Antileukemic Effects of the Novel Agent Elesclomol.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2736-2736 ◽  
Author(s):  
Sue Chow ◽  
Masazumi Nagai ◽  
Suqin He ◽  
Ronald K Blackman ◽  
James Barsoum ◽  
...  

Abstract Abstract 2736 Poster Board II-712 Elesclomol (N-malonyl-bis (N′-methyl-N′-thiobenzoyl hydrazide)) is an investigational first-in-class oxidative stress inducer that triggers apoptosis in cancer cells (Kirshner et al., Mol Cancer Ther 2008;7:2319–27). In the clinic, elesclomol is well tolerated in humans and showed activity in combination with paclitaxel in patients with refractory solid tumors (Berkenblit et al., Clin Cancer Res 2007;13:584–90). The aims of the current study are to examine the activity of elesclomol against a range of AML cell lines, including primary patient blast cultures, to investigate the mechanisms of drug action and the potential to combine elesclomol with other agents, and to identify candidate biomarkers for monitoring effects during treatment of leukemia patients with elesclomol. Here we describe the effects of elesclomol treatment in 4 AML cell lines selected based on their varying molecular attributes. Effects on cellular redox state and mitochondrial function were monitored using a flow cytometry incorporating the glutathione (GSH) probe monobromobimane, the reactive oxygen species (ROS) probe carboxy-dichlorofluorescin and the mitochondrial membrane potential stain DiIC(1)5. In addition, outer cell membrane integrity was determined by propidium iodide exclusion. Dual staining of fixed, permeabilized cells with phospho-specific antibodies to p38 and SAPK/JNK was used to determine if elesclomol treatment results in activation of the stress-activated MAP kinase pathways. Elesclomol showed potent anti-leukemic effects in vitro at concentrations as low as 10nM, which is well below the concentrations achieved in cancer patients, and greater toxicity was achieved with prolonged drug exposure. In OCI-AML2, a factor-independent, poorly differentiated AML cell line, toxicity was associated with loss of reduced GSH that coincided with a large increase in ROS generation and depolarization of the mitochondrial inner membrane, and later with loss of surface membrane integrity. A similar pattern was seen in OCI-M2, a p53-deficient erythroblastic leukemia cell line, except that during the early stages of drug effect these cells showed a large increase in reduced GSH, suggesting that initially they are able to compensate for drug-induced oxidative stress through enhanced cellular antioxidant production. In contrast, the factor-dependent line OCI-AML5, which appeared most sensitive to elesclomol, showed loss of outer membrane integrity without obvious prior oxidative stress while the Flt-3 ITD mutant line MV4-11 showed an initial loss of mitochondrial membrane potential without accompanying oxidative stress. Strikingly, we did not observe activation of the stress-responsive p38 or SAPK/JNK pathways in any of these 4 cell lines tested, suggesting that this is not a prominent response to elesclomol activity in AML and that additional mechanisms may be at work for the activity of elesclomol in these cells. Further investigations are ongoing and additional studies, including evaluation of elesclomol activity in primary leukemic cells from AML patients, will be presented. In summary, elesclomol is a potent novel compound that exerts anti-leukemic effects in tissue culture at drug concentrations that are well below those achieved in patients, suggesting that it might be active in leukemia patients. Disclosures: Chow: Synta Pharmaceuticals Inc.: Research Funding. Nagai:Synta Pharmaceuticals Inc.: Employment. He:Synta Pharmaceuticals Inc.: Employment. Blackman:Synta Pharmaceuticals Inc.: Employment, Equity Ownership. Barsoum:Synta Pharmaceuticals Inc.: Employment, Equity Ownership. Vukovic:Synta Pharmaceuticals Inc.: Employment, Equity Ownership. Hedley:Synta Pharmaceuticals Inc.: Research Funding.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1432-1432
Author(s):  
James M Bogenberger ◽  
William E. Pierceall ◽  
Ryan Lena ◽  
Reinhold Munker ◽  
Kaoru Tohyama ◽  
...  

Abstract Abstract 1432 Previously we demonstrated that inhibition of anti-apoptotic BCL-2 family members sensitize leukemic cells to 5-Azacytidine (5-Aza), using siRNA and pharmacological inhibition with the BH3-mimetic ABT-737, both in vitro and ex vivo (Bogenberger, JM., et. al. ASH Annual Abstracts 2011;118:Abstract 3513). Crucially, several anti-apoptotic BCL-2 members (e.g. BCL-2 and BCL-XL) required concurrent inhibition for potent and universal sensitization to 5-Aza. Anti-apoptotic BCL-2 proteins block the execution of programmed cell death (apoptosis) by binding to and counteracting two types of pro-apoptotic BCL-2 family proteins: the “BH3-only” proteins, including both activators (BIM and BID) and sensitizers (e.g. NOXA, PUMA, HRK), and the multi-domain effector proteins (BAX and BAK). Cells dependent on anti-apoptotic BCL-2 family members for survival have been defined as “primed for death” (Certo, M., et. al. Cancer Cell 2006 May;9(5):351-65). Importantly, the priming status reflects a dependence on anti-apoptotic BCL-2 family proteins and can be characterized with the BH3 profiling functional assay. This assay measures induction of mitochondrial outer membrane permeabilization (MOMP) in response to treatment with peptides derived from BH3-only proteins (Ni Chonghaile, T., et. al. Science 2011 Nov 25;334(6059);1129-33). Thus, the unique BH3 profile associated with a specific malignant cell population, is a function of the anti-apoptotic BCL-2 family member/s contextual expression in that cell population. Based on our observations of potent 5-Aza sensitization in combination with anti-apoptotic BCL-2 protein family inhibition, we hypothesized that BH3-profiling would predict response to 5-Aza. To address the potential utility of BH3 profiling in predicting response to 5-Aza in myeloid malignancies, we assayed a broad panel of AML-derived cell lines (N=13) by BH3 profiling and correlated BH3 metrics with 5-Aza response. Identical cell line passages were assayed by BH3 profiling and in 5-Aza drug dose response assays. The cell panel comprised lines derived from AML FAB subtypes M7, M6, M5, M4, M2 and M0, as well as diverse cytogenetics such as t(11;21), t(9;11), t(4;11), t(6;11), del 5q, del 7, and a broad spectrum of mutations such as FLT3, N-RAS, CDKN2A, NPM1 and DNMT3A. The panel included a normal karyotype AML cell line (CG-SH) (Munker, R., et. al. Leuk Res 2009 Oct:33(10):1405-8) and a blastic cell line derived from a patient with MDS (MDS-L) (Tohyama, K., et. al. Br J Haematol 1994 Jun;87(2):235-42). Partition modeling using several BH3 metrics discriminates 5-Aza response with statistical significance (N=13, Mann-Whitney p<0.01) between more sensitive (EC50<2uM) and less sensitive (EC50>2uM) AML lines. For instance, the Puma BH3 peptide alone distinguishes more sensitive from less sensitive cell lines (N=13, Mann-Whitney p=0.0046). Several more complex parameters, such as “Puma+Hrk”, “Puma+Noxa,” and “Bim+Puma+Noxa+Hrk,” also significantly discriminate 5-Aza response (Mann-Whitney p=0.0011). Using continuous variable analysis, a Puma+Hrk metric exhibited the strongest correlation with 5-Aza response (R2=0.85, p<0.0001), while an individual Puma metric yielded the second strongest correlation (R2=0.70, p=0.0004). In conclusion, BH3 profiling discriminates 5-Aza response and confirms a central role for anti-apoptotic BCL-2 members in 5-Aza response. Furthermore, HRK priming indicates that BCL-XL is an important, although not the sole, anti-apoptotic component determining 5-Aza response in myeloid cells. These BH3 profiling results are in agreement with our functional 5-Aza RNAi and BCL-XL/BCL-2 inhibitor data in combination with 5-Aza. Based on these results, we are currently evaluating BH3 profiles from 5-Aza responding and non-responding AML and MDS patients to validate BH3 profiling-derived metrics in predicting clinical outcomes to 5-Aza. Disclosures: Pierceall: Eutropics Pharmaceuticals: Employment, Equity Ownership. Lena:Eutropics Pharmaceuticals: Employment. Mesa:Sanofi: Research Funding; Incyte: Research Funding; Lilly: Research Funding; NS Pharma: Research Funding; YM Bioscience: Research Funding. Cardone:Eutropics Pharmaceuticals: Employment, Equity Ownership.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2648-2648 ◽  
Author(s):  
Sarah Gooding ◽  
Chad C. Bjorklund ◽  
Michael Amatangelo ◽  
Yan Ren ◽  
Mathieu Marella ◽  
...  

Abstract Background: Multiple myeloma (MM) is a clonally heterogeneous cancer that recurrently relapses during its clinical course, demonstrating how interplay among subclones can lead to differentially drug-tolerant populations. The determination of onset and mechanisms of drug resistance is of clinical interest, and the improved understanding of this will drive innovative therapeutic approaches. The IMiD® agents lenalidomide (Len) and pomalidomide (Pom) bind to the E3-ubiquitin ligase receptor, Cereblon (CRBN), leading to ubiquitination and subsequent proteosomal degradation of key transcription factors Ikaros (IKZF1) and Aiolos (IKZF3). As a consequence of this degradation, direct antitumor and immunostimulatory effects are observed. Previous studies have revealed potential mechanisms of resistance to Len and Pom in both MM cell lines and clinical cohorts, including mutations, transcriptional downregulation or alternative splicing of CRBN, or mutations elsewhere in the E3 ligase pathway (CUL4A, DDB1), IKZF1/3 or IRF4. In resistant cell lines and patients, activation of proliferation, cell cycle, IGF-1, MAPK and FOXM1 pathways are reported, in addition to non-activation of interferon signature genes (ISGs). We hypothesize that rare MM cells/subclones with genetic, epigenetic or proteomic architecture permitting survival against Len or Pom in vitro may exist prior to acquired resistance, but may not be detectable by bulk genomic, transcriptional or proteomic analyses. Our goal is to define such populations using a combination of single cell technologies, initially focusing on acquired Pom-resistance mechanisms and quantifying genetic and transcriptional changes in Cereblon and its mechanistic targets, and to identify features of pre-existing resistant cell populations. Methods: To investigate the mechanism and timing of onset of acquired resistance, we utilized H929 and MM1S MM cell lines made resistant to Pom by serial dose increments over time, to a final Pom concentration of 10 μM (H929/PR and MM1S/PR respectively). Samples were collected at various timepoints for analysis by genomic, bulk and single cell (10X genomics) RNASeq techniques, and single cell protein quantification of Cereblon, Ikaros and Aiolos by IHC. Results: Bulk genomic sequencing revealed the H929/PR cell line had gained two CRBN mutations, a frameshift deletion of the glutarimide-binding region present in 10.9% alleles, and a point mutation in the non-glutarimide binding region present in 71% alleles. Concurrent single-cell protein quantification showed a near complete loss of Cereblon expression in the majority of cells, with little change in Ikaros or Aiolos staining. Bulk RNASeq of the H929/PR cell line showed a significant enrichment of cell cycle, E2F and MYC target gene sets. Using single cell transcriptomics, we found high expression of the signature genes of the Pom-resistant population to co-localise with an independently derived FOXM1 target gene set to a cluster of sensitive single cells defined by a gene expression profile identifying cells at the G2M checkpoint. Sanger sequencing of CRBN in the MM1S/PR cell line showed a de novo 12 bp deletion in intron 5 which is required for spliceosome assembly, resulting in deletion of exon 6 of the mature mRNA transcript. Interestingly, CRBN exon 6 is required for interaction with DDB1, suggesting loss of a functional E3-ligase. Analysis by single cell IHC showed substrate levels similar to sensitive parental cell lines. As in the H929/PR cell line, bulk RNASeq again showed enrichment of E2F and MYC targets, and independently derived Pom-resistant and FOXM1 target gene sets co-localised to the same cluster of MM1S drug-naïve single cells, identified by a G2M checkpoint signature. Conclusions: A range of CRBN mutations, Cereblon protein loss and enrichment of cell cycle, FOXM1 target and G2M checkpoint-related signatures was found at the onset of Pom resistance. This association with altered cell cycle progression dynamics may lead to, correlate with or result from drug survival. Tracking mutation- and non mutation-associated transcription factor signatures through sequential time points from first drug exposure to full resistance may reveal mechanisms by which functional CRBN loss causes drug resistance. Disclosures Gooding: Celgene: Research Funding. Bjorklund:Celgene Corporation: Employment, Equity Ownership. Amatangelo:Celgene Corporation: Employment, Equity Ownership. Ren:Celgene Corporation: Employment, Equity Ownership. Marella:Celgene: Employment. Couto:Celgene: Employment. Towfic:Celgene Corporation: Employment, Equity Ownership. Oppermann:Bayer Pharma: Research Funding. Ramasamy:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Pierceall:Celgene Corporation: Employment, Equity Ownership. Thakurta:Celgene Corporation: Employment, Equity Ownership.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2768-2768
Author(s):  
Greg P Coffey ◽  
Jiajia Feng ◽  
Sabah Kadri ◽  
Y. Lynn Wang ◽  
Pin Lu ◽  
...  

Abstract Abnormal upregulation of NFκB activity is observed in a variety of B cell malignancies, resulting in proliferative and survival signals that contribute to tumor progression. Under normal resting conditions, NFκB is negatively regulated principally via its physical association with IκB (inhibitor of NFκB) family members, thereby inhibiting nuclear transport or access to DNA. In B cells, NFκB is typically activated via various external stimuli (e.g., ligation of the B cell antigen receptor (BCR), toll-like receptors, cytokine receptors, CD40), leading to IκB kinase complex-dependent phosphorylation of IκB members and targeting them for ubiquitination and degradation. In some cases, the need for external stimuli is diminished or completely circumvented by mutations to critical regulators of NFκB, as has been described in the context of activating mutations to CD79A/B, MYD88, and CARD11, as well as inactivation of negative regulators such as A20 and IκB family members (reviewed by Staudt, 2010). Each of these mutations has been observed clinically in patients with B cell malignancies (Wilson et al, 2012; Norenberg et al, 2015; Mansouri et al, 2015), and can impact the anti-tumor activity of selective BCR pathway inhibition (Davis et al, 2010; Wilson et al, 2012) in part via induction of autocrine cytokine stimulation leading to JAK/STAT-dependent up-regulation of MCL1 (Lam et al, 2008). We previously reported that cerdulatinib, a small molecule kinase inhibitor that dually targets SYK and the JAK family members JAK1, JAK3, and TYK2, maintained anti-tumor activity in DLBCL cell lines bearing mutations to CARD11, MYD88, and A20 (Ma et al, 2015). The majority of DLBCL cell lines exhibit various degrees of reliance on SYK and JAK signaling for survival, however in a screen of 15 DLBCL cell lines we found 3 that were completely resistant to cerdulatinib and are described here. In one of the cerdulatinib-resistant cell lines, RCK8, next generation sequencing revealed bi-allelic inactivation of the IκBα gene. One allele carries a frameshift mutation in exon 1 resulting in the generation of a premature stop codon, and the second allele is a nonsense mutation in exon 3 at Gln154, also leading to a stop codon. In accord with a previous report (Kalaitzidis et al, 2002), the cell line lacks IκBα protein expression. We therefore proceeded to explore the possibility that loss of IκBα was responsible for resistance to cerdulatinib. Consistent with the loss of IκBα, the RCK8 cell line exhibited enhanced basal NFκB activity. Genetic re-introduction of wild type IκBα led to rapid suppression of NFκB, and ultimately cell cycle arrest and cell death, indicating that the cell line was dependent upon loss of this gene for survival. Associated with suppression of NFκB was decreased phosphorylation of cellular pAKT S473 and pERK Y202, but not of pSTAT3 Y705. We then attempted to knock down IκBα in cerdulatinib-sensitive cell lines using siRNA to determine if resistance to SYK/JAK inhibition could be generated. None of the DLBCL cell lines tested (n=4) could tolerate IκBα gene knock down, suggesting an additional mutation in RCK8 enables survival under conditions of homozygous loss of IκBα. Ligation of CD40 leads to a transient down-regulation of IκBα at the protein level (Oeckinghaus and Ghosh, 2009). We therefore examined the effect of CD40L on multiple DLBCL cell lines and found that IκBα was maximally suppressed within 30-60 minutes post CD40 stimulation, returning to pre-treatment levels by 2-4 hours. In contrast, the impact on NFκB activation was much longer, and 5 of 7 cerdulatinib-sensitive cell lines tested were made resistant by incubation with CD40L. Associated with this resistance was not only induction of NFκB, but also pERK Y204, pAKT S473, and pSTAT3 Y705. Interestingly, whereas the CD40L-induced NFκB activation was not inhibited by cerdulatinib, the other signaling events were, despite the generation of resistance. Loss of IkB family members has been described in the context of Hodgkin's lymphoma, non-Hodgkin's lymphoma, and chronic lymphocytic leukemia (Cabannes et al, 1999; Norenberg et al, 2015; Mansouri et al, 2015). Here we demonstrate that loss of IκBα in multiple DLBCL cell lines generates resistance to cerdulatinib. We will be exploring the clinical relevance of these in vitro observations in cell lines as part of an ongoing phase II trial of cerdulatinib in patients with relapsed/refractory B cell malignancies. Disclosures Coffey: Portola Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties, Research Funding. Feng:Portola Pharmaceuticals: Employment, Equity Ownership, Research Funding. Wang:Portola Pharmaceuticals: Honoraria, Research Funding. Michelson:Portola Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties, Research Funding. Pandey:Portola Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties, Research Funding. Curnutte:3-V Biosciences: Equity Ownership; Portola Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties, Research Funding; Sea Lane Biotechnologies: Consultancy. Conley:Portola Pharmaceuticals: Employment, Equity Ownership, Patents & Royalties, Research Funding.


2021 ◽  
Vol 28 (1) ◽  
Author(s):  
Junqiang Yan ◽  
Hongxia Ma ◽  
Xiaoyi Lai ◽  
Jiannan Wu ◽  
Anran Liu ◽  
...  

Abstract Background Parkinson’s disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. The oxidative stress is an important component of the pathogenesis of PD. Artemisinin (ART) has antioxidant and neuroprotective effects. The purpose of this study is to explore the neuroprotective effect of ART on 1-methyl-4-phenyliodine iodide (MPP +)-treated SH-SY5Y cells and underlying mechanism. Methods We used MPP+-treated SH-SY5Y cells to study the neuroprotective effect of ART. Cell viability was measured by MTT assay after incubating the cells with MPP+ and/or ART for 24 h. DCFH-DA was used to detect the level of intracellular reactive oxygen species (ROS), and WST-8 was used to detect the level of superoxide dismutase (SOD). The level of intracellular reduced glutathione (GSH) was detected with 5,5΄-dithiobis-(2-nitrobenzoic acid), and the level of malondialdehyde (MDA) was assessed based on the reaction of MDA and thiobarbituric acid. A mitochondrial membrane potential detection kit (JC-1) was used to detect changes in the mitochondrial membrane potential (MMP), and an Annexin V-FITC cell apoptosis kit was used to detect cell apoptosis. The expression levels of caspase-3, cleaved caspase-3 and the autophagy-related proteins LC3, beclin-1, and p62 were detected by Western blotting. In addition, to verify the change in autophagy, we used immunofluorescence to detect the expression of LC3 and p62. Results No significant cytotoxicity was observed at ART concentrations up to 40 μM. ART could significantly increase the viability of SH-SY5Y cells treated with MPP+ and reduce oxidative stress damage and apoptosis. In addition, the Western blotting and immunofluorescence results showed that MPP+ treatment could increase the protein expression of beclin1 and LC3II/LC3I and decrease the protein expression of p62, indicating that MPP+ treatment could induce autophagy. Simultaneous treatment with ART and MPP+ could decrease the protein expression of beclin1 and LC3II/LC3I and increase the protein expression of p62, indicating that ART could decrease the level of autophagy induced by MPP+. Conclusion Our results indicate that ART has a protective effect on MPP+-treated SH-SY5Y cells by the antioxidant, antiapoptotic activities and inhibition of autophagy. Our findings may provide new hope for the prevention and treatment of PD.


2014 ◽  
Vol 306 (4) ◽  
pp. C334-C342 ◽  
Author(s):  
Eiji Takahashi ◽  
Michihiko Sato

To elucidate how tumor cells produce energy in oxygen-depleted microenvironments, we studied the possibility of mitochondrial electron transport without oxygen. We produced well-controlled oxygen gradients (ΔO2) in monolayer-cultured cells. We then visualized oxygen levels and mitochondrial membrane potential (ΔΦm) in individual cells by using the red shift of green fluorescent protein (GFP) fluorescence and a cationic fluorescent dye, respectively. In this two-dimensional tissue model, ΔΦm was abolished in cells >500 μm from the oxygen source [the anoxic front (AF)], indicating limitations in diffusional oxygen delivery. This result perfectly matched GFP-determined ΔO2. In cells pretreated with dimethyloxaloylglycine (DMOG), a prolyl hydroxylase domain-containing protein (PHD) inhibitor, the AF was expanded to 1,500–2,000 μm from the source. In these cells, tissue ΔO2 was substantially decreased, indicating that PHD pathway activation suppressed mitochondrial respiration. The expansion of the AF and the reduction of ΔO2 were much more prominent in a cancer cell line (Hep3B) than in the equivalent fibroblast-like cell line (COS-7). Hence, the results indicate that PHD pathway-activated cells can sustain ΔΦm, despite significantly decreased electron flux to complex IV. Complex II inhibition abolished the effect of DMOG in expanding the AF, although tissue ΔO2 remained shallow. Separate experiments demonstrated that complex II plays a substantial role in sustaining ΔΦm in DMOG-pretreated Hep3B cells with complex III inhibition. From these results, we conclude that PHD pathway activation can sustain ΔΦm in an otherwise anoxic microenvironment by decreasing tissue ΔO2 while activating oxygen-independent electron transport in mitochondria.


Sign in / Sign up

Export Citation Format

Share Document