Anti-Leukemia Effect of FF-10501-01, a Novel Inosine 5'-Monophosphate Dehydrogenase Inhibitor, in Acute Myeloid Leukemia

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2756-2756
Author(s):  
Hui Yang ◽  
Zachary S. Bohannan ◽  
Zhihong Fang ◽  
Irene Gañán-Gómez ◽  
Yue Wei ◽  
...  

Abstract Introduction Inosine 5'- monophosphate dehydrogenase (IMPDH) plays a critical role in nucleotide synthesis by serving as a rate-limiting step for the de novo production of guanine from its precursors. Overexpression of IMPDH has been observed in both solid and hematologic malignancies. FF-10501-01 is a potent new competitive IMPDH inhibitor. In this study, we systematically investigated the anti-leukemia effect of FF-10501-01 in acute myeloid leukemia (AML) cell lines, including hypomethylating agent (HMA)-resistant derivative cells. Methods Thirteen leukemia cell lines were studied, including 5 parental AML cell lines and their HMA-resistant derivatives (MOLM13, SKM1, HL60, TF1, and U937) as well as three other AML cell lines (KG1, HEL, and OCI-AML3). Cell proliferation was determined using trypan blue analysis. Flow cytometry was performed to detect drug-induced apoptosis and cell cycle status. High-performance liquid chromatography (HPLC) was performed to detect the intracellular concentration of guanine nucleotides, with mycophenolic acid (MPA) treated cells used as positive control. We also studied the effect of guanosine supplementation on FF-10501-01-treated cells. Results To understand whether FF-10501-01 was able to effectively limit AML cell proliferation, we subjected a variety of cell lines to 72 hours of FF-10501-01 treatment at various concentrations. We also included a large number of HMA-resistant cell lines in an effort to understand whether or not FF-10501-01 could be a useful secondary or complimentary treatment to HMA therapy. FF-10501-01 inhibited the proliferation of all 13 AML cell lines studied with 72 hours of treatment. The IC-50 of FF-10501-01 ranged between 4.3 and 144.5 µM. The IC50 values for HMA-resistant cells were all higher than those values in their HMA-sensitive counterparts, except SKM1, in which the HMA-sensitive line had a higher IC50 than the HMA-resistant SKM1 line. To further understand the mechanism by which FF-10501-01 effectively reduced cell numbers in these cell lines, we assessed the level of apoptosis in each line after FF-10501-01 exposure. FF-10501-01-induced apoptosis was observed in all of the studied cell lines in a dose-dependent manner except for the HMA-resistant TF-1 cell line. We then assessed whether FF-10501-01 affected cell cycle progression. This effect was highly variable. Increased numbers of cells in G1 phase and decreased numbers of cells in S phase were observed in MOLM13, SKM1, and TF-1 cell lines treated with less than 100 µM FF-10501-01. To understand the mechanistic effects of FF-10501-01, we performed rescue experiments with both HMA-resistant and HMA-sensitive MOLM13 and HL60 cells. Concurrent treatment with FF-10501-01 and guanosine in these cells partially rescued the antiproliferation effect of FF-10501-01. To further characterize the effect of FF-10501-01 on guanosine metabolism, we then performed HPLC experiments to analyze the levels of phosphoguanosine in treated MOLM13 and SKM1 cells. FF-10501-01 treatment effectively reduced the intracellular phosphoguanosine levels in both cell lines. This effect was seen for GMP, GDP, and GTP. We then sought to assess whether the combination of FF-10501-01 and HMAs could be effective in limiting MOLM13 and HL-60 cell proliferation, especially in their HMA resistant derivatives. The combination of HMA and FF-10501-01 showed little synergy beyond the effects of FF-10501-01 alone, regardless of HMA sensitivity, except in HMA-resistant HL-60 cells, in which FF-10501-01 showed moderate synergy with HMA. We further assessed the antiproliferative effect of FF-10501-01 in bone marrow blast samples taken from 3 AML patients. There was a minor dose-dependent antiproliferation effect seen in these samples, but this was not statistically significant. Notably, one patient showed a sharp increase in cell counts at the lowest concentration of FF-10501-01, but that sample's cell numbers decreased more rapidly as FF-10501-01 concentration increased. This implies that FF-10501-01 treatment response may be related to cell proliferation. Conclusions The IMPDH inhibitor FF-10501-01 can produce potent anti-proliferative and apoptotic induction effects on AML cell lines, including HMA-resistant cell lines, through inhibition of de novo guanine nucleotide synthesis. These results indicate that FF-10501-01 might be a promising new therapeutic agent for AML. Disclosures Paradiso: Strategia Therapeutics, Inc.: Employment. Iwamura:FUJIFILM Corporation: Employment.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-9
Author(s):  
Sujan Piya ◽  
Marla Weetall ◽  
Josephine Sheedy ◽  
Balmiki Ray ◽  
Huaxian Ma ◽  
...  

Introduction: Acute myeloid leukemia (AML) is characterized by both aberrant proliferation and differentiation arrest at hematopoietic progenitor stages 1,2. AML relies upon de novo nucleotide synthesis to meet a dynamic metabolic landscape and to provide a sufficient supply of nucleotides and other macromolecules 3,4. Hence, we hypothesized that inhibition of de novo nucleotide synthesis would lead to depletion of the nucleotide pool and pyrimidine starvation in leukemic cells compared to their non-malignant counterparts and impact proliferative and differentiation inhibition pathways. PTC299 is an inhibitor of dihydroorotate dehydrogenase (DHODH), a rate limiting enzyme for de novo pyrimidine nucleotide synthesis that is currently in a clinical trial for the treatment of AML. Aim: We investigated the pre-clinical activity of PTC299 against AML in primary AML blasts and cytarabine-resistant cell lines. To confirm that PTC299 effects are due to inhibition of de novo pyrimidine nucleotide synthesis for leukemic growth, we specifically tested the impact of uridine and orotate rescue. In addition, a comprehensive analysis of alteration of metabolic signaling in PI3K/AKT pathways, apoptotic signatures and DNA damage responses were analyzed by Mass cytometry based proteomic analysis (CyTOF) and immunoblotting. The potential clinical relevance of DHODH inhibition was confirmed in an AML-PDX model. Results: The IC50s for all tested cell lines (at 3 day) and primary blasts (at 5-7 day) were in a very low nanomolar range: OCI-AML3 -4.43 nM, HL60 -59.7 nM and primary samples -18-90 nM. Treatment of AML in cytarabine-resistant cells demonstrated that PTC299 induced apoptosis, differentiation, and reduced proliferation with corresponding increase in Annexin V and CD14 positive cells (Fig.1). PTC299-induced apoptosis and inhibition of proliferation was rescued by uridine and orotate. To gain more mechanistic insights, we used an immunoblotting and mass cytometry (CyTOF) based approach to analyze changes in apoptotic and cell signaling proteins in OCI-AML3 cells. Apoptotic pathways were induced (cleaved PARP, cleaved Caspase-3) and DNA damage responses (TP53, γH2AX) and the PI3/AKT pathway were downregulated in response to PTC299. In isogenic cell lines, p53-wildtype cells were sustained and an increased DNA damage response with corresponding increase in apoptosis in comparison to p53-deficient cells was shown. (Fig.2) In a PDX mouse model of human AML, PTC299 treatment improved survival compared to mice treated with vehicle (median survival 40 days vs. 30 days, P=0.0002) (Fig.3). This corresponded with a reduction in the bone marrow burden of leukemia with increased expression of differentiation markers in mice treated with PTC299 (Fig.3). Conclusion: PTC299 is a novel dihydroorotate dehydrogenase (DHODH) inhibitor that triggers differentiation, apoptosis and/or inhibition of proliferation in AML and is being tested in a clinical trials for the treatment of acute myeloid malignancies. Reference: 1. Thomas D, Majeti R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017; 129(12): 1577-1585. e-pub ahead of print 2017/02/06; doi: 10.1182/blood-2016-10-696054 2. Quek L, Otto GW, Garnett C, Lhermitte L, Karamitros D, Stoilova B et al. Genetically distinct leukemic stem cells in human CD34- acute myeloid leukemia are arrested at a hemopoietic precursor-like stage. The Journal of experimental medicine 2016; 213(8): 1513-1535. e-pub ahead of print 2016/07/06; doi: 10.1084/jem.20151775 3. Villa E, Ali ES, Sahu U, Ben-Sahra I. Cancer Cells Tune the Signaling Pathways to Empower de Novo Synthesis of Nucleotides. Cancers (Basel) 2019; 11(5). e-pub ahead of print 2019/05/22; doi: 10.3390/cancers11050688 4. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv 2016; 2(5): e1600200. e-pub ahead of print 2016/07/08; doi: 10.1126/sciadv.1600200 Disclosures Weetall: PTC Therapeutic: Current Employment. Sheedy:PTC therapeutics: Current Employment. Ray:PTC Therapeutics Inc.: Current Employment. Konopleva:Genentech: Consultancy, Research Funding; Rafael Pharmaceutical: Research Funding; Ablynx: Research Funding; Ascentage: Research Funding; Agios: Research Funding; Kisoji: Consultancy; Eli Lilly: Research Funding; AstraZeneca: Research Funding; Reata Pharmaceutical Inc.;: Patents & Royalties: patents and royalties with patent US 7,795,305 B2 on CDDO-compounds and combination therapies, licensed to Reata Pharmaceutical; AbbVie: Consultancy, Research Funding; Calithera: Research Funding; Cellectis: Research Funding; Amgen: Consultancy; Stemline Therapeutics: Consultancy, Research Funding; Forty-Seven: Consultancy, Research Funding; F. Hoffmann La-Roche: Consultancy, Research Funding; Sanofi: Research Funding. Andreeff:Amgen: Research Funding; Daiichi-Sankyo; Jazz Pharmaceuticals; Celgene; Amgen; AstraZeneca; 6 Dimensions Capital: Consultancy; Daiichi-Sankyo; Breast Cancer Research Foundation; CPRIT; NIH/NCI; Amgen; AstraZeneca: Research Funding; Centre for Drug Research & Development; Cancer UK; NCI-CTEP; German Research Council; Leukemia Lymphoma Foundation (LLS); NCI-RDCRN (Rare Disease Clin Network); CLL Founcdation; BioLineRx; SentiBio; Aptose Biosciences, Inc: Membership on an entity's Board of Directors or advisory committees. Borthakur:BioLine Rx: Consultancy; BioTherix: Consultancy; Nkarta Therapeutics: Consultancy; Treadwell Therapeutics: Consultancy; Xbiotech USA: Research Funding; Polaris: Research Funding; AstraZeneca: Research Funding; BMS: Research Funding; BioLine Rx: Research Funding; Cyclacel: Research Funding; GSK: Research Funding; Jannsen: Research Funding; Abbvie: Research Funding; Novartis: Research Funding; Incyte: Research Funding; PTC Therapeutics: Research Funding; FTC Therapeutics: Consultancy; Curio Science LLC: Consultancy; PTC Therapeutics: Consultancy; Argenx: Consultancy; Oncoceutics: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3253-3253
Author(s):  
Kevin R Kelly ◽  
Kelli Oberheu ◽  
Ernest Medina ◽  
Claudia M Espitia ◽  
Devalingam Mahalingam ◽  
...  

Abstract Abstract 3253 Drug resistance is a major cause of treatment failure for patients with acute myeloid leukemia (AML) and novel strategies that circumvent resistance mechanisms are urgently needed. The Pim kinases (PIM1, PIM2, PIM3) are a small family of proto-oncogenes within the CAMK superfamily with essential roles in the regulation of signal transduction cascades that promote cell survival, proliferation, and drug resistance. Accordingly, overexpression of Pim kinases has been reported in a wide range of malignancies including AML. However, the specific role of Pim kinases as regulators of sensitivity to standard agents utilized in AML therapy remains to be elucidated. We hypothesized that inhibiting Pim kinase activity would significantly increase the efficacy of the standard of care drug cytarabine by disrupting signaling events that enable cells to resist cytotoxic stress. In order to investigate a potential role for Pim kinases as mediators of cytarabine resistance, we first evaluated their expression levels in paired HL-60 cell lines that are sensitive and resistant to cytarabine. Our results showed that the levels of Pim-1 are significantly higher in cytarabine-resistant HL-60 cells compared with their sensitive counterparts. Consistent with this observation, treatment of cultured AML cell lines and primary AML blasts with cytarabine led to a dose-dependent increase in Pim-1 expression as assessed by quantitative RT-PCR and immunoblotting. SGI-1776 is novel small molecule inhibitor of Pim kinase activity that has entered Phase I clinical trials. To further test our hypothesis, we investigated the in vitro efficacy of SGI-1776 in a panel of 9 human AML cell lines. SGI-1776 led to a dose-dependent reduction in viability in all cell lines evaluated. Subsequent experiments showed that SGI-1776 impaired clonogenic survival, diminished cell proliferation as determined by BrdU incorporation, and induced apoptotic cell death. These effects were associated with a significant reduction in the phosphorylation of the Pim kinase substrate and apoptotic regulator Bad as well as an increase in the expression of the cyclin-dependent kinase inhibitor, p27. We next investigated whether inhibiting Pim signaling with SGI-1776 could augment the efficacy of cytarabine. Treatment of AML cells with the combination of cytarabine and SGI-1776 led to significantly greater inhibition of clonogenic survival and induction of apoptosis over what was achieved by either single agent. To validate the therapeutic utility of this combination, vehicle, cytarabine, SGI-1776, or cytarabine and SGI-1776 were administered to mice bearing MOLM-13 AML xenografts for 21 days. Treatment with the combination of these two agents was very well tolerated and significantly increased the efficacy of single agent cytarabine therapy. Immunohistochemical analyses of specimens from mice treated with this regimen revealed that SGI-1776 diminished Bad phosphorylation and cooperated with cytarabine in vivo to promote activation of caspase-3 and inhibit tumor cell proliferation as quantified by the expression of proliferating cell nuclear antigen (PCNA). Collectively, our data demonstrate that antagonizing Pim kinase activity represents a new strategy to increase the therapeutic efficacy of cytarabine. A clinical trial evaluating the safety and efficacy of this combination for patients with AML that are refractory to standard treatments is warranted. Disclosures: Taverna: SuperGen, Inc.: Employment. Choy:SuperGen, Inc.: Employment.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 50-50
Author(s):  
Kana Nakatani ◽  
Hidemasa Matsuo ◽  
Yutarou Harata ◽  
Moe Higashitani ◽  
Asami Koyama ◽  
...  

Acute myeloid leukemia (AML) is a genetically and clinically heterogeneous disease. Although t(8;21) AML patients have a more favorable prognosis than other cytogenetic subgroups, nearly 40% of t(8;21) AML patients experience relapse. Therefore, novel therapeutic approaches based on a better understanding of the biology of t(8;21) AML need to be developed. In this study, at first, we re-analyzed the sequencing data of 149 pediatric t(8;21) AML patients from St. Jude Children's Research Hospital tissue resource core facility and the JPLSG AML-05 study, and 134 adult t(8;21) AML patients from CALGB/Alliance trials and the University Hospital of Ulm. In pediatric patients, 13 CCND2 mutations were detected in 11 patients (11/149, 7.4%), and in adult patients, 14 CCND2 mutations were detected in 12 patients (12/134, 9.0%). In both cohorts, CCND2 mutations were located on the PEST domain, suggesting that the mutations stabilize the cyclin D2 protein. Next, we compared CCND2 mRNA expression between t(8;21) AML patients (n=24) and non-t(8;21) AML patients (n=163) using the TARGET AML cohort. In non-t(8;21) AML patients, CCND2 expression varied from low to high levels, whereas in t(8;21) AML patients, CCND2 expression was restricted to higher levels. Consistently, CCND2 expression was higher in t(8;21) AML cell lines (n=2: Kasumi-1 and SKNO-1), compared with non-t(8;21) AML cell lines (n=32). Kasumi-1 cells transfected with shCCND2 showed cell cycle arrest at G1 phase and impaired cell proliferation. These results suggest that the frequency of CCND2 mutations and CCND2 expression are increased in t(8;21) AML, and high CCND2 expression plays an important role in t(8;21) AML cell proliferation. Because CCND2 is not a druggable target, we examined the effect of CDK4/6 inhibitors (palbociclib and abemaciclib) on t(8;21) AML cells. Analysis of 19 AML cell lines showed that t(8;21) AML cells had lower IC50 values for CDK4/6 inhibitors than non-t(8;21) AML cells. CDK4/6 inhibitors caused cell cycle arrest at G1 phase and impaired cell proliferation in t(8;21) AML cells. To identify potential therapeutic approaches in combination with CDK4/6 inhibitors in t(8;21) AML, we performed microarray analysis and examined the effects of CDK4/6 inhibition. In addition to the pathways associated with the cell cycle (regulation of sister chromatid separation, retinoblastoma gene, and cell cycle), the MAP-ERK and PI3K-AKT-mTOR signaling pathways were downregulated by CDK4/6 inhibition. Because these pathways are involved in autophagy regulation via mTOR, we focused on examining autophagy in subsequent experiments. Assessment of the effect of CDK4/6 inhibition on autophagy in t(8;21) AML cells showed that the CDK4/6 inhibitor (abemaciclib) treatment induced LC3B-I to LC3B-II conversion in both Kasumi-1 and SKNO-1 cells. Transmission electron microscopic examination of autophagosome formation detected a large number of autophagosomes in the cytoplasm of Kasumi-1 and SKNO-1 cells treated with abemaciclib, whereas few autophagosomes were detected in control samples. These results suggest that autophagy is induced by CDK4/6 inhibition in t(8;21) AML cells. Autophagy is involved in the resistance to chemotherapy in cancer cells, therefore, we hypothesized that autophagy inhibition may be a promising therapeutic approach. Treatment of t(8;21) AML cells with the autophagy inhibitors chloroquine (CQ) or LY294002 in combination with abemaciclib significantly increased the frequency of apoptotic (Annexin V positive) cells compared with that in untreated cells, whereas CQ or LY294002 single treatment had no significant effect on apoptosis. Consistently, combinatorial inhibition of CDK4/6 and autophagy upregulated cleaved caspase 3 expression. The combinatorial effect was confirmed by silencing the autophagy-related protein ATG7 using small interfering RNA in abemaciclib-treated t(8;21) AML cells. These results suggest that autophagy inhibition enhances CDK4/6 inhibitor-induced apoptosis in t(8;21) AML cells. In conclusion, the present results indicate that inhibition of CDK4/6 and autophagy may be a novel and promising biomarker-driven therapeutic strategy for the treatment of t(8;21) AML. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (5) ◽  
pp. 1003
Author(s):  
Valentina Gaidano ◽  
Mohammad Houshmand ◽  
Nicoletta Vitale ◽  
Giovanna Carrà ◽  
Alessandro Morotti ◽  
...  

Dihydroorotate Dehydrogenase (DHODH) is a key enzyme of the de novo pyrimidine biosynthesis, whose inhibition can induce differentiation and apoptosis in acute myeloid leukemia (AML). DHODH inhibitors had shown promising in vitro and in vivo activity on solid tumors, but their effectiveness was not confirmed in clinical trials, probably because cancer cells exploited the pyrimidine salvage pathway to survive. Here, we investigated the antileukemic activity of MEDS433, the DHODH inhibitor developed by our group, against AML. Learning from previous failures, we mimicked human conditions (performing experiments in the presence of physiological uridine plasma levels) and looked for synergic combinations to boost apoptosis, including classical antileukemic drugs and dipyridamole, a blocker of the pyrimidine salvage pathway. MEDS433 induced apoptosis in multiple AML cell lines, not only as a consequence of differentiation, but also directly. Its combination with antileukemic agents further increased the apoptotic rate, but when experiments were performed in the presence of physiological uridine concentrations, results were less impressive. Conversely, the combination of MEDS433 with dipyridamole induced metabolic lethality and differentiation in all AML cell lines; this extraordinary synergism was confirmed on AML primary cells with different genetic backgrounds and was unaffected by physiological uridine concentrations, predicting in human activity.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5182-5182
Author(s):  
Xutao Guo ◽  
Bowen Yan ◽  
Yi Qiu

Acute myeloid leukemia (AML) exhibits large intrinsic variation in drug responsiveness due to its inherent heterogeneity. Therefore, it is important to understand the resistant mechanism in order to improve the treatment. In our previously study, the OCI-AML2-resistant cell lines were established to resist cytarabine (Ara-C) in the concentration of 50 µM (OCI-AML2 R50). The RNA-seq results showed that many genes changed in the resistant cells compared to wild type OCI-AML2 cells. One of the most remarkably decreased gene in resistant cells was HOXA11 (Homeobox A11). It is the part of the A cluster on chromosome 7 and encodes a DNA-binding transcription factor which regulates gene expression, morphogenesis, and differentiation. In this study, we have evaluated the importance of HOXA11 in AML chemoresistance. We found that knockdown of HOXA11 repressed the WT OCI-AML2 cell proliferation and increased the population of cells expressing CD123 and CD47 LSC (Leukemia stem cell) markers and enhanced the resistance to Ara-C in vitro, while overexpression of HOXA11 showed the reverse effect. These results support the idea that HOXA11 promotes drug sensitivity and apoptosis in AML. However, the result also showed that overexpression of HOXA11 repressed the OCI-AML2 R50 cell proliferation and enhanced the resistance. Therefore, HOXA11 plays opposite role in sensitive cells and resistant cells. We further investigated the mechanism for these effects. We found that knockdown of HOXA11 decreased the p53 gene expression and overexpression of HOXA11 increased the expression of p53 in OCI-AML2 and R50 cells. Further, in OCI-AML2 R50 cells p53 has a hotspot mutation in DNA binding site and studies have shown that p53 mutation enhance cancer cell survival and chemoresistance. Therefore, our study shows dual roles for HOXA11 in cell survival. In p53 wild type parental AML2 cells, HOXA11 induces wild type p53 expression to enhance drug sensitivity while in resistant cell, HOXA11 promotes mutant p53 expression and enhances the resistance of chemotherapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2951-2951
Author(s):  
Raoul Tibes ◽  
Ashish Choudhary ◽  
Amanda Henrichs ◽  
Sadia Guled ◽  
Irma Monzon ◽  
...  

Abstract In order to improve treatment strategies for Acute Myeloid Leukemia (AML), we adapted a functional genomics approach using RNAi screening to identify molecular targets that are vital to the growth of AML. Herein we report the first large-scale kinome gene silencing screen in AML. A high throughput RNAi screen was developed for the efficient siRNA transfection of AML cell lines. Eight commercially available cationic lipid-based transfection reagents were tested for their ability to transfect several AML cell lines with siRNA. These extensive transfection optimization experiments identified two AML cells lines TF-1 and ML4 with up to 95–100 and 70–75% transfection efficiency respectively. Two independent replicate kinome screens were performed on both cell lines using a siRNA library targeting 572 kinase genes with 2 siRNA/gene. At 96 hours post transfection, cell proliferation was assessed and the B-score method was used to background correct and analyze the screening data. Several siRNA to specific kinases were identified that significantly inhibit cell proliferation of up to ~40–88%. Hits were defined at two thresholds: siRNA having a B-score of <−2 providing a statistically significance of p<0.05 (confidence of > 95%) and a cutoff B-score of <−1.5 providing greater than 87% confidence for each siRNA hit. Two different kinases (2 siRNA/gene/screen) were identified as major growth regulating kinases in TF1 cells with all 4 siRNA/gene having a B-score <−2. For two additional kinases, 3/4 siRNA for each gene had a Bscore <−2. Expanding the cutoff to a B-score <−1.5 three further kinases were targeted by at least 3/4 siRNA/gene. Similar analysis using the same criteria for ML4 cells identified one kinase targeted by 3/4 siRNA at a B-score <−2, seven kinases with 2/4 siRNA <−2 and two kinases with 3/4 siRNA/gene at a B-score of <−1.5. Common hits for both cell lines with at least 6/8 siRNA per gene from 4 screens performing at a B-score <−2 identified two kinases, one of them PLK1. Applying a B-score threshold of <−1.5, we identified five kinases for which at least 5/8 siRNA/gene from 4 screens met these criteria. Kinases/genes will be presented at the meeting.Confirmation of gene silencing and validation of growth response is currently underway for a subset of genes. Among the strongest hits are siRNA targeting PLK1, as well as siRNA targeting three other kinase-genes involved in regulating cell cycle progression and checkpoints and gene ontology (GO) analysis showed enrichment in cell cycle and cell cycle-checkpoint processes. Inhibitors against PLK1 and other kinase hits identified in the screen are in (pre)-clinical development and if confirmed, our experiments provide a strong rational to test these in AML. The application of RNAi based screening is useful in the identification of genes important in AML proliferation, which could serve as targets for therapeutic intervention and guide AML drug development. Furthermore, results from these types of functional genomics approaches hold promise to be rapidly translated into clinical application.


Blood ◽  
2012 ◽  
Vol 120 (4) ◽  
pp. 858-867 ◽  
Author(s):  
Jing Fang ◽  
Garrett Rhyasen ◽  
Lyndsey Bolanos ◽  
Christopher Rasch ◽  
Melinda Varney ◽  
...  

Bortezomib (Velcade) is used widely for the treatment of various human cancers; however, its mechanisms of action are not fully understood, particularly in myeloid malignancies. Bortezomib is a selective and reversible inhibitor of the proteasome. Paradoxically, we find that bortezomib induces proteasome-independent degradation of the TRAF6 protein, but not mRNA, in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) cell lines and primary cells. The reduction in TRAF6 protein coincides with bortezomib-induced autophagy, and subsequently with apoptosis in MDS/AML cells. RNAi-mediated knockdown of TRAF6 sensitized bortezomib-sensitive and -resistant cell lines, underscoring the importance of TRAF6 in bortezomib-induced cytotoxicity. Bortezomib-resistant cells expressing an shRNA targeting TRAF6 were resensitized to the cytotoxic effects of bortezomib due to down-regulation of the proteasomal subunit α-1 (PSMA1). To determine the molecular consequences of loss of TRAF6 in MDS/AML cells, in the present study, we applied gene-expression profiling and identified an apoptosis gene signature. Knockdown of TRAF6 in MDS/AML cell lines or patient samples resulted in rapid apoptosis and impaired malignant hematopoietic stem/progenitor function. In summary, we describe herein novel mechanisms by which TRAF6 is regulated through bortezomib/autophagy–mediated degradation and by which it alters MDS/AML sensitivity to bortezomib by controlling PSMA1 expression.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2228-2228
Author(s):  
Xin Long ◽  
Laszlo Perlaky ◽  
Tsz-Kwong Chris Man ◽  
Michele S. Redell

Abstract Acute myeloid leukemia (AML) is a life-threatening bone marrow malignancy with a relapse rate near 50% in children, despite aggressive chemotherapy. Accumulating evidence shows that the bone marrow stromal environment protects a subset of leukemia cells and allows them to survive chemotherapy, eventually leading to recurrence. The factors that contribute to stroma-induced chemotherapy resistance are largely undetermined in AML. Our goal is to delineate the mechanisms underlying stroma-mediated chemotherapy resistance in human AML cells. We used two human bone marrow stromal cell lines, HS-5 and HS-27A, to study stroma-induced chemotherapy resistance. Both stromal cell lines are equally effective in protecting AML cell lines and primary samples from apoptosis induced by chemotherapy agents, including mitoxantrone, etoposide, and cytarabine. By gene expression profiling using the Affymetrix U133Plus 2 platform, we previously found that CYR61 was among the genes that were commonly upregulated in AML cells by both stromal cell lines. CYR61 is a secreted matricellular protein that is expressed at relatively low levels by AML cells, and at higher levels by stromal cells. CYR61 binds and activates integrins and enhances growth factor signaling in AML cells, and it has been associated with chemoresistance in other malignancies. Our current data provide functional evidence for a role for this protein in stroma-mediated chemoresistance in AML. First, we added anti-CYR61 neutralizing immunoglobulin (Ig), or control IgG, to AML-stromal co-cultures, treated with chemotherapy for 24 hours, and measured apoptosis with Annexin V staining and flow cytometry. In THP-1+HS-27A co-cultures treated with 50 nM mitoxantrone, the apoptosis rate was 33.0 ± 3.7% with anti-CYR61 Ig v. 16.3 ± 4.2% with control IgG; p=0.0015). Next, we knocked down CYR61 in the HS-5 and HS-27A stromal cell lines by lentiviral transduction of two individual shRNA constructs, and confirmed knockdown (KD) at the gene and protein levels for both cell lines. These CYR61-KD stromal cells provided significantly less protection for co-cultured AML cells treated with mitoxantrone, compared to stromal cells transduced with the non-silencing control. For example, the apoptosis rate for THP-1 cells co-cultured with CYR61-KD HS-27A cells was 10.8 ± 0.8%, compared to 6.8 ± 1.1% for THP-1 cells co-cultured with control HS-27A cells (p=0.02). Similar results were obtained with NB-4 AML cells. These results demonstrate that CYR61 contributes to stroma-mediated chemoresistance. CYR61 binds to integrin αvβ3 (Kireeva, et al, J. Biol. Chem., 1998, 273:3090), and this integrin activates spleen tyrosine kinase (Syk) (Miller, et al, Cancer Cell, 2013, 24:45). Using intracellular flow cytometry, we found that activated Syk (pSyk) increased in THP-1 and NB-4 cell lines, and in primary AML patient samples, upon exposure to control HS-27A cells. In primary samples, the mean fluorescence intensity (MFI) for pSyk averaged 11.7 ± 1.3 in co-culture v. 6.6 ± 0.6 for cells cultured alone (p=0.004, n=10). In contrast, pSyk did not significantly increase in AML cells co-cultured with CYR61-KD HS-27A cells (MFI for primary patient samples: 8.6 ± 0.8). This result implicates Syk as a downstream signaling mediator of CYR61. To determine the role of CYR61-induced Syk signaling in chemotherapy resistance, we treated AML-stromal cell co-cultures with 3 uM R406, a potent Syk inhibitor, or DMSO, then added 300 nM mitoxantrone, and measured apoptosis after 24 hours. In AML cells co-cultured with control HS-27A cells, mitoxantrone-induced apoptosis was significantly increased by Syk inhibition (THP-1 cells: 13.7 ± 0.7% with R406 v. 10.0 ± 0.3% with DMSO, p<0.05), consistent with reduced chemoresistance. Notably, R406 did not further increase mitoxantrone-induced apoptosis in AML cells co-cultured with CYR61-KD HS-27A stromal cells (THP-1 cells: 15.7 ± 0.2% with R406 v. 16.9 ± 0.4% with DMSO). Similar results were seen with NB-4 cells, as well. These results support the notion that CYR61 signals through the integrin-Syk pathway to protect AML cells from chemotherapy. Therefore, the CYR61 - integrin - Syk pathway may be a potential therapeutic target for overcoming stroma-induced chemotherapy resistance in AML. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document