Pre-Clinical Efficacy of Combined Therapy with Novel β-Catenin Antagonist BC2059 and Histone Deacetylase Inhibitor Against Cultured and Primary AML Blast Progenitor Cells

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 287-287
Author(s):  
Kapil Bhalla ◽  
Warren Fiskus ◽  
Stephen K. Horrigan ◽  
Sunil Abhyankar ◽  
Joseph McGuirk ◽  
...  

Abstract Abstract 287 The canonical WNT-β-catenin pathway is essential for self-renewal, growth and survival of AML stem and early blast progenitor cells (BPCs). Deregulated WNT signaling in AML inhibits polyubiquitylation and proteasomal degradation of β-catenin by the multi-protein complex. The inactivation of the degradation complex for β-catenin results in the preservation, nuclear translocation and activity of β-catenin in AML BPCs. This is especially notable in AML BPCs expressing FLT3-ITD, where increased activity of PI3K/AKT phosphorylates and inactivates GSK3β, thereby inhibiting phospho-degradation, increasing stability and nuclear accumulation of β-catenin. Activated FLT3 kinase has also been shown to directly phosphorylate β-catenin and promote its stabilization and nuclear localization. As a co-activator, in the nucleus β-catenin interacts with the bipartite T-cell factor (TCF)/lymphoid enhancer factor (LEF) transcription factor, which increases the expression of pro-growth and pro-survival genes, including cyclin D1, Myc and survivin. Aberrant expression of LEF1 in hematopoietic stem cells has been shown to induce AML. β-catenin is also required for the HOXA9 and MEIS1 or MLL-AF9-mediated transformation of hematopoietic stem/progenitor cells. Thus, reactivation of β-catenin is required for maintenance of leukemic transformation making it an attractive drug target in AML. Here, we determined the in vitro and in vivo anti-AML activity of BC2059 (β-Cat Pharmaceuticals), a potent, small molecule, anthraquinone oxime-analog, against cultured (HL-60, OCI-AML3 and MV4-11 cells) and primary AML BPCs with or without the expression of FT3-ITD. First, as compared to the normal human CD34+ cells, FLT3-ITD expressing AML BPCs showed increased levels of β-catenin, mostly in the nucleus, as determined by confocal immunofluorescence microscopy. Exposure to 100 nM of BC2059 (BC) induced β-catenin degradation through the proteasome, as well as attenuated the nuclear and cytoplasmic levels of β-catenin in the cultured and primary AML BPC. Chromatin immunoprecipitation with anti-β-catenin antibody demonstrated that treatment with BC2059 (BC) reduced the binding of β-catenin to the WNT response elements (WRE) in the promoter DNA of its target genes, including Myc, cyclin D1 and survivin. Estimation of the intracellular luciferase levels in AML cells transfected with the TOP/FLASH versus FOP/FLASH construct showed that treatment with BC2059 significantly reduced only the TOP-FLASH luciferase activity, indicating that BC inhibits the expression of genes with promoters containing WRE elements. This was associated with reduced mRNA and protein levels of cyclin D1, MYC and survivin. Treatment with BC dose-dependently induced apoptosis of cultured and primary AML BPCs (up to 70%), including apoptosis of the CD34+CD38-Lin- AML BPCs. In contrast, BC induced apoptosis in < 10% of normal CD34+ progenitor cells. We have previously reported that treatment with the histone deacetylase inhibitor panobinostat (PS) attenuates p-FLT3, p-AKT and p-GSK3β levels in AML BPCs expressing FLT3-ITD. Consistent with this, here, we determined that co-treatment with BC and PS (10 to 20 nM) synergistically induced apoptosis of cultured and primary AML BPCs. Against primary AML BPCs expressing FLT3-ITD, co-treatment with the FLT3 kinase inhibitor AC220 (100 nM) further augmented BC mediated depletion of the cytoplasmic and nuclear levels of β-catenin and significantly enhanced BC-induced apoptosis (p < 0.01). This was associated with induction of BIM and p27 with depletion of MCL-1 levels. Following tail vein infusion and establishment of AML by OCI-AML3 cells in NOD-SCID mice, treatment with BC (5 or 10 mg/kg b.i.w, IV) for three weeks demonstrated improved survival of the mice compared to the vehicle control treated mice (p <0. 001). Survival was further improved upon co-treatment with BC and PS (5 mg/kg IP, MWF). BC treatment (5 or 10 mg/kg IV) also dramatically improved survival of the NOD/SCID/IL2Rγ-depleted mice with established human AML following tail-vein injection of primary AML BPCs expressing FLT3 ITD. Mice did not experience any toxicity or weight loss. These findings support a compelling rationale for further development and in vivo testing of the BC-based combination with PS and FLT3 antagonist against human AML BPCs. Disclosures: Horrigan: BetaCat Pharmaceuticals: Employment. Sharma:BetaCat Pharmaceuticals: Equity Ownership.

Blood ◽  
2010 ◽  
Vol 116 (24) ◽  
pp. 5306-5315 ◽  
Author(s):  
Aditya Mandawat ◽  
Warren Fiskus ◽  
Kathleen M. Buckley ◽  
Kelly Robbins ◽  
Rekha Rao ◽  
...  

Abstract Stromal cell derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 are involved in the directional homing to the bone marrow niches and in peripheral mobilization of normal and transformed hematopoietic stem and myeloid progenitor cells. Elevated CXCR4 expression confers poor prognosis, whereas inhibition of CXCR4 signaling overcomes stroma-mediated chemoresistance in acute myeloid leukemia (AML). Here, we demonstrate that treatment with the pan-histone deacetylase inhibitor panobinostat (PS) depleted the mRNA and protein levels of CXCR4 in the cultured and primary AML cells. PS-induced acetylation of the heat shock protein (hsp) 90 reduced the chaperone association between CXCR4 and hsp90, directing CXCR4 to degradation by the 20S proteasome. PS treatment also depleted G protein–coupled receptor kinase 3, as well as attenuated the phosphorylation of AKT and ERK1/2 in AML cells, which was not affected by cotreatment with CXCL12. Compared with each agent alone, cotreatment with PS and CXCR4 antagonist AMD3100 or FC-131 synergistically induced apoptosis of cultured and primary AML cells. PS and FC-131 exerted more lethal effects on primary AML versus normal CD34+ bone marrow progenitor cells. These findings support the rationale to test the in vivo efficacy of PS in enhancing the lethal effects of CXCR4 antagonists against AML cells.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 65-65
Author(s):  
Warren Fiskus ◽  
Jacqueline Smith ◽  
Uma Mudunuru ◽  
Stacey Hembruff ◽  
Ruben Reyes ◽  
...  

Abstract Abstract 65 The canonical WNT-β-catenin pathway is essential to the cellular processes of self-renewal, growth and survival. Deregulated WNT-β-catenin in transformed hematopoietic progenitor cells inhibits the multi-protein degradation complex formed by axin, adenomatous polyposis coli (APC) and glycogen synthase kinase 3β (GSK3β). This results in the preservation, nuclear translocation and interaction of β-catenin with the T-cell factor (TCF)/lymphoid enhancer factor (LEF) transcription factor, which regulates the expression of genes such as cyclin D1, Myc, survivin and Axin. BC2059 (β-Cat Pharmaceuticals) is a potent small molecule, anthraquinone oxime-analog inhibitor of the WNT-β catenin pathway. Treatment with BC2059 mediates the degradation of β-catenin. In the present studies, we determined the activity of BC2059 in human cultured and primary CML and advanced MPN versus normal progenitor cells. Exposure to 50 to 100 nM of BC2059 induced cell cycle G1 phase accumulation and apoptosis (40 to 80%) of the cultured MPN cells HEL92.1.7 (HEL) and UKE1 cells expressing the mutant JAK2V617F, as well as of the CML K562 and LAMA-84 cells expressing BCR-ABL. BC2059 treatment also induced apoptosis of CD34+ primary MPN cells derived from the peripheral blood of patients with advanced MPN expressing mutant JAK2, as well as of primary CD34+ CML progenitor cells. In contrast, as compared to the untreated controls, BC2059 treatment did not induce apoptosis of normal CD34+ progenitor cells. Exposure to BC2059 resulted in marked down regulation of β-catenin protein levels and the activity of the LEF1/TCF4 transcription factor, which was accompanied with reduced levels of cyclin D1, MYC, survivin and up regulation of Axin 2 levels, as detected by immunoblot analyses of the cell lysates of BC2059-treated CML and MPN cells. We also determined the in vivo anti-MPN activity of BC2059. Following the tail vein infusion of HEL cells and establishment of MPN, NOD-SCID mice were treated with 15 or 20 mg/Kg of BC2059 administered b.i.w for three weeks via the tail vein. As compared to the control, BC2059-treated mice demonstrated significantly improved survival (p <0. 001). Next, we examined the effects of co-treatment with BC2059 (20 to 50 nM) and JAK2-targeted TKI TG101209 (TG) (200–1000 nM) or BCR-ABL-targeted TKI nilotinib (10–20 nM) against MPN or CML cells, respectively. As compared to treatment with each agent alone co-treatment with BC2059 and TG synergistically induced apoptosis of HEL and primary CD34+ MPN cells. Additionally, co-treatment with BC2059 and nilotinib induced synergistic apoptosis of K562 and primary CML progenitor cells. Further, combined treatment with BC2059 and the HDAC inhibitor panobinostat (10 to 20 nM) also induced significantly more apoptosis of HEL and K562 (p < 0.01), as well as of the primary CD34+ MPN and primary CML progenitor cells. In normal CD34+ progenitor cells, the BC2059-based combinations were remarkably less toxic (p < 0.01). These findings demonstrate that BC2059 exerts notable in vitro and in vivo activity against human MPN and CML versus normal CD34+ progenitor cells. Additionally, BC2059 may exert superior activity in combination with JAK2 or BCR-ABL-targeted TKI, or with pan-HDAC inhibitor against human MPN or CML progenitor cells. Disclosures: Reyes: Millennium, Sanofi Aventis: Consultancy. Horrigan:BetaCat Pharmaceuticals: Employment, Equity Ownership. Sharma:Beta Cat Pharmaceuticals: Equity Ownership.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 10605-10605
Author(s):  
Kapil N. Bhalla ◽  
Warren Fiskus ◽  
Sunil Sharma ◽  
Stephen Horrigan ◽  
Uma Mudunuru ◽  
...  

10605 Background: The canonical WNT-β-catenin pathway is essential for self-renewal, growth and survivalof AML stem and progenitor cells. Deregulated WNT signaling inhibits degradation of β-catenin, causing increased nuclear translocation and interaction of β-catenin with the TCF/LEF transcription factor, which up regulates cyclin D1, Myc and survivin expression in AML progenitor cells. BC2059 (β-Cat Pharmaceuticals) is a potent, small molecule, anthraquinone oxime-analog, which inhibits WNT-β catenin pathway by promoting the degradation and attenuation of β-catenin levels. Methods: We determined the in vitro anti-AML activity of BC2059 (BC) (250 to 1000 nM) against cultured and primary human AML blast progenitors, as well as evaluated the in vivo anti-AML efficacy of BC in NOD-SCID and NOD-SCID-IL2γ receptor deficient (NSG) mice. Results: BC induced cell cycle G1 phase accumulation and apoptosis (40%) of the cultured OCI-AML3, HL-60 and HEL92.1.7 (HEL) AML cells. BC dose-dependently also induced apoptosis of primary AML versus normal progenitors. Treatment with BC resulted in proteasomal degradation and decline in the nuclear levels of β-catenin, which led to decreased activity of the LEF1/TCF4 transcription factor highlighted by reduced TOP-FLASH luciferase activity in the AML cells. This was associated with reduced protein levels of cyclin D1, MYC and survivin. Co-treatment with BC and the histone deacetylase inhibitor panobinostat (PS) (10 to 20 nM) synergistically induced apoptosis of cultured and primary AML blasts. Following tail vein infusion and establishment of AML by OCI-AML3 or HEL cells in NOD-SCID mice, treatment with BC (5, 10 or 15 mg/kg b.i.w, IV) for three weeks demonstrated improved survival, as compared to the control mice (p <0. 001). Survival was further improved upon co-treatment with BC and PS (5 mg/kg IP, MWF). BC treatment (5 or 10 mg/kg IV) also dramatically improved survival of NSG mice with established human AML following tail-vein injection of primary AML blasts expressing FLT3 ITD. Mice did not experience any toxicity or weight loss. Conclusions: These findings highlight the notable pre-clinical in vitro and in vivo activity and warrant further development and in vivo testing of BC against human AML.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1417-1417
Author(s):  
Hilal Gul ◽  
Leah A. Marquez-Curtis ◽  
Ashraf Kharrat ◽  
Jencet Montano ◽  
A. Robert Turner ◽  
...  

Abstract Stromal-cell derived factor (SDF)-1α (CXCL12) is a potent chemoattractant for hematopoietic stem/progenitor cells (HSPC) whose chemotactic effect is mediated through the G protein-coupled receptor CXCR4. The expression level of CXCR4 on leukemic blasts is known to be a major prognostic factor in acute myeloid leukemia (AML) because it increases cell retention, survival and growth within the bone marrow (BM) microenvironment, resulting in resistance to conventional chemotherapy. Modulation of CXCR4 expression would be relevant not only in the trafficking of normal HSPC and leukemic cells but also in the response of the latter to chemotherapeutic agents. Previously, we demonstrated that valproic acid (VPA), an effective histone deacetylase inhibitor (HDI) known to induce differentiation in leukemic blasts, increases proliferation, self-renewal and engraftment of normal murine HSPC (Cancer Res.2005:65;2537), and increases the response of AML cells to chemotherapeutic agents (Blood2003:102). Because of this differential effect we hypothesized that VPA enhances homing/engraftment of normal human HSPC by increasing the cell surface expression of CXCR4 but induces the release of AML blasts from BM by decreasing it, making the AML blasts more susceptible to chemotherapy. Thus, we examined the effect of VPA on CXCR4 expression (by FACS analysis and real-time RT-PCR) and on functional response towards an SDF-1α gradient (by chemotaxis assay) of normal HSPC (CD34+ cells from cord blood (CB)) and AML cells (KG-1 and HL-60 cell lines and primary AML cells). Cells were incubated for 24 h and 48 h in IMDM supplemented with 20% FCS in the presence of 1 mM VPA. We found that VPA increases by about 2-fold the percentage of CXCR4-expressing CB CD34+ and KG-1 cells after 24 h incubation, but did not increase the percentage of CXCR4-expressing HL-60 or primary AML cells. Surprisingly, after 48 h incubation we found that VPA decreases (up to 3-fold) the percentage of CXCR4-expressing HL-60 and primary AML cells. These effects were also confirmed at the mRNA level in KG-1 cells (8-fold upregulation) and in HL-60 cells (2.5-fold downregulation). Consistent with these findings, VPA was also found to increase (1.8-fold for normal CD34+ cells and 1.6-fold for KG-1 cells) chemotaxis towards SDF-1α (20 ng/mL), which was inhibited in KG-1 cells by AMD3100, a potent antagonist of CXCR4. On the other hand, chemotaxis was decreased in VPA-treated HL-60 cells (4-fold) and AML primary cells (2.5-fold). Our present data support previous findings that there is a relationship between the differentiation status of cells and their response to HDI such as VPA. We propose that very immature cells (CD34+ cells) respond to VPA by upregulation of CXCR4 (thereby enhancing homing responses towards an SDF-1α gradient); whereas more differentiated cells (AML blasts) downregulate CXCR4 resulting in their egress from the BM. Hence, we suggest that ex vivo priming of HSPC with HDI may improve their marrow engraftment. On the other hand, HDI may mobilize AML cells from the protective stromal microenvironment and render them more susceptible to chemotherapy.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 6592-6592
Author(s):  
W. Fiskus ◽  
M. Pranpat ◽  
M. Balasis ◽  
P. Atadja ◽  
P. Manley ◽  
...  

6592 Background: AMN107 has potent in vitro and in vivo activity against the un-mutated and most mutant forms of Bcr-Abl tyrosine kinase (TK). We have previously demonstrated that in CML cells the pan-histone deacetylase inhibitor LBH589 depletes both unmutated and mutated Bcr-Abl, as well as attenuates p-AKT and Raf-1 levels. In the present studies we determined the effects of AMN107 and/or LBH589 in unmutated or mutated Bcr-Abl-expressing mouse BaF3 cells, or human CML K562 and LAMA-84 cells, as well as in primary CML cells procured from patients with imatinib mesylate (IM)-refractory CML. Methods: Cultured and primary CML cells were exposed to 20 to 100 nM AMN107 and/or LBH589 for 24 to 48 hours. Following this, the % of apoptotic cells was determined, as well as immunoblot analyses were performed to determine Bcr-Abl, p-STAT5, p-AKT, p-CrkL, Bim, Bcl-xL and p27 levels. Synergy between the apoptotic effects was determined by the median dose-effect isobologram analysis. Results: AMN107 was 20-fold more potent than IM, and was more potent in inhibiting Bcr-Abl TK activity and attenuating p-STAT5, p-AKT, Bcl-xL, and c-Myc levels in K562 and LAMA-84 cells. Co-treatment with LBH589 and AMN107 exerted synergistic apoptotic effects against human CML (but not the normal bone marrow progenitor cells), with more attenuation of p-STAT5, p-ERK1/2, c-Myc and Bcl-xL and increased p27 and Bim levels. LBH589 attenuated Bcr-Abl levels and induced apoptosis of mouse pro-B BaF3 cells containing ectopic expression of unmutated Bcr-Abl or the IM-resistant Bcr-AblT315I and Bcr-AblE255K. Treatment with LBH589 also depleted Bcr-Abl levels and induced apoptosis in IM-resistant primary human CML cells, including those with expression of Bcr-AblT315I. As compared to either agent alone, co-treatment with AMN107 and LBH589 induced more loss of cell viability of primary IM-resistant CML cells. Conclusions: These findings support in vivo testing of the combination of LBH589 and AMN107 against IM naïve or resistant CML, with the goal to eradicate IM sensitive or resistant CML. [Table: see text]


Haematologica ◽  
2007 ◽  
Vol 92 (4) ◽  
pp. 542-545 ◽  
Author(s):  
G. Bug ◽  
K. Schwarz ◽  
C. Schoch ◽  
M. Kampfmann ◽  
R. Henschler ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document