scholarly journals Novel Irreversible Menin Inhibitor, BMF-219, Shows Potent Single Agent Activity in Clinically Relevant DLBCL Cells

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4318-4318
Author(s):  
Priyanka Somanath ◽  
Daniel Lu ◽  
Brian Law ◽  
Tenley C. Archer ◽  
Alexandru Cacovean ◽  
...  

Abstract Introduction: Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of Non-Hodgkin Lymphoma. Double Hit Lymphomas (DHL) and Double Expresser Lymphomas (DEL) are high grade B-cell lymphomas (HGBLs) that have high MYC and BCL2 dependency. DHL harbor translocations of MYC and BCL2 (or BCL6 less frequently), which lead to increased cell growth and survival. Standard R-CHOP therapy is inadequate and results in poor patient outcomes. Novel targeted agents, which inhibit MYC and BCL2 are currently under investigation; however, there is no established standard-of-care for these highly aggressive lymphomas. Menin is a scaffold protein that is essential for cancers driven by oncogenic MLL-fusions, such as in acute leukemias. At present, the role of menin in DLBCL is not well described. A recent study demonstrated that MYC and menin co-localize on chromatin, leading to menin-mediated enhancement of MYC target gene expression in cancer cells. However, the relationship between menin and MYC and the direct effect of menin inhibition on MYC in liquid tumors has not been examined. Here, we demonstrate the ability of BMF-219, a novel, selective, orally bioavailable, irreversible inhibitor of menin, to modulate MYC expression in leukemia cells as the basis for exploring activity of the molecule in DLBCL cells. Methods: MOLM-13 cells incubated with BMF-219 for 6 and 24 hours at 0.5 μM and 1 μM were analyzed by RNA-seq on the Illumina NextSeq 550 platform. Differentially expressed genes were identified through DeSeq2 negative binomial model analysis. Transcription factor activity inference of differentially expressed genes was calculated using a published statistical framework and algorithm. DLBCL DHL cell lines, DB and Toledo, were treated with BMF-219 as monotherapy, and in combination with ABT-199, at a concentration range of 0.01 μM to 3 μM for a 4-day duration. Cell viability was measured using CellTiter Glo and IC 50 values were calculated. Results: RNA-seq analysis of MOLM-13 cells treated with BMF-219 showed a marked reduction of MYC transcript levels by ~25-fold at 6 hours of treatment at both concentrations tested, and ~110-fold and ~220-fold at 0.5 μM and 1 μM BMF-219 treatment, respectively, at 24 hours. Additionally, MYC and its co-factor, MAX, emerged as top candidates in transcription factor activity inference analysis based on ChIP-seq GEO repository datasets and differential expression of MOLM-13 cells treated with BMF-219 at 24 hours. These results prompted an examination of the effects of BMF-219 on lymphoid malignancies known to be highly dependent on MYC aberrations. Single-agent BMF-219 reduced >90% of cell viability in both DB and Toledo cells, at 1.0 μM and 0.36 μM, respectively. The IC 50 values of BMF-219 were calculated as 0.32 μM and 0.29 μM for DB and Toledo, respectively. Two reversible menin inhibitors tested were less effective in reducing growth of DHL cell lines. One reversible menin inhibitor exhibited IC 50 values of 3 μM and 1.5 μM in DB and Toledo, respectively, while neither cell line was sensitive to the other reversible compound. Given that a 20-fold reduction in BCL2 transcript occurred in MOLM-13 cells at 24 hours of 0.5 μM BMF-219 exposure, we investigated whether a combination of BCL2 inhibitor, ABT-199, with BMF-219 could further reduce cell viability of DHL cells. ABT-199 as a single agent reduced viability of Toledo cells, but not DB cells, at 0.33 μM. By contrast, the combination of BMF-219 and ABT-199 synergistically killed both DB and Toledo cells. Combination treatment using both inhibitors at 0.33 μM killed >80% of DB cells and 100% of Toledo cells. These results demonstrate that DLBCL DHL cells are sensitive to menin inhibition. Of note, initial data has indicated that multiple myeloma cells also manifest responses to BMF-219 similar to DHL DLBCL. Conclusions: Collectively, our study demonstrates the unique ability of BMF-219's irreversible menin inhibition, which leads to marked killing of DHL cells. BMF-219, as a single agent, substantially reduces MYC and BCL2 gene expression in liquid tumor cells, providing a molecular basis for targeting DLBCL and other tumors with these aberrations. Additionally, BMF-219 offers single agent as well as treatment synergy when combined with ABT-199 in DHL cells. These data serve as initial pre-clinical evidence for employing irreversible menin inhibition as a promising therapeutic strategy in DHL DLBCL. Disclosures Somanath: Biomea Fusion, Inc.: Consultancy, Current Employment, Current equity holder in publicly-traded company. Lu: Biomea Fusion Inc.: Current Employment, Current equity holder in publicly-traded company. Law: Biomea Fusion Inc.: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Archer: Biomea Fusion: Current Employment, Current holder of stock options in a privately-held company. Cacovean: Abbvie: Current equity holder in publicly-traded company; Lyell Imunopharma: Current equity holder in publicly-traded company; CytomX: Current equity holder in publicly-traded company; Iovance: Current equity holder in publicly-traded company, Ended employment in the past 24 months; Biomea Fusion: Current Employment, Current equity holder in publicly-traded company. Palmer: Nyrada, Inc.: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Exopharm: Consultancy; Biomea Fusion Inc.: Consultancy, Current equity holder in publicly-traded company. Kinoshita: Biomea Fusion INC.,: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company, Patents & Royalties. Butler: Biomea Fusion Inc.: Current Employment, Current equity holder in publicly-traded company.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2232-2232
Author(s):  
Jin Wang ◽  
Venu Valmeekam ◽  
Kaijin Wu ◽  
Jiang F. Zhong ◽  
Andres Stucky ◽  
...  

Abstract Background: Murine double minute 2 (MDM2) is the primary negative regulator of the tumor suppressor protein, p53. Navtemadlin (KRT-232), a potent, selective, orally available MDM2 inhibitor, restores p53 activity to drive apoptosis of cancer cells in TP53 WT malignancies through the intrinsic (mitochondrial) pathway. Navtemadlin is being evaluated in a phase 3 trial in relapsed or refractory myelofibrosis, and in phase 1b/2 trials in various hematologic malignancies and solid tumors. Because p53 is infrequently mutated and MDM2 is often overexpressed in acute myeloid leukemia (AML), navtemadlin represents a rational therapeutic candidate (Prokocimer et al. Blood. 2017). Synergy between navtemadlin and the hypomethylating agent decitabine has been demonstrated in preclinical AML models (Canon et al. AACR. 2016), although the mechanisms mediating this effect remain unclear. The present study aims to identify mechanisms of navtemadlin-mediated apoptosis within AML cell lines, timing of pathway activation, and effects of combination treatment with navtemadlin + decitabine. Methods: MOLM13 or MV-4-11 AML cell lines were treated with a vehicle control, 0.5-1.0 µM navtemadlin, 1.0 µM decitabine, or with a combination of navtemadlin + decitabine. Cells were pretreated with decitabine for 42-48 hours followed by navtemadlin + fresh decitabine for 0, 5, 10, or 24 hours, and were then harvested for RNA-Seq and immunoblotting analyses of apoptotic pathway proteins. Gene set enrichment analysis (GSEA) and signaling pathway impact analysis (SPIA) were performed on RNA-Seq data to determine the most differentially regulated pathways (using KEGG curated signaling pathways) between controls, single-agent navtemadlin or decitabine, or combination treatment. Protein expression analyses were conducted to validate RNA-Seq data. Results: GSEA of MOLM13 and MV-4-11 cells at 10 hours showed that, relative to controls, several signaling pathways were differentially regulated to a statistically significant level (P≤0.05) with navtemadlin alone, including activation of apoptosis and p53 signaling pathways, and suppression of DNA replication and cell cycle pathways, in line with previous reports (Figure 1a). In MOLM13 cells treated with single-agent decitabine, lipid and proteoglycan-related signaling pathways were activated, whereas several metabolic pathways were suppressed. With combination treatment in both cell lines, unique activated pathways relative to controls included immune-related and cancer-signaling pathways; and unique suppressed pathways included base excision repair and cellular senescence pathways. SPIA analysis of both cell lines at 10 hours produced higher perturbation scores for apoptosis and p53 signaling pathways with combination treatment vs navtemadlin alone (Figure 1b), indicating more robust pathway activation. Additionally, tumor microenvironment (TME) and extracellular matrix (ECM) remodeling pathways showed more robust perturbation when combined with navtemadlin vs decitabine alone. At 24 hours, only SPIA was conducted, which showed less apoptotic pathway induction relative to 10 hours in both cell lines, suggesting time-dependent signaling effects. Cellular senescence pathways were suppressed, whereas central carbon metabolism and HIF-1 signaling were induced by single-agent navtemadlin relative to controls in both cell lines, with combination treatment further enhancing these effects. Combination treatment also induced significant changes in the expression of ECM remodeling genes including MMP9, MMP2, and ITGB7. Protein expression analyses in both cell lines confirmed early upregulation of key apoptosis regulators including p53, p21, PUMA, cleaved caspase-3 and cleaved-PARP with navtemadlin alone, and the DNA-damage response protein, yH2AX, with decitabine alone. The combination enhanced the induction of p53, p21, and yH2AX vs controls or single agents alone. Conclusion: Initial results from this study suggest that navtemadlin + decitabine enhanced apoptosis and p53 signaling pathways to a greater degree than either single agent in AML cell lines. Combination treatment also induced several unique DNA damage response, ECM-remodeling, and TME-related pathways. Activation of both proapoptotic and tumor-stromal interaction pathways suggest a unique mechanism of navtemadlin + decitabine for inhibiting AML cell growth. Figure 1 Figure 1. Disclosures Valmeekam: Telios Pharmaceuticals: Current Employment, Current holder of stock options in a privately-held company, Other: travel, accommodations, expenses. Canon: Oncovalent Therapeutics: Current Employment; Amgen, Inc.: Current holder of individual stocks in a privately-held company, Ended employment in the past 24 months. Krejsa: Acerta Pharma: Current holder of individual stocks in a privately-held company; AstraZeneca: Current equity holder in publicly-traded company; Seattle Genetics: Current equity holder in publicly-traded company, Divested equity in a private or publicly-traded company in the past 24 months; Kartos Therapeutics: Current Employment, Current holder of stock options in a privately-held company, Other: travel, accommodations, expenses. Kelly: AstraZeneca: Consultancy; Bayer: Speakers Bureau; Verastem: Consultancy; Amgen: Consultancy; Berkley Lights: Current equity holder in publicly-traded company; Denovo Biopharma: Consultancy; Takeda: Consultancy; Sanofi-Aventis: Consultancy; Novartis: Speakers Bureau; Janssen: Speakers Bureau; Agios: Current equity holder in publicly-traded company; Celgene: Speakers Bureau; Epizyme: Speakers Bureau; Pharmacyclics: Speakers Bureau; Karyopharm: Speakers Bureau; Gilead: Speakers Bureau. OffLabel Disclosure: Yes, navtemadlin (KRT-232) is an investigational small molecule inhibitor.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3356-3356
Author(s):  
Melinda A. L. Day ◽  
Philipp Sergeev ◽  
Caroline A. Heckman ◽  
Anna Schinzel ◽  
Nikolaus D. Obholzer ◽  
...  

Abstract Spleen tyrosine kinase (SYK) is a non-receptor tyrosine kinase that mediates integrin and Fc receptor signaling in myeloid cells. SYK has been implicated as an oncogenic driver in acute myeloid leukemia (AML) with aberrant expression of HOXA9 and MEIS1 and cooperates with FLT3 internal tandem duplication to drive leukemogenesis. The oral SYK inhibitor entospletinib (ENTO) has demonstrated clinical activity in HOXA9/MEIS1 driven AML and is currently being investigated in a phase 3 trial of previously untreated patients with nucleophosmin1-mutated (NPM1 mut) AML. Lanraplenib (LANRA) is a next generation oral SYK inhibitor with potency and selectivity comparable to ENTO. In healthy volunteers and patients with autoimmune disease, LANRA has shown pharmacokinetic properties that compare favorably with ENTO. To support the clinical development of LANRA for the treatment of AML, ex vivo treatment of patient-derived AML cells was used to compare its activity to that of ENTO, both as a single-agent and in combination with other AML therapies. First, ENTO and LANRA single-agent activities were evaluated in peripheral blood-derived blasts from 15 AML patients, representing different mutational backgrounds including NPM1, FLT3, PTPN11, and NRAS mutations. AML cells were seeded into 96 well plates and treated with ENTO and LANRA for 6 days. Comparable effects on viability were observed across the 15 models with the 2 compounds, and in 11 of the models, the half maximal inhibitory concentration (IC 50) values were within 2-fold of each other. ENTO had a slightly lower IC 50 value than LANRA in the FLT3-mutated models possibly due to the direct FLT3 inhibitory activity of ENTO. Next, we tested the activity of ENTO and LANRA ex vivo in bone marrow-derived AML blasts from 29 AML patients representing diverse mutational backgrounds, including NPM1, IDH1, FLT3, and RAS mutations as well as MLL rearrangements. The models were treated for 9 days with either ENTO or LANRA, and viability was assessed using Annexin V and 7-aminoactinomycin D staining. Again, ENTO and LANRA showed comparable effects on cell viability with no significant differences between the compounds when compared across the different mutational backgrounds. Both studies suggest the potential for anti-leukemic activity in several different genetically defined subsets of AML. Matrix combination assays were performed by combining ENTO or LANRA with either cytarabine (NPM1 mut), gilteritinib (FLT3 mut), or trametinib (RAS mut) with cell viability and death assessed after a 3-day incubation period. Increased cell death in an additive manner was observed in all combinations tested, with results for ENTO and LANRA being similar, indicating the utility of both compounds in combinatorial treatment paradigms. These results support the clinical evaluation of LANRA in genetically defined subsets of AML. A phase 1b/2 study of LANRA in combination with the selective FLT3 inhibitor gilteritinib, in patients with relapsed or refractory FLT3 mut AML is planned for the end of this year. Disclosures Day: Cyteir Therapeutics: Current equity holder in publicly-traded company, Ended employment in the past 24 months; Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company. Heckman: Novartis: Research Funding; Orion Pharma: Research Funding; Celgene/BMS: Research Funding; Oncopeptides: Consultancy, Research Funding; Kronos Bio, Inc.: Research Funding. Schinzel: Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company. Obholzer: Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Lin: Kronos Bio, Inc.: Current Employment. Kumar: Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company. DiMartino: Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company. Saffran: Kronos Bio, Inc.: Current Employment, Current equity holder in publicly-traded company.


Development ◽  
1997 ◽  
Vol 124 (10) ◽  
pp. 1963-1973 ◽  
Author(s):  
K.J. Kaneko ◽  
E.B. Cullinan ◽  
K.E. Latham ◽  
M.L. DePamphilis

mTEF-1 is the prototype of a family of mouse transcription factors that share the same TEA DNA binding domain (mTEAD genes) and are widely expressed in adult tissues. At least one member of this family is expressed at the beginning of mouse development, because mTEAD transcription factor activity was not detected in oocytes, but first appeared at the 2-cell stage in development, concomitant with the onset of zygotic gene expression. Since embryos survive until day 11 in the absence of mTEAD-1 (TEF-1), another family member likely accounts for this activity. Screening an EC cell cDNA library yielded mTEAD-1, 2 and 3 genes. RT-PCR detected RNA from all three of these genes in oocytes, but upon fertilization, mTEAD-1 and 3 mRNAs disappeared. mTEAD-2 mRNA, initially present at approx. 5,000 copies per egg, decreased to approx. 2,000 copies in 2-cell embryos before accumulating to approx. 100,000 copies in blastocysts, consistent with degradation of maternal mTEAD mRNAs followed by selective transcription of mTEAD-2 from the zygotic genome. In situ hybridization did not detect mTEAD RNA in oocytes, and only mTEAD-2 was detected in day-7 embryos. Northern analysis detected all three RNAs at varying levels in day-9 embryos and in various adult tissues. A fourth mTEAD gene, recently cloned from a myotube cDNA library, was not detected by RT-PCR in either oocytes or preimplantation embryos. Together, these results reveal that mTEAD-2 is selectively expressed for the first 7 days of embryonic development, and is therefore most likely responsible for the mTEAD transcription factor activity that appears upon zygotic gene activation.


2017 ◽  
Vol 28 (2) ◽  
pp. 182-191 ◽  
Author(s):  
Jake Yeung ◽  
Jérôme Mermet ◽  
Céline Jouffe ◽  
Julien Marquis ◽  
Aline Charpagne ◽  
...  

2008 ◽  
Vol 7 (1) ◽  
pp. 27 ◽  
Author(s):  
Audrey Sermeus ◽  
Jean-Philippe Cosse ◽  
Marianne Crespin ◽  
Veronique Mainfroid ◽  
Francoise de Longueville ◽  
...  

2014 ◽  
Vol 25 (1) ◽  
pp. 184-195 ◽  
Author(s):  
Rachael A. Bowe ◽  
Orla T. Cox ◽  
Verónica Ayllón ◽  
Emilie Tresse ◽  
Nollaig C. Healy ◽  
...  

Epithelial cell differentiation and polarized migration associated with epithelial-to-mesenchymal transition (EMT) in cancer requires integration of gene expression with cytoskeletal dynamics. Here we show that the PDZ-LIM domain protein PDLIM2 (Mystique/SLIM), a known cytoskeletal protein and promoter of nuclear nuclear factor κB (NFκB) and signal transducer and activator of transcription (STAT) degradation, regulates transcription factor activity and gene expression through the COP9 signalosome (CSN). Although repressed in certain cancers, PDLIM2 is highly expressed in invasive cancer cells. Here we show that PDLIM2 suppression causes loss of directional migration, inability to polarize the cytoskeleton, and reversal of the EMT phenotype. This is accompanied by altered activity of several transcription factor families, including β-catenin, Ap-1, NFκB, interferon regulatory factors, STATs, JUN, and p53. We also show that PDLIM2 associates with CSN5, and cells with suppressed PDLIM2 exhibit reduced nuclear accumulation and deneddylation activity of the CSN toward the cullin 1 and cullin 3 subunits of cullin-RING ubiquitin ligases. Thus PDLIM2 integrates cytoskeleton signaling with gene expression in epithelial differentiation by controlling the stability of key transcription factors and CSN activity.


Author(s):  
Stephen P. Methot ◽  
Jan Padeken ◽  
Giovanna Brancati ◽  
Peter Zeller ◽  
Colin E. Delaney ◽  
...  

AbstractThe developmental role of histone H3K9 methylation (H3K9me), which typifies heterochromatin, remains unclear. In Caenorhabditis elegans, loss of H3K9me leads to a highly divergent upregulation of genes with tissue and developmental-stage specificity. During development H3K9me is lost from differentiated cell type-specific genes and gained at genes expressed in earlier developmental stages or other tissues. The continuous deposition of H3K9me2 by the SETDB1 homolog MET-2 after terminal differentiation is necessary to maintain repression. In differentiated tissues, H3K9me ensures silencing by restricting the activity of a defined set of transcription factors at promoters and enhancers. Increased chromatin accessibility following the loss of H3K9me is neither sufficient nor necessary to drive transcription. Increased ATAC-seq signal and gene expression correlate at a subset of loci positioned away from the nuclear envelope, while derepressed genes at the nuclear periphery remain poorly accessible despite being transcribed. In conclusion, H3K9me deposition can confer tissue-specific gene expression and maintain the integrity of terminally differentiated muscle by restricting transcription factor activity.


1996 ◽  
Vol 74 (4) ◽  
pp. 523-534 ◽  
Author(s):  
Keith Wheaton ◽  
Peter Atadja ◽  
Karl Riabowol

Several lines of evidence suggest that the limited replication potential of normal human cells is due to the presence of an intrinsic genetic programme. This "senescence programme" is believed to reduce the incidence of cancer by limiting the growth of most of the transformed cells arising in vivo, although some cells do escape senescence becoming both immortalized and transformed. Here we review the literature that describes the senescence process in terms of gene expression and the regulation of gene expression by a variety of mechanisms affecting transcription factor activity. We focus on regulation of the c-fos gene through posttranslational modification of the serum response factor (SRF) as an example of altered gene expression during cellular aging.Key words: cellular aging, transcription, Fos, SRF, phosphorylation.


Sign in / Sign up

Export Citation Format

Share Document