Abstract 1319: A novel formulation of CX-5461, a small-molecule inhibitor of rRNA synthesis, and its use for treatment of acute myeloid leukemia models

Author(s):  
Ada W.Y. Leung ◽  
Malathi Anantha ◽  
Kathleen E. Prosser ◽  
Mohamed Wehbe ◽  
Charles J. Walsby ◽  
...  
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 ◽  
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 ◽  
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.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1021-1021
Author(s):  
Ronan T. Swords ◽  
Kevin R. Kelly ◽  
Peter G. Smith ◽  
James J. Gansey ◽  
Devalingam Mahalingam ◽  
...  

Abstract Abstract 1021 Poster Board I-43 The coordinated balance between the synthesis and degradation of proteins is an important regulator of cancer cell biology. The ubiquitin-proteasome system (UPS) is responsible for the timed destruction of many proteins including key mediators of fundamental signaling cascades and critical regulators of cell cycle progression and transcription. Within the UPS, the E3 ligases are multi-protein complexes whose specificity is established by their individual components as well as post-translational modifications by various factors including the ubiquitin-like molecule, Nedd8. The Nedd8 activating enzyme (NAE) has been identified as an essential regulator of the Nedd8 conjugation pathway, which controls the activity of the cullin-dependent E3 ubiquitin ligases. The cullins direct the ubiquitination and subsequent degradation of many proteins with important roles in cell cycle progression (p27, cyclin E), DNA damage (Cdt-1), stress response (NRF-2, HIF1α) and signal transduction (IκBα). Acute myeloid leukemia (AML) is a disease of the elderly and prognosis is extremely poor with a median overall survival of just 2 months for untreated patients. As such, novel therapeutic strategies are urgently needed to improve clinical outcomes. Considering that Nedd8-mediated control of protein homeostasis is vitally important for the survival of AML cells, we hypothesized that disrupting this process would inhibit proliferation and induce cell death. We tested this hypothesis by investigating the preclinical anti-leukemic activity of MLN4924, a novel first in class small molecule inhibitor of the Nedd8 activating enzyme. MLN4924 induced DNA damage followed by rapid and selective caspase-dependent cell death in AML cell lines and primary AML cells from patients, but not in peripheral blood mononuclear cells from healthy donors. Transient exposure to MLN4924 impaired colony formation in a dose-dependent manner. Kinetic analysis of drug-induced effects on cell cycle distribution revealed that AML cells treated with MLN4924 initially arrested at the G1 transition prior to their subsequent accumulation in the sub-G1 compartment. Assays conducted using MV-411 cells with and without stable shRNA-mediated knockdown of FLT3 expression demonstrated that MLN4924 is highly effective independent of FLT3 status. Further investigation revealed that the activity of MLN4924 was preserved when cells were co-cultured with bone marrow stromal cells indicating that it has the ability to overcome the effects of stromal-mediated survival signaling that has been established to blunt the efficacy of relevant standard of care agents. MLN4924 induced a dose and time dependant increase in the expression of phospo-IκB, an important target for degradation through the Nedd8 conjugation pathway. The inhibitory effects of MLN4924 on NFκB were confirmed by demonstrating that the transcriptional activity of the NFκB p65 subunit was significantly reduced following drug exposure. Moreover, treatment of immunodeficient mice implanted with HL-60 human leukemia cells with MLN4924 led to an inhibition of neddylated cullins, accumulation of phospho-IκBα and achieved complete and stable disease regression. Our results indicate that MLN4924 is a highly promising novel agent for the treatment of AML and warrants further evaluation in clinical trials. Disclosures: Smith: Millennium Pharmaceuticals: Employment. Gansey:Millennium Pharmaceuticals: Employment.


Sign in / Sign up

Export Citation Format

Share Document