads to redox perturbations in AML cells, coincident with impaired leukemic cell function. In contrast, USP15 is dispensable for human and mouse normal hematopoietic cells in vitro and in vivo. A preclinical small-molecule inhibitor of USP15 induced the KEAP1-NRF2 axis and impaired AML cell function, suggesting that targeting USP15 catalytic function can suppress AML. Based on these findings, we report that USP15 drives AML cell function, in part, by suppressing a critical oxidative stress sensor mechanism and permitting an aberrant redox state. Furthermore, we postulate that inhibition of USP15 activity with small molecule inhibitors will selectively impair leukemic progenitor cells by re-engaging homeostatic redox responses while sparing normal hematopoiesis.


2018 ◽  
Author(s):  
Gerard Minuesa ◽  
Steven K. Albanese ◽  
Arthur Chow ◽  
Alexandra Schurer ◽  
Sun-Mi Park ◽  
...  

SUMMARYThe MUSASHI family of RNA binding proteins (MSI1 and MSI2) contribute to a wide spectrum of cancers including acute myeloid leukemia. We found that the small molecule Ro 08–2750 (Ro) directly binds to MSI2 and competes for its RNA binding in biochemical assays. Ro treatment in mouse and human myeloid leukemia cells resulted in an increase in differentiation and apoptosis, inhibition of known MSI-targets, and a shared global gene expression signature similar to shRNA depletion of MSI2. Ro demonstrated in vivo inhibition of c-MYC and reduced disease burden in a murine AML leukemia model. Thus, we have identified a small molecule that targets MSI’s oncogenic activity. Our study provides a framework for targeting RNA binding proteins in cancer.


Sign in / Sign up

Export Citation Format

Share Document