Single Cell Network Profiling (SCNP) to Evaluate the Mechanism of Action of ON 01910.Na, A Novel Clinical Trial Stage Compound.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3827-3827 ◽  
Author(s):  
David M. Soper ◽  
Ying-Wen Huang ◽  
Francois Wilhelm ◽  
S. C. Cosenza ◽  
E. Premkumar Reddy ◽  
...  

Abstract Abstract 3827 Poster Board III-763 Background ON 01910.Na, a small molecule multikinase inhibitor, promotes G2/M arrest and apoptosis. Key targets for this inhibitor include Plk1 (polo-like kinase, a cell cycle regulator), Cdk1, (cyclin dependent kinase, a mitotic regulator) and the PI-3 kinase pathway (Ramana Reddy et al. J. Med. Chem. 2008, Park et al, Oncogene, 2007, Gumireddy et al., Cancer Cell, 2005). The drug has been shown to have anti-tumor activity in in vitro and in vivo models. Phase I studies in >100 advanced cancer patients revealed that the drug is well tolerated. Further, in several ongoing Phase 1 clinical trials in patients with myelodysplastic syndromes (MDS), positive effects on hematological indicators have been noted (Sloand et al, ASH 2008). Based on these data, a Phase 2 single-arm study is in progress to assess the efficacy and safety of the drug in IPSS Intermediate-2 and High risk MDS patients. Single Cell Network Profiling (SCNP) using flow cytometry is a platform that measures multiple fluorescent parameters (up to 10) in each cell, including both surface markers and intracellular signaling proteins in response to extracellular network inputs. By simultaneously measuring the effects of drug exposure on several pathways within each cell type in a heterogeneous patient tissue sample, valuable data can be gained about drug interactions with specific cellular pathways and cell type selectivity. This information has potential implications for dose/schedule optimization and development of patient stratification biomarkers. Objectives Studies were designed to evaluate the in vitro effects of ON 01910.Na, at clinically relevant concentrations, on intracellular pathways in the human GM-CSF-dependent erythroblastic TF-1 cell line using SCNP in order to monitor transitional changes in the cell cycle, with a focus on the G2-M phase and to perform dose-dependent titrations of drug using these cell cycle readouts. Methods The reagents chosen to measure cell cycle readouts were fluorochrome-conjugated antibodies that recognize cyclin B1, p-histone H3(S28) and p-Cdk1(Y15) and 4'6'-diamino-2-phenylindole (DAPI), a fluorescent dye that binds strongly to DNA. The phosphorylation status of p-histone H3(S28) and p-Cdk1(Y15), and the level of cyclin B1 expression are all determinants of the G2-M and/or M phase of the cell cycle. Dose dependent titrations of ON 01910.Na and its inactive analog ON 01911 were performed over a dose range starting at 10-5 M and decreasing to 10-10 M (dose range which includes pharmacologically achievable concentrations in humans) with 3-fold serial dilutions for eleven points after an exposure to the drug for either 24 or 48 hrs. Cells were processed for multiparameter flow cytometry by fixation, permeabilization and incubation with fluorchrome-conjugated antibodies. Results The data showed that at 24 hours after ON 01910.Na exposure there was a simultaneous increase in phosphorylation of histone H3(S28), a decrease in phosphorylation of Cdk-1(Y15), and accumulation of cyclin B1. These data suggest that ON 01910 exposure disrupted the G2/M cell cycle transition leading to mitotic arrest with subsequent apoptosis. TF-1 cell DNA content measured by DAPI verified this to be the case as increases in G2/M and sub-G1 (a measure of apoptotic cell death) were simultaneously observed. No significant effects on G2/M targets were observed when TF-1 cells were exposed to ON 01911, indicating the effects of ON 01910.Na on the cell cycle were specific to the drug. Maximal effects of ON-01910.Na on cell cycle signaling molecules were observed at a drug concentration of 0.37 mM and no further changes were seen at higher concentrations. These effects were also observed at 48 hours, although with more cell death. Conclusions These data indicate that intracellular phosphorylation changes of histone H3(S28) and Cdk-1(Y15), in addition to accumulation of cyclin B1 with subsequent apoptosis, reflect possible mechanisms of action of ON 01910.Na. The assay will be used in ongoing clinical trials to measure the pharmacodynamic activity of the drug in MDS patient samples pre- and post-treatment. Disclosures: Soper: Nodality Inc.: Employment, Equity Ownership. Huang:Nodality Inc.: Employment, Equity Ownership. Wilhelm:Onconova Therapeutics Inc: Employment. Cosenza:Onconova Therapeutics Inc.: Consultancy. Reddy:Onconova Therapeutics Inc.: Consultancy, Equity Ownership, Grantee, Membership on an entity's Board of Directors or advisory committees. Cesano:Nodality Inc.: Employment, Equity Ownership. Greenberg:Nodality Inc.: Research Funding; Onconova Therapeutics Inc.: Research Funding. Fantl:Nodality, Inc.: Employment, Equity Ownership.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4131-4131 ◽  
Author(s):  
Trinayan Kashyap ◽  
Irfana Muqbil ◽  
Amro Aboukameel ◽  
Boris Klebanov ◽  
Ramzi Mohammad ◽  
...  

Abstract Background: XPO1 (exportin-1/CRM1) mediates nuclear export of proteins containing leucine-rich amino-acid consensus sequences. XPO1 cargo proteins include many of the major tumor suppressor proteins (p53, IkB, pRB, FOXOs) and their export leads to the inactivation of cell cycle checkpoints. Overexpression of XPO1 has been reported to correlate with poor cancer prognosis. The Selective Inhibitor of Nuclear Export (SINE) compound, selinexor, binds covalently to the cargo pocket on XPO1, inhibits nuclear export which leads to cell cycle arrest and specific cancer cell death. Selinexor is currently in advanced clinical trials for patients with solid and hematological malignancies including patients with relapsed/refractory Diffuse Large B-Cell Lymphoma (DLBCL) (NCT02227251). Using preclinical models, we recently demonstrated that proteasome inhibitors (PI) can re-sensitize multiple myeloma that acquired resistance to selinexor. Here, we aimed to find if treatment with selinexor and bortezomib is beneficial for the treatment of DLBCL. Methods: DLBCLcell lines were treated with selinexor in combination with bortezomib. Cell viability was examined using standard viability assays after 72 hours of treatment. Whole cell protein lysates were evaluated by immunoblotting. NF-κB transcriptional activity was analyzed using an ELISA assay. WSU-DLCL2 cells were grown as sub-cutaneous tumors in ICR SCID mice. Tumor bearing mice were divided into 4 groups and were administered either vehicle, sub-maximum tolerated doses of selinexor (10 mg/kg p.o. twice a week, M, Th), bortezomib (1 mg/kg i.v. twice a week, M, TH) and the combination of selinexor (10 mg/kg p.o. twice a week) plus bortezomib (1 mg/kg i.v. twice a week). Results: The combination treatment of selinexor with bortezomib synergistically killed DLBCL cells compared to the single agents alone. Co-treatment with bortezomib enhanced selinexor mediated nuclear retention of IκB-α. Selinexor plus bortezomib treatment decreased NF-κB transcriptional activity. Finally, the combination of selinexor with bortezomib showed superior anti-tumor efficacy in the combination group compared to single agent treatments in WSU-DLCL2 xenograft model. Conclusions: Based on our results, inhibition of NF-κB transcriptional activity through forced nuclear retention of IκB appears to be an important mechanism underlying the synergistic effects of selinexor plus bortezomib in many different cell lines including DLBCL. The superior efficacy of selinexor plus bortezomib combination both in vitro and in vivo when compared to single agents along provides a rational for conducting clinical trials with these combinations in DLBCL patients. Disclosures Kashyap: Karyopharm Therapeutics: Employment, Equity Ownership. Klebanov:Karyopharm Therapeutics: Employment, Equity Ownership. Senapedis:Karyopharm Therapeutics: Employment, Equity Ownership. Shacham:Karyopharm Therapeutics: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Kauffman:Karyopharm Therapeutics Inc: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Landesman:Karyopharm Therapeutics: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4931-4931
Author(s):  
Monica Hellesøy ◽  
Katarzyna Wnuk-Lipinska ◽  
Anna Boniecka ◽  
Eline Milde Nævdal ◽  
Hakon Reikvam ◽  
...  

Abstract Axl is a member of the Tyro3, Axl, Mer (TAM) receptor tyrosine kinase family that regulate a wide range of cellular functions, including cell survival, proliferation, migration/invasion and adhesion. Axl has been shown to play a key role in the survival and metastasis of many tumors, and has also been found to be upregulated and constitutively active in human AML. Indeed, Axl has been reported as an independent prognostic marker and a potential novel therapeutic target in AML. BGB324 is a first-in-class highly selective small molecule inhibitor of Axl. BGB324 has been shown to be safe and well tolerated in clinical safety studies in healthy volunteers at doses up to 1500 mg/day with a predictable PK profile and long plasma half-life, and is currently in phase I b clinical trials for AML and non-small cell lung cancer. In this study, we use phosphoflow cytometry to measure changes in signal transduction nodes in single AML cells treated with BGB324. We are applying this approach to monitor signaling profiles in primary AML cells harvested from patients undergoing BGB324 treatment. Results: The human AML cell line MOLM13 was treated in vitro with BGB324 (0.5 and 1µM for 1 hour) and analyzed for signal transduction changes by phosphoflow cytometry. We found a significant reduction in phosphorylation of Axl (pY779), Akt(pS473), Erk1/2(pT202/Y204) and PLCɣ1(pY783). Next we established a systemic MOLM13 preclinical AML model in NOD/SCID mice. The mice were treated with 25 or 50 mg/kg BGB324 until moribund (up to 16 days). We found a dose-dependent and significant increase in overall survival in BGB324-treated mice. We further investigated intracellular signaling in BGB324-treated cells in vivo. Mice carrying systemic AML disease (MOLM13) were treated with BGB324 at 50mg/kg for 4 days, and we monitored CD33/45-positive MOLM13 cells harvested from spleen and bone marrow by flow cytometry. BGB324-treated mice showed a significant reduction in pErk and pPLCɣ1 relative to mice in the control group. PBMCs from peripheral blood of AML patients treated with BGB324 400 mg x1 at day 1 and 2, and thereafter 100 mg daily were collected for single cell signal profiling of signal transduction changes by conventional flow cytometry (phospho-flow) and mass cytometry (CyTOF). Preliminary phopho-flow analyses show decrease of pAkt(T308) and pPLCgamma1(Y783) in one patient. Further analyses are ongoing and will be presented. Figure 1. In vitro response to 1 hour BGB324 treatment in human AML cell line MOLM13 at 0.5 and 1µM doses. Response was evaluated in pAxl, pErk1/2, pAkt and pPLCγ1. n=3, *p≤0.05, **p≤0.005. Figure 1. In vitro response to 1 hour BGB324 treatment in human AML cell line MOLM13 at 0.5 and 1µM doses. Response was evaluated in pAxl, pErk1/2, pAkt and pPLCγ1. n=3, *p≤0.05, **p≤0.005. Figure 2. Dose-dependent response in overall survival in a MOLM13 systemic xenograft model (n=10). Figure 2. Dose-dependent response in overall survival in a MOLM13 systemic xenograft model (n=10). Figure 3. Response to BGB324-treatment in pErk, pPLCγ1 and pAkt in CD33/CD45-positive cells harvested from spleens (left) and bone marrows (right) of mice with systemic MOLM13 xenografts. n=5, *p≤0.05, **p≤0.005. Figure 3. Response to BGB324-treatment in pErk, pPLCγ1 and pAkt in CD33/CD45-positive cells harvested from spleens (left) and bone marrows (right) of mice with systemic MOLM13 xenografts. n=5, *p≤0.05, **p≤0.005. Disclosures Hellesøy: BerGenBio AS: Other: Previous employee. Stock option holder. Wnuk-Lipinska:BerGenBio AS: Employment. Boniecka:BerGenBio AS: Employment. Nævdal:BerGenBio AS: Employment. Loges:BerGenBio: Honoraria, Other: travel support, Research Funding. Cortes:Teva: Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; BerGenBio AS: Research Funding; Ariad: Consultancy, Research Funding; Astellas: Consultancy, Research Funding; Ambit: Consultancy, Research Funding; Arog: Research Funding; Celator: Research Funding; Jenssen: Consultancy. Lorens:BerGenBio AS: Employment, Equity Ownership. Micklem:BerGenBio AS: Employment, Equity Ownership. Gausdal:BerGenBio AS: Employment. Gjertsen:Haukeland University Hospital: Research Funding.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e16024-e16024
Author(s):  
Qingdi Quentin Li ◽  
Iawen Hsu ◽  
Thomas Sanford ◽  
Reema S. Railkar ◽  
Piyush K. Agarwal

e16024 Background: Protein Kinase D (PKD) is implicated in tumor growth, death, invasion, and progression. CRT0066101 is an inhibitor of PKD and has antitumor activity in several types of carcinomas. However, the effect and mechanism of CRT0066101 in bladder cancer remain unknown. Methods: The MTS assay was used to evaluate the ability of CRT0066101 to inhibit cellular proliferation in bladder cancer cells. Cell cycle was analyzed by flow cytometry. Protein expression and phosphorylation were assessed by western blotting. Results: We showed that CRT0066101 suppressed the proliferation and migration of 4 bladder cancer cell lines in vitro. We also demonstrated that CRT0066101 inhibited tumor growth in an in vivo mouse model of bladder cancer. To verify the role of PKD in bladder tumor, we found that PKD2 was highly expressed in 8 bladder cancer lines and that RNA interference-mediated silencing of the PKD2 gene dramatically reduced bladder cancer growth in vitro and in vivo, suggesting that the effect of the compound in bladder cancer is mediated through inhibition of PKD2. This notion was confirmed by demonstrating that the levels of PKD2 and phospho-PKD2 (Ser-876) were markedly decreased in CRT0066101-treated bladder cancer. In addition, our cell cycle analysis by flow cytometry revealed that CRT0066101 arrested bladder cancer cells at the G2-M phase. We further validated these data by immunoblotting showing that treatment of bladder carcinoma cells with CRT0066101 downregulated the expression of cyclin B1, cdc2 and cdc25C, but elevated the levels of p27kip1, gadd45a, chk1/2, and wee1. Finally, CRT0066101 was found to increase the phosphorylation of cdc2 and cdc25C, which lead to reduction in cdc2-cyclin B1 activity. Conclusions: These novel findings suggest that CRT0066101 inhibits bladder cancer growth through modulating the cell cycle G2 checkpoint and inducing cell cycle G2-M arrest, which lead to blockade of cell cycle progression. QQL and IH contributed equally to this work.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2753-2753
Author(s):  
Todd M. Covey ◽  
Michael Gulrajani ◽  
Heiko Becker ◽  
Jason C. Chandler ◽  
Sebastian Schwind ◽  
...  

Abstract Abstract 2753 Aberration in kinase activity by either the gain-of-function mutations or overexpression of the encoding genes plays a pivotal role in myeloid leukemogenesis. An increasing number of kinase inhibitors are being developed as “targeted therapies” for the treatment of acute myeloid leukemia (AML) and other myeloproliferative disorders. However, given the biologic and clinical heterogeneity inherent to these diseases, an unmet medical need exists for tools to guide the choice of inhibitor(s) most relevant for individual patients. With the aim of developing a platform for the biological characterization of patient-specific tumors, which could assist patient stratification strategies for clinical trials, we combined signaling pathway analysis and drug response profiling in AML samples using Single Cell Network Profiling (SCNP) assays. This technology allows for the simultaneous measurement of the activation state of multiple signaling proteins at the single cell level. Cryopreserved mononuclear cells from blood leukapheresis of patients with AML (N=6) were analyzed in two experimental arms. #1 Signaling Arm: A panel of kinase inhibitors targeting FLT3, cKit, PI3 kinase, mTor, MEK, and JAK proteins was added at varying concentrations to the AML cells followed by stimulation with G-CSF, IL-27, cKit ligand (SCF), FLT3 ligand (FLT3L), or a vehicle control. Using multiparameter flow cytometry, the phosphoylation status of AKT, ERK, S6 Ribosome, STAT1, STAT3, and STAT5 were measured in multiple leukemia cell subsets defined by expression of CD34, cKit, CD3, and light scatter properties. Per sample, there were a total of 68 treatments measuring 3 phospho-proteins in 3 cell subsets. #2 Apoptosis/Cytostasis Arm: The leukemic cells were driven into cell cycle by exposure to IL-3, SCF, and FLT3L, followed by a 48-hr incubation with a combination of 1 to 5 kinase inhibitors targeting the same pathways referred to previously. The kinase inhibitor impact was measured on distal functional readouts, including apoptosis (cleaved PARP) and cell cycle (CyclinB1-S/G2 phase; p-Histone H3-M phase). These results were compared with results using bone marrow samples from healthy donors (N=6). Results: Each patient's sample generated a unique signaling profile. A broad range of protein-specific phosphorylation status of AKT, ERK, S6 Ribosome, STAT1, STAT3, and STAT5 was observed in response to growth factor stimulation. Response was measured by setting a region gate that captures the overall percentage of cells with fluorescence above the unstimulated level. The percentage of SCF, G-CSF and FLT3L responsive cells ranged between 6%-49%, 3%-56%, and 3%-22%, respectively. Overall, patient samples could be grouped based on their signaling profile, proliferative potential, and sensitivity to kinase inhibitor treatment. Specifically, the 2 samples with the greatest SCF and G-CSF signaling response also showed the most robust in vitro proliferation and were most sensitive to the JAK inhibitor, CP-690,550 (1μM) (as measured by cytostasis readouts). Whereas, 2 other samples that displayed only modest SCF and G-CSF signaling, but robust FLT3L signaling expanded slowly in culture and were particularly sensitive to the cytostatic effects of the PI3K inhibitor, GDC-0941, (1uM) or the Flt3 inhibitor, tandutinib, (1uM). Finally, the last 2 AML samples had weak growth factor signaling and did not proliferate in culture and therefore could not be tested for kinase inhibitor-induced cytostasis. While the successfully tested patient samples showed variable sensitivity (as measured by cytostasis and apoptosis) to different drug combinations, the samples from healthy donors showed considerable similarity in response across all inhibitor combinations. Conclusions: This study provides preliminary proof-of-concept on the utility of SCNP to dissect the pathophysiologic heterogeneity of hematologic tumors and assess their differential response to single and combination therapies. Ultimately, this functional pathway profiling and drug sensitivity assay may be useful to stratify patients to different kinase combination treatments tested in clinical trials. Disclosures: Covey: Nodality Inc.: Employment, Equity Ownership. Gulrajani:Nodality Inc.: Employment, Equity Ownership. Cesano:Nodality Inc.: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3472-3472 ◽  
Author(s):  
Leona E Ling ◽  
Sucharita Roy ◽  
Thomas Daly ◽  
Edward Cochran ◽  
Steven Tyler ◽  
...  

Abstract Introduction IgG antibodies are the primary pathogenic agent in a number of auto- or allo-immune diseases. Efficacious therapies which decrease systemic levels of pathogenic antibodies include treatment with IVIG therapeutic plasmapheresis or immunoabsorption. Here, a novel strategy was evaluated to induce IgG clearance in diseases driven by IgG autoantibodies by blockade of FcRn-mediated IgG recycling. Methods M281 was developed as a high affinity, effectorless IgG1anti-FcRn monoclonal antibody. M281 effect on IgG recycling was evaluated in human umbilical vein endothelial cells in vitro. In vivo studies in transgenic human FCGRT/mouse FCGRT null mice and cynomolgus monkey were performed to characterize the pharmacokinetics, biodistribution, target occupancy, specificity of M281 and its efficacy in mouse models of thrombocytopenia and arthritis. Results M281 demonstrates specific dose-dependent, albumin-sparing IgG clearance in human FCGRT transgenic/mouse FCGRT null mice and in cynomolgus monkeys. M281 inhibits IgG recycling in endothelial cells in vitro and IgG clearance in vivo. Pharmacokinetics, target occupancy, pharmacodynamics and biodistribution indicate typical recombinant antibody biodistribution with rapid, dose dependent target inhibition and systemic clearance. M281 also demonstrated efficacy in mouse idiopathic thrombocytopenia purpura and collagen antibody-induced arthritis models of disease. Conclusions These findings support the evaluation of M281 as a strategy for the rapid and reversible suppression of pathogenic autoantibodies or alloantibodies in the setting of immune cytopenias, acquired inhibitors, thrombotic states and other disorders. Disclosures Ling: Momenta Pharmaceuticals: Employment, Equity Ownership. Roy:Momenta Pharmaceuticals: Employment, Equity Ownership. Daly:Momenta Pharmaceuticals: Employment, Equity Ownership. Cochran:Momenta Pharmaceuticals: Employment, Equity Ownership. Tyler:Momenta Pharmaceuticals: Employment, Equity Ownership. Markowitz:Momenta Pharmaceuticals: Employment, Equity Ownership. Bulik:Momenta Pharmaceuticals: Employment, Equity Ownership. Choudhury:Momenta Pharmaceuticals: Employment, Equity Ownership. Meador:Momenta Pharmaceuticals: Employment, Equity Ownership. Parge:Momenta Pharmaceuticals: Employment. Mekala:Momenta Pharmaceuticals: Employment, Equity Ownership. Sipsey:Momenta Pharmaceuticals: Employment, Equity Ownership. Gurnani:Momenta Pharmaceuticals: Employment, Equity Ownership. Duffner:Momenta Pharmaceuticals: Employment, Equity Ownership. Lee:Momenta Pharmaceuticals: Employment, Equity Ownership. Washburn:Momenta Pharmaceuticals: Employment, Equity Ownership. Meccariello:Momenta Pharmaceuticals: Employment, Equity Ownership. Schaeck:Momenta Pharmaceuticals: Employment, Equity Ownership. Wang:Momenta Pharmaceuticals: Employment, Equity Ownership. Schultes:Momenta Pharmaceuticals: Employment, Equity Ownership. Hillson:Momenta Pharmaceuticals: Employment, Equity Ownership. Avery:Momenta Pharmaceuticals: Employment, Equity Ownership. Kaundinya:Momenta Pharmaceuticals: Employment, Equity Ownership. Manning:Momenta Pharmaceuticals: Employment, Equity Ownership.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 607-607 ◽  
Author(s):  
Emma M Smith ◽  
Brian A Walker ◽  
Emma L Davenport ◽  
Lauren I Aronson ◽  
David Krige ◽  
...  

Abstract Abstract 607 Myeloma cells rely on a number of mechanisms to maintain their survival, including the silencing of genes that would normally check uncontrolled proliferation and lead to apoptosis. Epigenetic alterations such as histone deacetylation contribute significantly to the pathogenesis of both solid and haematological malignancies and are associated with the silencing of tumour suppressor genes. Pan-histone deacetylase inhibitors (HDACs), such as SAHA, panobinostat, depsipeptide, and numerous others have been published to have anti-myeloma activity. Here we report the effects of a novel pan HDAC inhibitor, CHR-3996, on multiple myeloma cells. Results: CHR-3996 was potently cytotoxic against a panel of myeloma cell lines (H929, KMS11, LP-1, MM1s, RPMI-8226, and U266) with a GI50 ranging from 9 to 65 nM and was equally as potent in primary patient myeloma cells. The loss of cell viability was associated with an increase in apoptotic cells; EndonucleaseG and Noxa were both up-regulated and caspase 9 was cleaved. Furthermore a decrease in apoptosis was demonstrated in the presence of a pan-caspase inhibitor indicating cell death is largely dependent on caspase-mediated apoptosis. There was no effect on bone marrow stromal (BMS) cell viability but there was an observed decrease in the IL-6 and VEGF the BMS cells secrete, both of which promote the growth and survival of myeloma cells in the bone marrow microenvironment. CHR-3996 caused an increase in acetylated H3K9 but there was minimal change in the levels of ubiquitinated proteins in the cell or acetylated alpha-tubulin, indicating low activity against HDAC6 or the proteasome (also demonstrated by an assay to specifically measure proteasome function). In addition to induction of apoptosis, cell cycle analysis showed an increased proportion of cells in G0/G1 indicating cell cycle arrest. In-keeping with data from treating myeloma cells with SAHA and panobinostat, an increase in cell cycle inhibitor p21 was observed. Gene expression profiling (GEP) identified changes in cell cycle regulators, indications of increased cell stress (elevated CHOP, ATF3, and TAO kinase 3), repression of Wnt (up-regulation of NLK, GSK3beta) and mTOR (decreased 4E-BP1) signalling, and changes in key pro- and anti-apoptotic proteins (for example SMAD3, BCL2, BIM, BID, and BIRC5). When used in combination studies CHR-3996 was highly synergistic in vitro with tosedostat, an aminopeptidase inhibitor, which we have previously shown to have anti-myeloma activity via the induction of the amino acid deprivation response and autophagy. One of the largest changes the GEP analysis identified was BIRC3 (CIAP2), an inhibitor of NF-kappaB signalling, which increased 23.5 fold with CHR-3996 as a single agent and over 100-fold when added to H929 cells in combination with tosedostat. Additionally both CHR-3996 and todestat independently up-regulated expression of members of the IkappaB family. Altered expression and nuclear localisation of canonical and non-canonical NF-kappaB family members were observed by immuno-fluorescence and immunoblotting, suggesting targeting of NF-kappaB signalling as a reason for the high degree of synergy between these two compounds. Early data suggest oral CHR-3996 is effective in a subcutaneous in vivo myeloma model and, reflecting the in vitro data, there is a degree of synergy when administered with tosedostat. Conclusions: The novel compound CHR-3996 is a potent HDAC inhibitor that leads to increased H3K9 acetylation but has no detectable activity against HDAC6 or proteasome activity in myeloma. It induces apoptosis of myeloma cells without affecting BMS cell viability. CHR-3996 shows a very high degree of synergy with an aminopeptidase inhibitor, tosedostat, potentially through targeting the NF-kappaB pathway. It has exciting therapeutic potential either as a mono-therapy or in combination with other agents. Disclosures: Krige: Chroma Therapeutics Ltd: Employment, Equity Ownership. Hooftman:Chroma Therapeutic Ltd: Employment, Equity Ownership. Drummond:Chroma therapuetics: Employment, Equity Ownership.


Circulation ◽  
2018 ◽  
Vol 138 (Suppl_1) ◽  
Author(s):  
Ruya Liu ◽  
Rajaganapathi Jagannathan ◽  
Feng Li ◽  
Jeongkyung Lee ◽  
Vijay K Yechoor ◽  
...  

Introduction: Mammalian cardiomyocyte (CM) proliferation peaks in the embryonic and neonatal periods. TEAD1, a key transcription factor regulated by the Hippo pathway, is critical for early embryonic CM proliferation. But mid gestation lethality of Tead1 germline deletion precluded the study of its role in CMs at later developmental stages. We recently generated Tead1 floxed (Tead1 F/F ) mice which allows the study of TEAD1 function in CMs at later stages. The objective of this study was to determine requirement of TEAD1 for neonatal CM proliferation. Hypothesis: TEAD1 remains critical for CM proliferation in late embryonic and early neonatal periods through transcriptional regulation of cell cycle promoting genes. Methods and Results: We observed that TEAD1 cardiac expression peaks in the perinatal period. Using Myh6-Cre deletor mice, we knocked out Tead1 in CMs at E10.5 (referred as cKO). cKO pups were born in expected Mendelian frequency, but survived only till day of life (DOL) 9. Systolic dysfunction was evident by ECHO in DOL1 cKO pups and progressed to frank heart failure (HF) by DOL9. Histological exam showed decreased myocardial mass with increased intercellular fibrosis. Ventricles of DOL1 cKO pups demonstrated increased expression of Acta1, Nppa, and Nppb, consistent with HF but showed decreased expression of Myh7, suggesting an impairment in the typical fetal gene program activated in HF. Myocardial immunostaining showed reduction in Ki67 (G1/S/G2/M phase marker) (Fig 1) and PH3-S10 (M phase marker) positive CMs by 82% and 46% respectively in DOL1 cKO hearts, indicating significantly reduced CM proliferation. The expression of essential cell cycle proteins showed a significant decrease in the levels of G1/S regulating proteins, CDK4, CDK6, ppRB S807/811 and S/G2 and G2/M regulating proteins, pWEE1 S642 and Cyclin B1 in cKO hearts (Fig 2). Similar results in ex vivo and in vitro Tead1 knockout models in CMs using neonatal Tead1 F/F CMs and HL1 cells validated the cell autonomous regulation of CM cell cycle by TEAD1. Conclusions: TEAD1 is required for embryonic and neonatal CM proliferation and its loss at mid gestation leads to neonatal HF associated with impaired fetal gene program activation and decreased expression of cell cycle promoting genes.


2021 ◽  
Vol 12 ◽  
Author(s):  
Tai-Hsin Tsai ◽  
Ann-Shung Lieu ◽  
Tzuu-Yuan Huang ◽  
Aij-Lie Kwan ◽  
Chih-Lung Lin ◽  
...  

Background: Glioblastoma multiforme (GBM) is the vicious malignant brain tumor in adults. Despite advances multi-disciplinary treatment, GBM constinues to have a poor overall survival. CDDO-trifluoroethyl-amide (CDDO-TEFA), a trifluoroethylamidederivative of CDDO, is an Nrf2/ARE pathway activator. CDDO-TEFEA is used to inhibit proliferation and induce apoptosis in glioma cells. However, it not clear what effect it may have on tumorigenesis in GBM.Methods: This in vitro study evaluated the effects of CDDO-TFEA on GBM cells. To do this, we treated GBM8401 cell lines with CDDO-TFEA and assessed apoptosis, cell cycle. DNA content and induction of apoptosis were analyzed by flow cytometry and protein expression by Western blot analysis.Results: CDDO-TFEA significantly inhibited the cell viability and induced cell apoptosis on GBM 8401 cell line. The annexin-FITC/PI assay revealed significant changes in the percentage of apoptotic cells. Treatment with CDDO-TFEA led to a significant reduction in the GBM8401 cells’ mitochondrial membrane potential. A significant rise in the percentage of caspase-3 activity was detected in the treated cells. In addition, treatment with CDDO-TFEA led to an accumulation of G2/M-phase cells. In addition, these results suggest that regarding increased protein synthesis during mitosis in the MPM-2 staining, indicative of a delay in the G2 checkpoint. An analysis of Cyclin B1, CDK1, Cyclin B1/CDK1 complex and CHK1 and CHK2 expression suggested that cell cycle progression seems also to be regulated by CDDO-TFEA. Therefore, CDDO-TFEA may not only induce cell cycle G2/M arrest, it may also exert apoptosis in established GBM cells.Conclusion: CDDO-TFEA can inhibit proliferation, cell cycle progression and induce apoptosis in GBM cells in vitro, possibly though its inhibition of Cyclin B1, CDK1 expression, and Cyclin B1/CDK1 association and the promotion of CHK1 and CHK2 expression.


2011 ◽  
Vol 2011 ◽  
pp. 1-10 ◽  
Author(s):  
Ting-Tsz Ou ◽  
Chau-Jong Wang ◽  
Guang-Uei Hung ◽  
Cheng-Hsun Wu ◽  
Huei-Jane Lee

Shi-Liu-Wei-Liu-Qi-Yin (SLWLQY) was traditionally used to treat cancers. However, scientific evidence of the anticancer effects still remains undefined. In this study, we aimed to clarify the possible mechanisms of SLWLQY in treating cancer. We evaluated the effects of SLWLQY on apoptosis-related experiments inducing in TSGH-8301 cells by (i) 3-(4,5-dimethylthiazol-zyl)-2,5-diphenylterazolium bromide (MTT) for cytotoxicity; (ii) cell-cycle analysis and (iii) western blot analysis of the G2/M-phase and apoptosis regulatory proteins. Human bladder carcinoma TSGH-8301 cells were transplanted into BALB/c nude mice as a tumor model for evaluating the antitumor effect of SLWLQY. Treatment of SLWLQY resulted in the G2/M phase arrest and apoptotic death in a dose-dependent manner, accompanied by a decrease in cyclin-dependent kinases (cdc2) and cyclins (cyclin B1). SLWLQY stimulated increases in the protein expression of Fas and FasL, and induced the cleavage of caspase-3, caspase-9 and caspase-8. The ratio of Bax/Bcl2was increased by SLWLQY treatment. SLWLQY markedly reduced tumor size in TSGH-8301 cells-xenografted tumor tissues. In the tissue specimen, SLWLQY up-regulated the expression of Fas, FasL and Bax proteins, and down-regulated Bcl2as well as inin vitroassay. Our results showed that SLWLQY reduced tumor growth, caused cell-cycle arrest and apoptosis in TSGH-8301 cells via the Fas and mitochondrial pathway.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2745-2745
Author(s):  
David B. Rosen ◽  
James A Cordeiro ◽  
David M. Soper ◽  
Ying-Wen Huang ◽  
Donna E. Hogge ◽  
...  

Abstract Abstract 2745 Poster Board II-721 Background: Gemtuzumab Ozogamicin (GO, Mylotarg), a humanized CD33 monoclonal antibody linked to calicheamicin was approved by the US FDA for use as a monotherapy in patients older than 60 years with relapsed acute myeloid leukemia (AML) unfit to tolerate standard salvage therapy. GO is internalized rapidly after infusion, and calicheamicin, a potent enediyene, is subsequently released and acts as a cytotoxic agent by causing double strand DNA breaks. Currently GO is in multiple clinical trials as a single agent or in combination with other therapies for both induction and consolidation treatment of various clinical subgroups of AML. However, the mechanisms of action and resistance of GO are incompletely understood and it is unclear which patient subgroups benefit from GO-based therapy. Single cell network profiling (SCNP) has shown promise as a methodology wherein multiple signaling networks are measured after treatment with an exogenous modulator such as a growth factor, cytokine or therapeutic agent and the identified signaling profiles can be used as clinical and therapeutic enablement tools. Objectives: SCNP using multiparameter flow cytometry was used to identify intracellular pathways that were associated with responsiveness or refractoriness to in vitro GO exposure in both cancer cell lines and primary AML samples. Methods: Signaling pathways emphasizing DNA damage response, cell cycle, apoptosis and drug transporter activity were measured by SCNP after in vitro exposure of cell lines and AML primary samples to clinically relevant concentrations of GO. Samples were processed for cytometry by paraformaldehyde /methanol fixation and permeabilzation followed by incubation with fluorochrome-conjugated antibody cocktails that recognize cell surface proteins to delineate cell subsets and intracellular signaling molecules. Results: In cell lines, responsiveness to in vitro GO exposure was defined as a) induction of DNA Damage as measured by increased p-ATM, p-Chk2 and p-H2AX, b) cell cycle arrest at G2/M as measured by increased cyclin B1 and DNA content & c) induction of apoptosis as measured by cleaved PARP and viability dyes. Of note, inhibition of drug transporter activity in 2 MDR-1+ cell lines did not restore GO responsiveness, suggesting the presence of additional relevant resistance mechanisms in these cell lines. In primary AML diagnostic samples, DNA damage and apoptosis pathway readouts were able to identify responsiveness or refractoriness to GO exposure. In the GO responsive profile, induction of both DNA damage responses and apoptosis were seen. Within the refractory samples, two distinct profiles were observed: a) robust and early induction of DNA damage response without apoptosis and 2) delayed and attenuated DNA damage response without apoptosis. Conclusions: Characterization of intracellular Cell Cycle, DNA Damage, and Apoptosis networks in single cells after GO exposure distinguishes GO responsive from refractory AML cells. Further, these pathway signatures provide information about mechanisms of refractoriness. (e.g. a block between a successful DNA damage response and initiation of apoptosis versus a block in the initial induction of DNA damage after GO exposure). The ability of the same profiles to predict clinical responses to the drug will be tested in future studies. Disclosures: Rosen: Nodality, Inc.: Employment, Equity Ownership. Cordeiro:Nodality Inc.: Employment, Equity Ownership. Soper:Nodality Inc.: Employment, Equity Ownership. Huang:Nodality Inc.: Employment, Equity Ownership. Cesano:Nodality Inc.: Employment, Equity Ownership. Fantl:Nodality Inc.: Employment, Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document