Journal articles on the topic 'Equity lines'

To see the other types of publications on this topic, follow the link: Equity lines.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Equity lines.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

ANNAOUI, Ilias, and Pascal BARNETO. "The Determinants of Equity Lines Financing: An International Study." Bankers, Markets & Investors 171, no. 3 (December 22, 2022): 43–50. http://dx.doi.org/10.54695/bmi.171.8461.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Equity lines financing allows companies to increase their capital by issuing shares in successive tranches, as and when required over an agreed period of time, in order to boost equity and cash flow. The purpose of this article is to examine the factors which inform the use of this method, considering the specific parameters of businesses using equity lines, as well as variables linked to the institutional and economic context. Employing a model based on the Generalized Method of Moments (GMM) technique, we studied 407 firms in 15 countries making using of equity lines Financing in the period 2004-2018. The results confirm that the existence of opportunities for growth, the existence of a well-developed secondary market (maturity, expertise in financial intermediation activities, volume of transactions etc.) and the broader economic outlook are the principal explanatory variables behind the determinants of equity line financing.
2

Ebens, Allen J., Leanne Berry, Yung-Hsiang Chen, Gauri Deshmukh, Jake Drummond, Changchun Du, Michael Eby, et al. "A Selective PIM Kinase Inhibitor Is Highly Active In Multiple Myeloma: The Biology of Single Agent and PI3K/AKT/mTOR Combination Activity." Blood 116, no. 21 (November 19, 2010): 3001. http://dx.doi.org/10.1182/blood.v116.21.3001.3001.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 3001 PIM kinases co-regulate several important elements of the PI3K/AKT/mTOR pathway in myeloma cells (Munugalavadla V. et al., ASH 2010 submitted abstr.). In this study we show that pan-PIM inhibition suppresses growth in myeloma cell lines, xenografts, and primary patient samples, both as a single-agent as well acting synergistically in combination with PI3K, AKT, and mTOR inhibition. The PIM kinases are a family of 3 ser/thr growth factor- & cytokine-induced proteins hypothesized to have redundant survival and growth functions. Although PIM-1,2 have been noted as highly expressed in myeloma (Claudio et al., 2002), there are few data to support potential therapeutic utility of PIM inhibition in this indication. We show myeloma cell lines express all 6 PIM protein isoforms to varying extents, and we describe the properties of a novel pan-PIM inhibitor GNE-652 with picomolar biochemical potency, an excellent selectivity profile, and favorable ADME properties. Myeloma cell lines exhibit a striking prevalence of response to GNE-652 (23 of 25 lines with IC50 < 1 micromolar, median < 0.1 micromolar) and synergy in combination with the PI3K inhibitor GDC-0941 (mean combination index values ~0.2 (n=25)). We used an unrelated compound GNE-568 which has a PIM-1,3 selective profile to test the hypothesis that PIM-2 may have a non-redundant role in myeloma cells. GNE-568 while having cellular potency against PIM-1 and PIM-3, did not have single agent activity in myeloma cell lines nor did it act synergistically with GDC-0941 (n=10 cell lines). Interestingly, PI3K and AKT inhibitors showed the greatest extent of synergy with GNE-652, whereas mTOR-selective inhibitors were synergistic to a lesser extent. Standard of care agents dexamethasone, revlimid, velcade, and melphalan also combined well with GNE-652, but to a lesser extent and not as broadly. The synergistic anti-tumor activity of GNE-652 and PI3K inhibitor GDC-0941 on cell lines or on primary myeloma bone marrow aspirates in vitro was associated with cell-cycle arrest and marked apoptosis. In addition, we found 4 of 4 myeloma xenograft mouse models tested with GNE-562 and GDC-0941 showed excellent combination efficacy that correlated with modulation of the expected pharmacodynamic markers. These results provide the rationale for further preclinical development of PIM inhibitors and provide the basis for a possible clinical development plan in multiple myeloma. Disclosures: Ebens: Genentech: Employment, Equity Ownership. Berry:Genentech: Employment, Equity Ownership. Chen:Genentech: Employment, Equity Ownership. Deshmukh:Genentech: Employment, Equity Ownership. Drummond:Genentech: Employment, Equity Ownership. Du:Genentech: Employment, Equity Ownership. Eby:Genentech: Employment, Equity Ownership. Fitzgerald:Genentech: Employment, Equity Ownership. S. Friedman:Genentech: Employment, Equity Ownership. E. Gould:Genentech: Employment, Equity Ownership. Kenny:Genentech: Employment, Equity Ownership. Maecker:Genentech: Employment, Equity Ownership. Moffat:Genentech: Employment, Equity Ownership. Moskalenko:Genentech: Employment, Equity Ownership. Pacheco:Genentech: Employment, Equity Ownership. Saadat:Genentech: Employment, Equity Ownership. Slaga:Genentech: Employment, Equity Ownership. Sun:Genentech: Employment, Equity Ownership. Wang:Genentech: Employment, Equity Ownership. Yang:Genentech: Employment, Equity Ownership. Munugalavadla:Genentech: Employment, Equity Ownership.
3

Annaoui, Ilias, and Pascal Barneto. "Mécanismes de gouvernance et financement par Equity lines." Management & Avenir N° 135, no. 3 (June 6, 2023): 37–60. http://dx.doi.org/10.3917/mav.135.0037.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Ngo, Anh, Oscar Varela, and Xie Feixue. "The effects of lines of credit on market timing and the underpricing of seasoned equity offerings." Review of Accounting and Finance 18, no. 1 (February 11, 2019): 157–75. http://dx.doi.org/10.1108/raf-09-2016-0153.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
PurposeThis paper aims to examine the effects of lines of credit on a firm’s market timing behavior and the pricing of its seasoned equity offerings (SEOs). It shows that firms with lines of credit are more likely to time the equity market and receive less underpricing for their SEOs. It also shows that the propensity of firms with lines of credit to time the market is particularly significant for financially unconstrained firms. The results are robust to different measures of market timing and financial constraint, and these fill the gap in the literature that, to the best of the authors’ knowledge, has not examined the relation between lines of credit, market timing and value creation as related to equity offerings.Design/methodology/approachThe paper first investigates the relationship between lines of credit and the probability of a firm issuing SEOs using a logistic model. The paper then investigates whether firms with lines of credit engage in market timing behavior using ordinary least square regressions with two-way cluster-robust standard errors (standard errors that are robust to simultaneous correlation along two dimensions, such as firms and time) with two measures of market timing and two measures for financial constraints. Finally, the paper examines the relationship between lines of credit and SEO underpricing.FindingsIt was found that firms with lines of credit are more likely to time the equity market, perhaps driven by the financing flexibility resulting from the existence of their lines of credit. This finding comes mainly from financially unconstrained firms, as such an effect is not observed among financially constrained firms with lines of credit. It is further shown that firms with lines of credit are more likely to experience less severe equity underpricing, perhaps owing to market timing behavior. The results provide evidence on how lines of credit may create value to a firm through its market timing.Originality/valueThe paper sheds new light on how lines of credit may create value to a firm through the market timing channel.
5

Thomenius, Michael J., Jennifer Totman, Kat Cosmopoulos, Dorothy Brach, Lei Ci, Neil Farrow, Jesse J. Smith, et al. "Identification of a First-in-Class SETD2 Inhibitor That Shows Potent and Selective Anti-Proliferative Activity in t(4;14) Multiple Myeloma: T(4;14) Multiple Myeloma Cells Are Dependent on Both H3K36 Di and Tri-Methylation." Blood 132, Supplement 1 (November 29, 2018): 3207. http://dx.doi.org/10.1182/blood-2018-99-110803.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract t(4;14) chromosome translocations are found in 15% of newly diagnosed multiple myeloma (MM) cases and are associated with high risk. MM cells with t(4;14) over-express the histone methyltransferase (HMT), WHSC1/MMSET/NSD2, which leads to deregulation of gene expression due to increased di-methylation of Histone H3 at lysine 36 (H3K36me2). This activity has been shown to be essential for the survival of t(4;14) MM cells. In addition to WHSC1, another HMT, SETD2, has been shown to methylate H3K36. SETD2 is the only known enzyme capable of tri-methylation of H3K36 and has been reported to play a role in transcriptional elongation and alternative splicing. CRISPR pooled screening has shown that SETD2 activity is required for viability of a variety of cancer cell lines. This led Epizyme to develop small molecule inhibitors of SETD2 enzyme activity in order to understand the role of SETD2 in tumorigenesis. Through our drug discovery efforts, we identified EPZ-040414, a potent and selective inhibitor of SETD2 with low nM cell biochemical activity and broad selectivity against a panel of other HMTs. The proposed role of SETD2 in H3K36 methylation led us to test a panel of MM cells, including 6 t(4;14) cell lines with EPZ-040414. Inhibition of SETD2 resulted in reduced global tri-methylation of H3K36 in t(4;14) bearing MM cell lines. In contrast, there was no effect on global di-methyl H3K36 levels, indicating that WHSC1 activity is not affected by SETD2 inhibition. Moreover, 5/6 t(4;14) MM cell lines showed a cytotoxic response to treatment with EPZ-040414 with IC50s ranging between 60 and 200 nM, while all non-t(4;14) MM cell lines showed limited responses between 1 and 8 μM. Moreover, screening of a 280 cancer cell line panel with a SETD2 inhibitor showed minimal anti-proliferative activity in most cell lines tested. These findings show that t(4;14) MM cell lines require SETD2 activity for survival, suggesting that SETD2 inhibitors are strong candidates for the treatment of this high risk subgroup of MM. Efforts to further understand the interaction between SETD2 and WHSC1 in the molecular pathogenesis of t(4;14) myeloma will be presented. The current chemical series represented by EPZ-040414 is potent, selective, orally available, and currently under further evaluation for its therapeutic potential. Figure. Figure. Disclosures Thomenius: Epizyme Inc.: Employment, Equity Ownership. Totman:Epizyme Inc.: Employment, Equity Ownership. Cosmopoulos:Epizyme Inc.: Employment, Equity Ownership. Brach:Epizyme Inc.: Employment, Equity Ownership. Ci:Epizyme Inc.: Employment, Equity Ownership. Farrow:Epizyme Inc.: Employment, Equity Ownership. Smith:Epizyme Inc.: Employment, Equity Ownership. Chesworth:Epizyme Inc.: Employment, Equity Ownership. Duncan:Epizyme Inc.: Employment, Equity Ownership. Tang:Epizyme Inc.: Employment, Equity Ownership. Riera:Epizyme Inc.: Employment, Equity Ownership. Lampe:Epizyme Inc.: Employment, Equity Ownership.
6

Munugalavadla, Veerendra, Leanne Berry, Yung-Hsiang Chen, Gauri Deshmukh, Jake Drummond, Changchun Du, Michael Eby, et al. "A Selective PIM Kinase Inhibitor Is Highly Active In Multiple Myeloma: Mechanism of Action and Signal Transduction Studies." Blood 116, no. 21 (November 19, 2010): 4084. http://dx.doi.org/10.1182/blood.v116.21.4084.4084.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 4084 Related work from our group has shown the therapeutic utility of PIM inhibition in multiple myeloma cell lines, xenografts, and primary patient samples (Ebens A. et al., ASH 2010 submitted abstr.). In this study we provide detailed mechanistic findings to show that PIM kinase inhibition co-regulates several important elements of the PI3K/AKT/mTOR pathway, resulting in significant synergy for combination drug treatments. The PIM kinases are a family of 3 ser/thr growth factor- & cytokine-induced proteins hypothesized to have redundant survival and growth functions. GNE-652 is a pan-PIM kinase inhibitor with picomolar biochemical potencies and an excellent kinase selectivity profile. Myeloma cell lines exhibit sensitivity to single agent PIM inhibition and a striking synergy in combination with the PI3K inhibitor GDC-0941. Cells respond to this combination with cell cycle arrest and marked apoptosis in vitro. We tested a panel of selective PI3K/AKT/mTOR inhibitors and found PI3K and AKT inhibitors showed the greatest extent of synergy with GNE-652, whereas mTOR inhibitors were synergistic to a lesser extent. These results suggest that PIM signaling converges on both TORC1 and AKT to generate these differential synergies. BAD is a negative regulator of both Bcl-2 and Bcl-XL, and we were able to confirm previous reports that AKT and PIM cooperate to inactivate BAD (Datt et al., 1997; Yan et al., 2003). Pim has been shown to potentially inactivate PRAS40, a negative regulator of TORC1 (Zhang et al., 2009). We demonstrate that PIM or PI3K inhibition caused a loss of phosphorylation on PRAS40 and results in a physical association of PRAS40 and TORC1 and a decrease in phosphorylated p70S6K and S6RP. These reductions were apparent in 7 of 7 cell lines assayed and enhanced by the combination of PI3K and PIM inhibition in these cell lines. Consistent with prior reports (Hammerman et al., 2005), we show that a second node of convergence between PIM and TORC1 is 4E-BP1. Both GDC-0941 and GNE-652 treatments reduced phosphorylation of 4E-BP1 in 7 of 7 myeloma cell lines. Since dephosphorylated 4E-BP1 competes with eIF4G for the mRNA cap binding factor eIF4E, we assayed immunoprecipitates of eIF4E for the presence of eIF4G and 4E-BP1 and observed increased BP1 and decreased 4G. The combination treatment significantly enhanced the loss of 4G relative to either single agent, and importantly, even at 5× the IC50 concentrations for single agents, combination drug treatment achieved greater extent of effect than single agent treatment. Thus PI3K and PIM pathways are redundant at the level of cap-dependent translational initiation mediated by eIF4E. It has been hypothesized a subset of mRNAs are particularly sensitive to inhibition of cap-dependent translation, and that this includes a number of oncogenes such as cyclin D1. We assayed global protein synthesis in MM1.s cells using 35S-methionine and as expected we observed only a modest ≂∼f20% decrease caused by either GNE-652 or GDC-0941 and this decrease was not enhanced by combination treatment. However, we noted across 7 different myeloma cell lines, strong decreases in levels of cyclin D1 that were enhanced by combination treatment. In summary, we have identified several points at which PIM and PI3K/AKT/mTOR converge to provide synergistic apoptosis in multiple myeloma cell lines. These results provide the rationale for further preclinical development of PIM inhibitors and provide the basis for a possible clinical development plan in multiple myeloma. Disclosures: Munugalavadla: Genentech: Employment, Equity Ownership. Berry:Genentech: Employment, Equity Ownership. Chen:Genentech: Employment, Equity Ownership. Deshmukh:Genentech: Employment, Equity Ownership. Drummond:Genentech: Employment, Equity Ownership. Du:Genentech: Employment, Equity Ownership. Eby:Genentech: Employment, Equity Ownership. Fitzgerald:Genentech: Employment, Equity Ownership. S.Friedman:Genentech: Employment, Equity Ownership. E.Gould:Genentech: Employment, Equity Ownership. Kenny:Genentech: Employment, Equity Ownership. Maecker:Genentech: Employment, Equity Ownership. Moffat:Genentech: Employment, Equity Ownership. Moskalenko:Genentech: Employment, Equity Ownership. Pacheco:Genentech: Employment, Equity Ownership. Saadat:Genentech: Employment, Equity Ownership. Slaga:Genentech: Employment, Equity Ownership. Sun:Genentech: Employment, Equity Ownership. Wang:Genentech: Employment, Equity Ownership. Yang:Genentech: Employment, Equity Ownership. Ebens:Genentech Inc: Employment, Equity Ownership.
7

Dornan, David, Bart Burington, Peng Yue, Xiaoyan Shi, Ranjana Advani, Nancy Yu, Jeffrey T. Lau, et al. "CD40 Pathway Activation Status Predicts Response to CD40 Targeted Therapy in Diffuse Large b-Cell Lymphoma." Blood 114, no. 22 (November 20, 2009): 2721. http://dx.doi.org/10.1182/blood.v114.22.2721.2721.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 2721 Poster Board II-697 The role of the CD40 pathway in B-cell cancers is rather enigmatic due to having polar opposite roles in tumor biology. Stimulation of the CD40 receptor can reduce cell viability of NHL cell lines and xenograft tumor models, whereas, in sharp contrast, constitutive CD40 stimulation can actually promote proliferation of NHL cell lines, as well as drive lymphomagenesis and transformation of primary B-cells. Therefore, targeting the CD40 pathway has been challenging due to a knowledge gap in understanding the factors that may dictate a pro- or anti-tumor role. To address this directly, we employed a large panel of NHL cell lines and xenografts models to simulate a patient population and to define the molecular parameters to test in the context of clinical trials. Using baseline gene expression profiling, we found that the in vitro activity of an antibody that stimulates the CD40 pathway, Dacetuzumab (SGN-40), correlated strikingly with a transcriptional profile that was representative of an inactive CD40 pathway. Resistant cells, on the other hand, displayed a transcriptional profile indicative of an activated CD40 pathway. Consistent with these observations, constitutive phosphorylation of ERK, a node downstream of CD40 signaling, was present predominantly in cell lines that were resistant to Dacetuzumab. Fifteen genes, based on their association with in vitro activity and their biology as components of the CD40 network, were selected as a classifier from these pre-clinical data and independently tested in a panel of cell lines and xenografts models. Testing the selected gene classifier by qRT-PCR from baseline fixed tumor tissue of 39 patients treated with Dacetuzumab in phase I and II clinical trials revealed the ability to predict sensitivity in patients, as determined by tumor shrinkage >10%, with an overall accuracy of 80%. Consistent with the observed tumor shrinkage, the classifier positive, predicted to respond to Dacetuzumab, patients also exhibited a prolonged progression free survival (PFS) compared to classifier negative patients, predicted not to respond to Dacetuzumab (HR = 0.23, p = 0.001). Finally, using a novel microarray approach to genome-wide expression profiling on fixed tumor tissue revealed that CD40 pathway activation was indeed an important predictor of response. Overall, these data suggest that sensitivity to Dacetuzumab can be predicted by pre-determining the CD40 pathway activation status of tumors. Disclosures: Dornan: Genentech, Inc.: Employment, Equity Ownership. Burington:Genentech, Inc.: Employment, Equity Ownership. Yue:Genentech, Inc.: Employment, Equity Ownership. Shi:Genentech, Inc.: Employment, Equity Ownership. Advani:Seattle Genetics, Inc.: Research Funding. Yu:Genentech, Inc.: Employment, Equity Ownership. Lau:Genentech, Inc.: Employment, Equity Ownership. Yu:Genentech, Inc.: Employment, Equity Ownership. De Vos:Seattle Genetics, Inc.: Research Funding. Forero-Torres:Seattle Genetics, Inc.: Research Funding. Modrusan:Genentech, Inc.: Employment, Equity Ownership. Koeppen:Genentech, Inc.: Employment, Equity Ownership. Yang:Genentech, Inc.: Employment, Equity Ownership. Du:Genentech, Inc.: Employment, Equity Ownership. Elkins:Genentech, Inc.: Employment, Equity Ownership. Polson:Genentech, Inc.: Employment, Equity Ownership. Offringa:Genentech, Inc.: Employment, Equity Ownership. Whiting:Seattle Genetics, Inc.: Employment, Equity Ownership. Ebens:Genentech, Inc.: Employment, Equity Ownership.
8

SALANDRO, DAN, and WILLIAM B. HARRISON. "Determinants of the Demand for Home Equity Credit Lines." Journal of Consumer Affairs 31, no. 2 (December 1997): 326–45. http://dx.doi.org/10.1111/j.1745-6606.1997.tb00394.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Liu, Yang, and J. Jimmy Yang. "Private debt, unused credit lines, and seasoned equity offerings." Quarterly Review of Economics and Finance 51, no. 4 (November 2011): 376–88. http://dx.doi.org/10.1016/j.qref.2011.07.006.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Nadelson, Louis S., Rachelle G. Miller, Helen Hu, Na Mi Bang, and Brandy Walthall. "Is Equity on Their Mind? Documenting Teachers’ Education Equity Mindset." World Journal of Education 9, no. 5 (October 17, 2019): 26. http://dx.doi.org/10.5430/wje.v9n5p26.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
An education equity mindset is fundamental to assuring all students are supported to achieve to their highest capacity.We have identified six attributes of an educational equity mindset critical to assuring teachers’ practices andprofessional choices are aligned with meeting the needs of all students. In an effort to promote education equitymindset among teachers we determined there was a need to first empirically document the construct. We surveyed 452teachers in the southern region of the United States. Our results indicate that we effectively measured our definition ofeducation equity mindset. We found multiple differences in the mindset attributes based on personal and professionalvariables. Our data indicate that teachers may hold competing or fragmented mindsets. Our research uncoveredmultiple needed lines of research and implications for teacher preparation and professional development.
11

Eastman, Stephen, Christopher Shelton, Ira Gupta, Julie Krueger, Christina Blackwell, and Paul M. Bojczuk. "Synergistic Activity of Belantamab Mafodotin (anti-BCMA immuno-conjugate) with PF-03084014 (gamma-secretase inhibitor) in Bcma-Expressing Cancer Cell Lines." Blood 134, Supplement_1 (November 13, 2019): 4401. http://dx.doi.org/10.1182/blood-2019-123705.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Multiple myeloma (MM) is a plasma cell malignancy characterized by clonal proliferation of plasma cells within the bone marrow. B cell maturation antigen (BCMA) is a cell-surface receptor required for the survival of plasma cells and is also ubiquitously expressed on MM cells. Belantamab Mafodotin (GSK2857916) is a humanised monoclonal anti-BCMA antibody, which is afucosylated and conjugated to the microtubule-disrupting agent monomethyl auristatin-F (MMAF). Upon binding to BCMA on the cell surface, belatamab mafodotin is rapidly internalised and the cytotoxic moiety (cys-mcMMAF) is released, leading to direct cell death. BCMA is directly shed from the plasma membrane by gamma-secretase, a type-I sheddase. In order to further enhance belantamab mafodotin activity, we sought to increase cell surface levels of BCMA by blocking shedding of BCMA with a gamma-secretase inhibitor (GSI). We then determined the effect on the activity of belantamab mafodotin by combining belantamab mafodotin with PF-03084014, a highly-selective GSI. In order to understand combination effects against immuno-conjugate activity, a 3-day proliferation assay on a panel of multiple myeloma and lymphoma cell lines with varying levels of BCMA expression was conducted. The assay showed a 50 to 3,000-fold EC50 shift in cell lines sensitive to belantamab mafodotin across multiple lymphoma cell types. Antibody-dependent cellular cytotoxicity (ADCC) activity of belantamab mafodotin in combination with PF-03084014 was also examined. In a 24-hour ADCC Jurkat reporter assay, an EC50 shift across multiple BCMA-expressing cell lines was observed. Even cell lines with very low BCMA expression, such as Raji, showed a synergistic increase in ADCC activity in combination with PF-03084014. Cell lines that were non-responsive in the cell proliferation assay, showed activity in the ADCC assay, indicating low-expressing BCMA cell lines remain sensitive to belantamab mafodotin, alone and in combination with PF-03084014. Synergistic effect from this preclinical work provided rationale to support clinical evaluation of belantamab mafodotin in combination with PF-03084014 in a planned clinical trial (DREAMM-5). Disclosures Eastman: GlaxoSmithKline: Employment, Equity Ownership. Shelton:GlaxoSmithKline: Employment, Equity Ownership. Gupta:GlaxoSmithKline: Employment, Equity Ownership. Krueger:GlaxoSmithKline: Employment, Equity Ownership. Blackwell:GlaxoSmithKline: Employment, Equity Ownership. Bojczuk:GlaxoSmithKline: Employment, Equity Ownership.
12

Ulanet, Danielle, Victor Chubukov, John Coco, Gabrielle McDonald, Mya Steadman, Rohini Narayanaswamy, Sebastien Ronseaux, et al. "Hematologic Malignancies Exhibit Selective Vulnerability to Inhibition of De Novo Pyrimidine Biosynthesis By AG-636, a Novel Inhibitor of Dihydroorotate Dehydrogenase in Phase 1 Clinical Trials." Blood 134, Supplement_1 (November 13, 2019): 1570. http://dx.doi.org/10.1182/blood-2019-123471.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Rapidly proliferating cells reprogram metabolism to support increased biosynthetic demands, a feature that can expose targetable vulnerabilities for therapeutic intervention. A chemical biology screen was performed in an effort to identify metabolic vulnerabilities in particular tumor subtypes, and revealed potent and selective activity of a novel dihydroorotate dehydrogenase (DHODH) inhibitor, AG-636, in cancer cell lines of hematologic origin. In contrast, cancer cell lines of solid tumor origin exhibited comparatively poor sensitivity. Evaluation of a lymphoma cell line panel demonstrated broad responsiveness to DHODH inhibition, independent of clinical subtype (e.g. ABC, GCB, double-hit). The on-target cellular activity of AG-636 was evaluated by examining the metabolic effects of AG-636 on cells and by evaluating the ability of extracellular uridine to rescue the effects of AG-636 on proliferation and viability. The metabolic changes incurred upon treatment of cells with AG-636 were consistent with a mechanism of action driven by inhibition of DHODH and de novo pyrimidine biosynthesis. Supraphysiologic concentrations of extracellular uridine rescued the effects of AG-636 on growth and viability as well as the effects on metabolism, further confirming on-target activity. The mechanistic basis for differential sensitivity to AG-636 was assessed by comparing the activity of the de novo pyrimidine biosynthesis and uridine salvage pathways in cancer cell lines of hematologic or solid tumor origin with similar proliferative rates. Differential response to AG-636 could not be attributed to varying abilities to utilize the de novo pyrimidine biosynthesis pathway or to salvage extracellular uridine. Real-time imaging of cells treated with AG-636, along with monitoring of extracellular uridine concentrations, demonstrated immediate effects on the viability of lymphoma cell lines in the setting of depleted extracellular uridine. In contrast, solid tumor cell lines were able to maintain growth for an additional period of time, suggestive of adaptive mechanisms to supply pyrimidine pools and/or to cope with nucleotide stress. The high in vitro activity of AG-636 in cancer cells of hematologic origin translated to xenograft models, including an aggressive, patient-derived xenograft model of triple-hit lymphoma and an ibrutinib-resistant model of mantle cell lymphoma in which complete tumor regression occurred. These studies support the development of AG-636 for the treatment of hematologic malignancies. A phase 1 study has been initiated in patients with relapsed/refractory lymphoma (NCT03834584). Disclosures Ulanet: Agios: Employment, Equity Ownership. Chubukov:Agios: Employment, Equity Ownership. Coco:Agios: Employment, Equity Ownership. McDonald:Agios: Employment, Equity Ownership. Steadman:Agios: Employment, Equity Ownership. Narayanaswamy:Agios: Employment, Equity Ownership. Ronseaux:Agios: Employment, Equity Ownership. Choe:Agios: Employment, Equity Ownership. Truskowski:Agios: Employment, Equity Ownership. Nellore:Aurigene Discovery Technologies: Employment. Rao:Firmus Laboratories: Employment, Equity Ownership. Lenz:Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Agios: Research Funding; Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Gilead: Consultancy, Honoraria, Research Funding, Speakers Bureau; BMS: Consultancy; AstraZeneca: Consultancy, Honoraria, Research Funding; Bayer: Consultancy, Honoraria, Research Funding, Speakers Bureau; Roche: Employment, Honoraria, Research Funding, Speakers Bureau. Cooper:Agios: Employment, Equity Ownership. Murtie:Agios: Employment. Marks:Agios: Employment, Equity Ownership.
13

Sampath, Deepak, Elizabeth Punnoose, Erwin R. Boghaert, Lisa Belmont, Jun Chen, Franklin Peale, Nguyen Tan, et al. "Expression of Bcl-2 Pro-Survival Family Proteins Predicts Pharmacological Responses to ABT-199, a Novel and Selective Bcl-2 Antagonist, in Multiple Myeloma Models." Blood 120, no. 21 (November 16, 2012): 5028. http://dx.doi.org/10.1182/blood.v120.21.5028.5028.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 5028 Multiple myeloma (MM) is a hematological malignancy of the bone marrow caused by the dysregulated proliferation of monoclonal antibody producing plasma cells. A hallmark feature of cancer is the ability to evade cell death signals induced by stress response cues. The Bcl-2 family of proteins regulates the intrinsic apoptosis pathways and consists of pro-apoptotic (Bax, Bak, Bad, Bim, Noxa, Puma) and pro-survival (Bcl-2, Bcl-xL, Mcl-1); the balance of which dictates the life or death status of MM tumor cells. Thus, there is a strong rationale to target members of the Bcl-2 proteins for the treatment of MM. ABT-199 is a potent BH3-only mimetic that selectively antagonizes Bcl-2 and is currently in phase I clinical trials for the treatment of hematological malignancies. Therefore, we evaluated the efficacy of ABT-199 as a single agent and in combination with standard of care drugs such as Velcade (bortezomib) in preclinical models of MM. A panel of 21 human MM cell lines was evaluated in vitro for to sensitivity to ABT-199. ABT-199 potently inhibited cell viability in a sub-set of MM cell lines (7/21) with EC50 values less than 1 μM. Expression of Bcl-2, Bcl-xL, Mcl-1, Bim and other Bcl-2 family proteins were evaluated by protein and mRNA. Cell line modeling identified thresholds for expression of Bcl-2, Bcl-xL and Mcl-1 that best predicted sensitivity and resistance to ABT-199 and the dual Bcl-2/Bcl-xL antagonist, navitoclax. Consistent with the target inhibition profile of these drugs, we found that MM lines that were Bcl-2high/Bcl-xLlow/Mcl-1low are the most sensitive to ABT-199 treatment. Whereas cell lines that are Bcl-xLhigh remain sensitive to navitoclax but not ABT-199. MM cell lines that are Mcl-1high are less sensitive to both ABT-199 and navitoclax, suggesting that Mcl-1 is a resistance factor to both drugs. Utilizing a novel Mesoscale Discovery based immunoassay we determined that levels of Bcl-2/Bim complexes also correlated with sensitivity of ABT-199 in the MM cell lines tested. In addition, the t(11;14) status in these cell lines associated with sensitivity to ABT-199. The clinical relevance of the Bcl-2 pro-survival expression pattern in MM cell lines, was determined by a collection of bone marrow biopsies and aspirates (n=27) from MM patients by immunohistochemistry for prevalence of Bcl-2 and Bcl-xL. Similar to our in vitro observations, the majority (75%) of the MM bone marrow biopsies and aspirates had high Bcl-2 levels whereas 50% had high Bcl-xL expression. Therefore, a subset of patient samples (33%) were identified with a favorable biomarker profile (Bcl-2high/Bcl-xLlow) that may predict ABT-199 single agent activity. ABT-199 synergized with bortezomib in decreasing cell viability in the majority of MM cell lines tested in vitro based on the Bliss model of independence analyses (Bliss score range = 10 to 40). However the window of combination activity was reduced due to high degree of sensitivity to bortezomib alone. Therefore, the combination efficacy of ABT-199 and bortezomib was further evaluated in vivo in MM xenograft models that expressed high levels of Bcl-2 protein (OPM-2, KMS-11, RPMI-8226, H929 and MM. 1s). Bortezomib treatment alone at a maximum tolerated dose resulted in tumor regressions or stasis in all xenograft models tested. ABT-199 at a maximum tolerated dose was moderately efficacious (defined by tumor growth delay) as a single agent in xenograft models that expressed high protein levels of Bcl-2 but relatively lower levels of Bcl-xL. However, the combination of ABT-199 with bortezomib significantly increased the overall response rate and durability of anti-tumor activity when compared to bortezomib, resulting in increased cell death in vivo. Treatment with bortezomib increased levels of the pro-apoptotic BH3-only protein, Noxa, in MM xenograft models that expressed high levels of Mcl-1. Given that the induction of Noxa by bortezomib results in neutralization of Mcl-1 pro-survival activity in MM models [Gomez-Bougie et al; Cancer Res. 67:5418–24 (2007)], greater efficacy may be achieved when Bcl-2 is antagonized by ABT-199 thereby inhibiting pro-survival activity occurring through either Bcl-2 or Mcl-1 and increasing cell death. Thus, our preclinical data support the clinical evaluation of ABT-199 in combination with bortezomib in MM patients in which relative expression of the Bcl-2 pro-survival proteins may serve as predictive biomarkers of drug activity. Disclosures: Sampath: Genentech: Employment, Equity Ownership. Punnoose:Genentech: Employment, Equity Ownership. Boghaert:Abbott Pharmaceuticals: Employment, Equity Ownership. Belmont:Genentech: Employment, Equity Ownership. Chen:Abbott Pharmaceuticals: Employment, Equity Ownership. Peale:Genentech: Employment, Equity Ownership. Tan:Genentech: Employment, Equity Ownership. Darbonne:Genentech: Employment, Equity Ownership. Yue:Genentech: Employment, Equity Ownership. Oeh:Genentech: Employment, Equity Ownership. Lee:Genentech: Employment, Equity Ownership. Fairbrother:Genentech: Employment, Equity Ownership. Souers:Abbott Pharmaceuticals: Employment, Equity Ownership. Elmore:Abbott Pharmaceuticals: Employment, Equity Ownership. Leverson:Abbott Pharmaceuticals: Employment, Equity Ownership.
14

Erickson-Miller, Connie L., Jennifer Kirchner, Kodandaram Pillarisetti, Lone Ottesen, Yasser Mostafa Kamel, Yuan Liu, Anne-Marie Martin, and Conrad Messam. "Eltrombopag Decreases Proliferation of Ovarian, Lung and Breast Tumor Cell Lines." Blood 114, no. 22 (November 20, 2009): 2409. http://dx.doi.org/10.1182/blood.v114.22.2409.2409.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 2409 Poster Board II-386 Background: Eltrombopag (Promacta®) is a novel, oral thrombopoietin receptor (TpoR) agonist that interacts with the TpoR on bone marrow progenitors to stimulate megakaryocyte production, thus increasing platelet counts in thrombocytopenic patients. The effects of eltrombopag on the proliferation of solid tumor cell lines and the expression of thrombopoietin receptor (MPL, TpoR) on patient tumors is of interest given that chemotherapy can cause thrombocytopenia. Materials and methods: Proliferation was measured by Cell Titer Glo assay on 3 ovarian (OVCAR3, OVCAR4, SKOV3), 4 lung (A549, NCI-H226, NCI-H510, NCI-H460) and 3 breast (BT-474, MCF7, HCC1937) cancer cell lines from the ATCC treated with 0.01 – 100 ug/mL eltrombopag. Quantitative RT-PCR (qRT-PCR) for MPL expression was performed on the tumor cell lines and on 40 tumor samples, each from subjects with ovarian, lung or breast cancer. Microarray analysis for MPL mRNA expression was examined from 118 subjects with breast cancer and 29 with non-small cell lung cancer (NSCLC). Microarray data was normalized using robust multiarray average (RMA) and relative mRNA expression was determined. To determine expression of TpoR protein, western blot analyses was performed on some of the tumor cell lines. Results: Eltrombopag induced an inhibition of proliferation on all of the ovarian, lung and breast solid tumor cell lines tested. The IC50 ranged from 3.7 to 49.7 ug/mL (see table below). The Cmax of ITP patients treated with 75 mg eltrombopag is 11.4 ug/mL, demonstrating that these concentrations are clinically achievable. There was no enhancement of proliferation at any concentration of eltrombopag, consistent with the very low or undetectable level of MPL expression on samples of tumors from patients with these diseases. MPL was expressed at very low or undetectable levels in these tumor cell lines with the exception of the lung cancer line, NCI-H510. However, western blot analyses showed no detectable TpoR protein expression regardless of the higher levels of MPL mRNA in NCI-H510 cells. Erythropoietin receptor (EPOR) mRNA was expressed at low-to-moderate levels, while ERBB2 and IGF1R were expressed at higher levels in these cell lines. Microarray analysis showed undetectable MPL mRNA levels in all 118 samples from patients with breast cancer and 52% of the NSCLC samples, the remaining NSCLC samples expressed low levels of MPL. In contrast, EPOR was expressed in 75–100% of the breast cancer, and NSCLC samples. ERBB2 was expressed in 97–100% of the samples and IGF1R was expressed in 54–100% of the samples. When 40 other tumor samples each from subjects with ovarian, lung and breast cancer were examined by qRT-PCR, MPL mRNA levels were also very low or undetectable. EPOR, ERBB2, and IGF1R expression levels varied according to tumor type, but were greater than MPL levels. Conclusions: In summary, similar to its effects on leukemia and lymphoma cell lines, all of the nine lung, ovarian, breast or prostate tumor cell lines demonstrated decreased proliferation in response to eltrombopag. The undetectable or very low levels of expression of MPL mRNA in tumors of patients with lung, ovarian, breast or prostate cancer supports the proliferation results. Disclosures: Erickson-Miller: GlaxoSmithKline: Employment, Equity Ownership, Patents & Royalties, Research Funding. Kirchner:GlaxoSmithKline: Employment. Pillarisetti:GSK: Employment, Equity Ownership, Patents & Royalties. Ottesen:GSK: Employment, Equity Ownership. Mostafa Kamel:GSK: Employment, Equity Ownership. Liu:GSK: Employment, Equity Ownership. Martin:GSK: Employment, Equity Ownership. Messam:GSK: Employment, Equity Ownership.
15

Pramanik, Anik, Pan Xu, and Yifan Xu. "Equity Promotion in Public Transportation." Proceedings of the AAAI Conference on Artificial Intelligence 37, no. 10 (June 26, 2023): 11890–98. http://dx.doi.org/10.1609/aaai.v37i10.26403.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
There are many news articles reporting the obstacles confronting poverty-stricken households in access to public transits. These barriers create a great deal of inconveniences for these impoverished families and more importantly, they contribute a lot of social inequalities. A typical approach addressing the issue is to build more transport infrastructure to offer more opportunities to access the public transits especially for those deprived communities. Examples include adding more bus lines connecting needy residents to railways systems and extending existing bus lines to areas with low socioeconomic status. Recently, a new strategy is proposed, which is to harness the ubiquitous ride-hailing services to connect disadvantaged households with the nearest public transportations. Compared with the former infrastructure-based solution, the ride-hailing-based strategy enjoys a few exclusive benefits such as higher effectiveness and more flexibility. In this paper, we propose an optimization model to study how to integrate the two approaches together for equity-promotion purposes. Specifically, we aim to design a strategy of allocating a given limited budget to different candidate programs such that the overall social equity is maximized, which is defined as the minimum covering ratio among all pre-specified protected groups of households (based on race, income, etc.). We have designed a linear-programming (LP) based rounding algorithm, which proves to achieve an optimal approximation ratio of 1-1/e. Additionally, we test our algorithm against a few baselines on real data assembled by outsourcing multiple public datasets collected in the city of Chicago. Experimental results confirm our theoretical predictions and demonstrate the effectiveness of our LP-based strategy in promoting social equity, especially when the budget is insufficient.
16

Maira Muniz Almeida, Wanda, and Eliane Guaraldo. "Urban Green Equity." Life Style 8, no. 2 (June 2, 2022): 64–74. http://dx.doi.org/10.19141/2237-3756.lifestyle.v8.n2.p64-74.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
The growing concern with the impacts of climate change and the fast urbanization of cities has led international policies to guide changes in attitudes by governments and civil society, directing effective models of sustainable governance aimed at environmental health and equal access for society environmental benefits. In this study, we seek to understand the evolution of research and scientific production on a topic of great relevance today – green equity. Through scientometric analysis based on a systematic literature review, we analyzed articles published in the period between 1992 and 2021. We used the open source R-tool Biblioshiny, which processes information from academic databases to carry out the analyses. Through this methodology it was possible to identify the main fields of research and relate the results obtained with important historical milestones for sustainable development. We also seek to highlight the evolution of research lines and highlight the significant flows in the global collaboration network. We found that the theme of green equity was driven by international agendas such as the SDG and has been gaining more space in scientific production, linked to a greater variety of issues such as climate change, accessibility to green spaces, ecosystem services, green infrastructure and socioeconomic issues.
17

Prabhu, Varun V., Rohinton S. Tarapore, Mathew J. Garnett, Ultan McDermott, Cyril H. Benes, Jessica M. Wagner, Wafik S. El-Deiry, Martin Stogniew, Wolfgang Oster, and Joshua E. Allen. "Potent Anti-Leukemic Effects of Small Molecule ONC212, a Member of the Imipridone Class of Anti-Cancer Compounds." Blood 128, no. 22 (December 2, 2016): 5133. http://dx.doi.org/10.1182/blood.v128.22.5133.5133.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract ONC201, the founding member of the imipridone class of anti-cancer compounds, is a highly selective small molecule GPCR antagonist that is in Phase I/II advanced cancer clinical trials. In this study, we evaluated the anti-cancer effects of ONC212, an ONC201 analogue that possess the same unique core chemical structure shared by imipridones. The in vitro efficacy of ONC212 was assessed in the Genomics of Drug Sensitivity in Cancer collection of cell lines (>1,000 cell lines). Cell viability assays were performed to generate dose responses curves at concentrations from 78nM upto 20uM and at 72 hours post-treatment. ONC212 was broadly efficacious across most solid tumors and hematological malignancies in the low nanomolar range. Ranking the ONC212 sensitivity dataset revealed that leukemia is the most responsive tumor type based on completeness of response (area under the dose-response curve, AUC). ONC212 was tested in 65 leukemia cell lines in this study that is comprised of acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML) and hairy cell leukemia. ONC212 demonstrated broad spectrum anti-leukemic activity and was equally efficacious across all leukemia subtypes tested, in terms of AUC. Most cell lines (63/65) were responsive to ONC212 with GI50 ranging from <78nM to 312nM. Within ALL, both B-cell and T-cell ALL were highly sensitive to ONC212. ONC212 reduced cell viability in AML independent of complex karyotypes that are associated with poor clinical prognoses. Thus, ONC212 possesses robust anti-leukemic activity irrespective of subtype and provides further validation of the anti-cancer efficacy of the novel imipridone class of small molecules. Disclosures Prabhu: Oncoceutics: Employment. Tarapore:Oncoceutics: Employment, Equity Ownership. El-Deiry:Oncoceutics: Equity Ownership. Stogniew:Oncoceutics Inc.: Employment, Equity Ownership. Oster:Oncoceutics: Employment, Equity Ownership. Allen:Oncoceutics: Employment, Equity Ownership.
18

Thakurta, Anjan, Anita K. Gandhi, Michelle Waldman, Chad C. Bjorklund, Suzanne Lentzsch, Stephen A. Schey, Yolanda Calle, et al. "Absence Of Mutation In Cereblon (CRBN) and DNA Damage Binding Protein 1 (DDB1) Genes In Myeloma Cells and Patients and Its Clinical Significance." Blood 122, no. 21 (November 15, 2013): 3139. http://dx.doi.org/10.1182/blood.v122.21.3139.3139.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Background CRBN, a target of thalidomide and IMiDs® immunomodulatory agents lenalidomide (LEN) and pomalidomide (POM), is a component of the E3 ubiquitin cullin 4 ring ligase (CRL4) complex that also includes DDB1, Roc1, and Cul4. Two CRBN mutations have been reported in multiple myeloma (MM) patients: truncating mutation (Q99) and point mutation (R283K). One copy of the CRBN gene was shown to be deleted in the MM1S and MM1S.R cell lines. No DDB1 mutation has been described previously. Results We investigated the incidence of CRBN and DDB1 mutations by next-generation sequencing in 20 MM cell lines and MM subjects. Of 90 MM patients, 24 were newly diagnosed and 66 were relapsed and refractory of which 36 patients were LEN resistant. Out of the cell lines tested, 1 heterozygous CRBN mutation (D249Y) was found in the LEN-resistant ANBL6R cells, which is located in the putative DDB1 binding domain, and 2 single silent mutations were identified in the KMS-12-BM (rs17027638) and OPM-2 cells. One DDB1 heterozygous mutation (E303D) was identified in ANBL6 cells. In the cohort of patients assessed, no CRBN mutation was detected; however, 5 single nucleotide variations (SNV) were identified. Three of the 5 SNVs were at position 735 (Y245Y) and 1 each at position 219 (H73H) and 939 (C313C), respectively. The first 2 SNVs (rs17027638 and rs1045309) are described but not the last. We found a single SNV (P51P; rs2230356) in DDB1 gene the patient samples. Conclusion Mutations within the coding sequences of CRBN and DDB1 are rare in MM patients and cell lines. Most intrinsically LEN-resistant cells and cell lines made resistant to LEN or POM do not have CRBN or DDB1 mutations, suggesting the potential role of other sources, such as genetic or epigenetic pathways in developing resistance to IMiD drug–based therapy. Disclosures: Thakurta: Celgene: Employment, Equity Ownership. Gandhi:Celgene: Employment, Equity Ownership. Waldman:Celgene: Employment, Equity Ownership. Bjorklund:Celgene: Employment, Equity Ownership. Lentzsch:Celgene: Research Funding. Schey:Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau; NAPP: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau; BMS: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau. Orlowski:Bristol-Myers Squibb: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Millennium: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Onyx: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Resverlogix: Research Funding; Array: Honoraria, Membership on an entity’s Board of Directors or advisory committees; Genentech: Honoraria, Membership on an entity’s Board of Directors or advisory committees; Merck: Membership on an entity’s Board of Directors or advisory committees. Madan:Covance Genomics Lab: Employment. Ning:Celgene: Employment, Equity Ownership. Mendy:Celgene: Employment, Equity Ownership. Lopez-Girona:Celgene: Employment, Equity Ownership. Schafer:Celgene: Employment, Equity Ownership. Avet-Loiseau:Celgene: Research Funding. Chopra:Celgene: Employment, Equity Ownership.
19

Arora, Shilpi, Kaylyn Williamson, Shruti Apte, Srividya Balachander, Jennifer Busby, Charlie Hatton, Barbara Bryant, and Patrick Trojer. "EZH2 Inhibitors Are Broadly Efficacious in Multiple Myeloma As Single Agent and in Combination with Standard of Care Therapeutics." Blood 128, no. 22 (December 2, 2016): 5672. http://dx.doi.org/10.1182/blood.v128.22.5672.5672.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Post-translational modifications of the histone proteins play a key role in regulating processes that require access to DNA. Specifically, methylation of lysine 27 on histone 3 (H3K27) is intimately linked with transcriptional repression. EZH2, a histone lysine methyl transferase is the catalytic component of the PRC2 complex, which catalyzes H3K27 methylation. EZH2 dysregulation has been observed in different malignancies and inhibition of its catalytic activity has emerged as a novel therapeutic approach to treat human cancers. Potent, selective and reversible EZH2 small molecule inhibitors are currently being tested in Ph. 1 clinical trials. We and others have reported EZH2 dependencies across non-Hodgkin Lymphoma subtypes in cancer cell lines, in xenograft mouse models and in lymphoma patients. We identified Multiple Myeloma as potential clinical application for EZH2 inhibitors. Treatment with EZH2 inhibitors such as CPI-360, CPI-169 and CPI-1205 cause apoptosis in multiple myeloma and plasmacytoma cell models and causes tumor growth inhibition in myeloma xenograft models at well tolerated doses. An EZH2-controlled transcriptional signature across various multiple myeloma was identified using integrated RNA-sequencing and ChIP-sequencing data. Combination studies testing EZH2 inhibitors with standard of care (SOC) agents across a panel of multiple myeloma cell lines showed synergistic responses with several of the SOC agents in vitro and in vivo. Disclosures Arora: Constellation Pharmaceuticals: Employment, Equity Ownership. Williamson:Constellation Pharmaceuticals: Employment, Equity Ownership. Apte:Constellation Pharmaceuticals: Employment, Equity Ownership. Balachander:Constellation Pharmaceuticals: Employment, Equity Ownership. Busby:Constellation Pharmaceuticals: Employment, Equity Ownership. Hatton:Constellation Pharmaceuticals: Employment, Equity Ownership. Bryant:Constellation Pharmaceuticals: Employment, Equity Ownership. Trojer:Constellation Pharmaceuticals: Employment, Equity Ownership.
20

Wang, Xiaomin, and Wenxin Zhang. "Efficiency and Spatial Equity Impacts of High-Speed Rail on the Central Plains Economic Region of China." Sustainability 11, no. 9 (May 5, 2019): 2583. http://dx.doi.org/10.3390/su11092583.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Efficiency impacts can be assessed based on improvements in accessibility promoted by the high-speed rail (HSR) project, focusing mainly on major urban areas. Spatial equity impacts originate from changes in the distribution of accessibility levels observed across such cities. This study uses the weighted average travel time and coefficient of variation to explore the impact of HSR on efficiency and spatial equity within the Central Plains Economic Region (CPER) and to build an “efficiency-equity” model to identify optimal upgrading of conventional rail (CR) lines to improve the accessibility of the CPER and mitigate regional disparities. The results indicate that since the operation of HSR, accessibility levels across the CPER have improved by roughly 24.56%, and the spatial distribution of gains has been uneven since the most significant improvements have occurred in HSR cities and large cities. Inequality in regional accessibility has increased by 28.12%, creating a more heterogeneous pattern of accessibility. With goals of “efficiency” and “equity”, an examination of the upgrading of CR as an approach shows that “Xinxiang-Jiaozuo” and “Xinxiang-Jiaozuo-Sanmenxia-Yuncheng” are the best upgraded lines for improving the efficiency of the CPER and limiting regional disparities. The results of this study serve as supportive information for the planning and construction of HSR lines and networks and for underdeveloped regional transport system policymaking.
21

Ahsan, Aarif, Ann Polonskaia, Chih-Chao Hsu, Chad C. Bjorklund, Maria Ortiz Estevez, Fadi Towfic, Nizar J. Bahlis, et al. "Super-Enhancer Driven Regulation of CKS1B in Multiple Myeloma: Implications in Mediating Response to BET Inhibitor and Celmod Agent Combination." Blood 138, Supplement 1 (November 5, 2021): 2658. http://dx.doi.org/10.1182/blood-2021-150641.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Introduction: The Myeloma Genome Project (MGP) characterized the genomic landscape of patients with newly diagnosed multiple myeloma (NDMM) (Walker BA, et al. Blood 2018; 132[6]:587-597). Using a multi-omics unsupervised clustering approach, 12 molecularly-defined disease segments were identified (Ortiz M, et al. Blood 2018; 132[suppl 1]:3165). Here, we performed experimental validation of CDC28 Protein Kinase Regulatory Subunit 1B (CSK1B) that was identified as a putative target from the disease segment with poorest clinical outcome. CKS1B was selected for in-depth validation due to their role in cell cycle pathways associated with high-risk disease, biological mechanisms of chromosome 1q amplification and druggability. Methods: Association of CKS1B with outcomes was analyzed in NDMM patients, across relapses and with clinical outcome datasets from MGP and Mayo clinic. Inducible shRNAs of CKS1B and bromodomain containing protein 4 (BRD4, a member of the BET [bromodomain and extra terminal domain] family) were generated in MM cell lines. BRD4 and Aiolos ChIP-seq datasets were analyzed for binding on CKS1B gene. BRD4 inhibitors JQ1 and CC-90010 were utilized for inhibition studies in MM cell lines. Results: Higher expression of CKS1B was associated with significantly poorer PFS, OS, disease severity and relapse. Knock-down of CKS1B in MM cells led to a significant decrease in proliferation (P&lt;0.001) and enhanced apoptosis in MM cell lines. BRD4-ChIP sequencing studies revealed that the expression of CKS1B was regulated by super-enhancer (SE) associated elements. As expected, two BRD4 inhibitors, JQ1 and CC-90010 and inducible BRD4 shRNAs downregulated the expression of CKS1B resulting in decreased proliferation, cell cycle arrest and apoptosis in MM cell lines. Furthermore, MM cell lines harboring chromosome 1q gain/amp showed higher sensitivity to BRD4 inhibition compared to cell lines with normal 1q copy number. Mechanistic studies revealed that BRD4inh and BRD4 shRNAs downregulated the expression of Aiolos and Ikaros in MM cell lines. Interestingly, Aiolos ChIP-sequencing studies demonstrated the binding of Aiolos at the transcriptional start sites of CKS1B with the transcriptional activation mark. The immunomodulatory agent (IMiD ®) pomalidomide (Pom) transcriptionally downregulated CKS1B in Pom-sensitive cells downstream of Aiolos, Ikaros degradation. Based on these mechanisms, IMiD agents, lenalidomide, Pom and the novel Cereblon E3 ligase modulating degrader (CELMoD ®) agent CC-92480 in combination with BRD4inh promoted a synergistic decrease in proliferation, cell cycle arrest and increase in apoptosis in both Pom-sensitive and -resistant cell lines. The combination of IMiD or novel CELMoD agent with BRD4inh also promoted deeper downregulation of CKS1B, Aiolos, Ikaros, c-Myc and survivin proteins with enhanced levels of apoptotic marker cleaved Caspase 3 as compared to single agents alone. Conclusions: In summary, we have identified CKS1B as a key target associated with poor outcome in MM patients. Translational studies suggest a profound downregulation of CKS1B and key pro-survival effector proteins following combination treatment with BRD4inh and IMiD agents/novel CELMoD agents resulting in synergistic anti-tumor effects. These data provide rationale for testing these agents in the clinic for high-risk and IMiD-relapsed patients. Figure: Changes in cell proliferation and protein levels of key signaling mediators were studied in K12PE cell line treated with increasing doses of Lenalidomide, Pomalidomide and CC-92480 in combination with JQ1. Figure 1 Figure 1. Disclosures Ahsan: BMS: Current Employment, Current equity holder in publicly-traded company. Polonskaia: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Hsu: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Bjorklund: BMS: Current Employment, Current equity holder in publicly-traded company. Ortiz Estevez: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Towfic: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Bahlis: Takeda: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria; GlaxoSmithKline: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Genentech: Consultancy; Pfizer: Consultancy, Honoraria; BMS/Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria. Pourdehnad: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties: No royalty. Flynt: BMS: Current Employment, Current equity holder in publicly-traded company. Ahsan: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Thakurta: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties.
22

Chen, Janice, Christopher L. Brooks, Peter McDonald, Jonathan D. Schwartz, Rebecca S. Schneider, Keiichi Sakakibara, Naoya Saito, Takuji Sato, Takumi Kawabe, and Eric K. Rowinsky. "SL-801, a Novel, Reversible Inhibitor of Exportin-1 (XPO1) / Chromosome Region Maintenance-1 (CRM1) with Broad and Potent Anti-Cancer Activity." Blood 126, no. 23 (December 3, 2015): 4433. http://dx.doi.org/10.1182/blood.v126.23.4433.4433.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract XPO1/CRM1, the principal nuclear export protein in eukaryotic cells, is required for the nuclear-cytoplasmic transport of both proteins and RNAs. Overexpression of XPO1 is reported in many cancers, causing dysregulated protein localization, aberrant cell proliferation, and resistance to apoptosis, and is associated with aggressive characteristics and poor patient outcome. Recent work has revealed XPO1 to be a clinically relevant target, and nuclear export inhibitors have emerged as a new class of anti-cancer agents with clinical activity in multiple hematologic and solid malignancies. SL-801 is a novel small molecule that binds covalently to Cys528 of XPO1, blocking the ability of XPO1 to interact with substrate cargos (e.g., p53, FOXO, p21, p27, and others). In contrast to the prototypical XPO1 inhibitor leptomycin B, which binds irreversibly to XPO1 and caused significant toxicities in Phase 1 trials, SL-801 binding to XPO1 is reversible, a characteristic that may be exploited to maximize its therapeutic index. Exposure to SL-801 results in potent inhibition of XPO1-dependent nuclear export, cell cycle arrest, and induction of apoptosis in a time- and dose-dependent manner. Here, the anti-tumor activity of SL-801 was investigated against a panel of 240 cell lines representing a broad range of solid and hematologic malignancies and confirmed in several SCID xenograft models. The OncoPanelTM high content screening platform was used to evaluate the cytotoxicity of SL-801 against 205 solid tumor and 35 liquid tumor cell lines. SL-801 demonstrated potent activity, with 50% growth inhibitory (GI50) values ≤ 10 nM in 51/240 (21.3%) cell lines and GI50 values ≤ 100 nM in 230/240 (95.8%) cell lines. SL-801 sensitivity was independent of cell proliferation rate or XPO1 expression levels. While SL-801 was broadly cytotoxic, cell lines of hematopoietic origin exhibited greater sensitivity. GI50s in hematologic cancers ranged from 3-93 nM in leukemias, 1-103 nM in lymphomas, and 3-11 nM in multiple myelomas. SL-801 also inhibited solid tumor growth, with GI50s ≤ 10 nM in several breast, brain, cervical, ovarian, gastric, kidney, liver, lung, melanoma, prostate, and sarcoma lines. In addition, a 5-fold increase in active caspase-3 staining was observed at SL-801 concentrations ≤ 100 nM in 117/240 (48.8%) cell lines, consistent with induction of apoptosis. To understand tumor sensitivity to SL-801, results of the cell line cytotoxicity screen were analyzed against publicly available genomic datasets. This analysis revealed that SL-801 was cytotoxic towards cell lines regardless of mutation status of key oncogenes (e.g., KRAS) and tumor suppressor genes (e.g., TP53). The in vitro cytotoxicity of SL-801 against tumor cell lines was further validated in several xenograft models in SCID mice. In the RPMI-8226 multiple myeloma xenograft model, tumor growth was significantly inhibited at oral SL-801 doses of 31.25 mg/kg administered daily for five days for two weeks. In the ARH-77 human multiple myeloma xenograft model, overall survival was significantly prolonged by daily oral administration of 125 mg/kg SL-801 for ten days. This dose and regimen were well tolerated, and 90% of SL-801-treated mice survived > 150 days, whereas median survival was 39.5 days in the vehicle-treated group (p < 0.001). Significant tumor growth inhibition was also observed in the NCI-H226 non-small cell lung cancer and 22RV1 prostate cancer xenograft models. These data demonstrate that SL-801 is a promising clinical candidate that inhibits a novel, clinically validated target and support its clinical development in a broad range of oncologic indications. The reversible binding of SL-801 to XPO1 may offer the potential to develop dosing schedules to enable recovery in normal tissues, thus broadening the therapeutic index of this class of agents. IND-enabling work is underway to support entry into clinical trials, and a Phase I trial design will be discussed. Disclosures Chen: Stemline Therapeutics, Inc.: Employment, Equity Ownership. Brooks:Stemline Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. McDonald:Stemline Therapeutics, Inc.: Employment, Equity Ownership. Schwartz:Stemline Therapeutics, Inc.: Employment, Equity Ownership. Schneider:Stemline Therapeutics, Inc.: Employment. Sakakibara:CanBas Co., Ltd.: Employment, Equity Ownership. Saito:CanBas Co., Ltd.: Employment, Equity Ownership. Sato:CanBas Co., Ltd.: Employment, Equity Ownership, Patents & Royalties. Kawabe:CanBas Co., Ltd.: Employment, Equity Ownership, Patents & Royalties. Rowinsky:Stemline Therapeutics: Employment, Equity Ownership.
23

Liu, Yang. "The Sources of Debt Matter Too." Journal of Financial and Quantitative Analysis 41, no. 2 (June 2006): 295–316. http://dx.doi.org/10.1017/s0022109000002076.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
AbstractThis paper examines the effects of different types of private debt on firm cash balances, equity risk, and investment. Firms with more bank loans have more cash and investment, but lower equity risk. Firms with more nonbank private debt have more cash, lower equity risk, and less investment. Firms with more unused credit lines have less cash and lower equity risk, but greater investment. Results suggest that financial intermediaries' monitoring intensity increases with loan size. Depending on type, private debt mitigates information asymmetry or asset substitution, or both. Deposit relations associated with bank borrowing also contribute to banks' information advantage.
24

Peng, Mao Yu, Yasmin Abaza, Martina Mcdermott, Monica Mead, Dennis J. Slamon, and Sarah Larson. "Activity of Carfilzomib (CFZ) in Acute Myeloid Leukemia (AML) As a Single Agent and in Novel Combinations." Blood 136, Supplement 1 (November 5, 2020): 6–7. http://dx.doi.org/10.1182/blood-2020-141459.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Background:Recent advances in targeted therapy have expanded the available therapeutic optionsfor patients with AML. However, many patients still have suboptimal outcomes, particularly in the relapsed/refractory setting, underscoring the need for novel therapeutic strategies. Proteasome inhibitors (PIs), such as bortezomib, exhibit antitumor activity in AML through inhibition of the nuclear factor κB pathway and induction of apoptosis. CFZ, a second-generation PI, has preferential preclinical activity in AML compared to bortezomib making it an agent of interest in AML therapy. Here we assessed the activity of CFZ as a single agent and in novel combinations with Ara-C and/or other agents targeting potential vulnerabilities in AML cell lines. Methods:20 AML cell lines were treated with a single dose of CFZ for 7 days, proliferation inhibition was measured using an IC50 cutoff for CFZ of 10 nM. 2 sensitive (ML2 and MV411) and 2 resistant (AML193 and NOMO1) cell lines were selected for further analysis. Apoptosis, cell cycle, and cell senescence analysis were performed after 72 hours of CFZ exposure at 10 nM. Combination assays using CFZ 10 nM and Ara-C 200 nM were performed to evaluate for potential interaction in the form of antagonism or potentiation. Proteomic analysis was performed at baseline using reverse phase protein assay (RPPA). Cell lines were aligned according to CFZ IC50. Several proteins involved in various physiological pathways exhibited a potential correlation with CFZ sensitivity. Combination treatments with CFZ and agents targeting these pathways were carried out in selected cell lines. Results:Single-agent CFZ induced apoptosis with apoptotic rates &gt;85% in sensitive cell lines and only 10% in resistant cell lines. Similarly, CFZ resulted in G0/G1 cell cycle arrest in sensitive, but not resistant AML cell lines. Lack of difference in cellular senescence confirmed apoptosis as the major mechanism of CFZ-induced growth inhibition in AML cell lines. No antagonism was noted when CFZ was combined with Ara-C. RPPA revealed that AML cell lines with higher expression of autophagy-related proteins (Atgs) were more resistant to CFZ treatment. Combining autophagy inhibitor hydroxychloroquine (HCQ) or ROC-325 with CFZ produced a synergistic effect to induce apoptosis in several CFZresistant cell lines. RPPA also revealed that lower basal levels of fatty acid synthase (FASN), a key enzyme involved in lipogenesis, correlated with CFZ sensitivity and CFZ resistant lines tendedto have higher basal FASN levels. The combination of CFZ with a FASN inhibitor resulted in a significant synergistic apoptosis-inducing effect that was observed in the AML lines tested. Conclusion:CFZ demonstrated single agent activity in the nanomolar range in human AML cell lines. The addition of standard-of -care chemotherapy to CFZ did not show antagonism. Combining CFZ with agents targeting autophagy or lipid-metabolism showed synergistic effect in apoptosis. These results suggest a role for CFZ in combination therapeutic strategies for AML patients. Disclosures Mcdermott: TORL Biotherapeutics:Current equity holder in private company;1200 Pharma:Current equity holder in private company.Slamon:TORL Biotherapeutics:Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees;1200 Pharma:Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees;Novartis:Consultancy, Research Funding;Eli Lilly:Consultancy;Bayer:Consultancy, Research Funding;Pfizer:Consultancy, Other: stock, Research Funding;Syndax:Research Funding;Aileron:Research Funding;Genetech:Research Funding;Biomarin:Membership on an entity's Board of Directors or advisory committees;Seattle Genetics:Other: Stock;Amgen:Other: Stock.Larson:BMS, Bioline, Celgene, Juno, Janssen:Research Funding;TORL Biotherapeutics:Current equity holder in private company.
25

Zhang, Yue, Julie Parmentier, Zhongwu Lai, Greg O'Connor, Melissa Passino, Francoise Powell, Erik Devereaux, and Kate Byth. "Spleen Tyrosine Kinase Inhibitor Fostamatinib Blocks B Cell Receptor Signaling and Reduces Viability of BCR Subtype Diffuse Large B Cell Lymphoma." Blood 120, no. 21 (November 16, 2012): 3720. http://dx.doi.org/10.1182/blood.v120.21.3720.3720.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 3720 Spleen tyrosine kinase (SYK) plays a key role in B cell receptor mediated survival in certain B cell malignancies including Diffuse Large B Cell Lymphoma (DLBCL). Therefore, targeting SYK represents an emerging therapeutic approach for the treatment of DLBCL. Indeed, fostamatinib, an orally available SYK inhibitor, has shown promising clinical activity in non-Hodgkin lymphoma (Friedberg et al. 2010 Blood 115: 2578). However, the overall response rate for novel targeted agents in unselected relapsed refractory DLBCL patients remains 25–30%, highlighting the opportunity for development of better treatment strategies. Here, we used preclinical models to study the mechanism underlining the efficacy of R406, the metabolic active form of fostamatinib. R406 was characterized in a heterogeneous panel of 17 DLBCL cell lines including both ABC and GCB subtypes. Overall, the cellular selectivity of R406, particularly in ABC-subtype DLBCL cell lines, was consistent with other BCR-targeted agents such as BTK inhibitor PCI-32765 (ibrutinib) and PI3 kinase δ inhibitor CAL-101. This strongly suggests that R406 functions through inhibition of BCR-mediated survival signaling. Furthermore, phospho-flow analysis and Western Blotting have demonstrated the effect of R406 on both basal and anti-IgG/M stimulated BCR signaling. Specifically, R406 decreases phosphorylation of proximal BCR pathway regulators BLNK, PLCγ2, as well as key players in downstream effect pathways such as ERK, AKT, RPS6, 4EBP and STAT3. To investigate whether the inhibition of BCR signaling by R406 affects cell viability, flow-cytometry based apoptosis analysis was employed. R406 induced apoptosis in sensitive cell lines in a dose-dependent manner. Mechanistically, treatment with R406 reduced MCL1 protein level and down-regulated Bfl-1 expression in sensitive, but not resistant, cell lines, which may partially contribute to the observed efficacy. Given the critical role of NFκB-dependent survival signaling in ABC subtype DLBCL, we then investigated the effect of R406 on this pathway by monitoring NF-κB target gene expression via quantitative real time PCR. Consistent with previous publication (Davis et al. 2010, Nature 463: 88), R406 significantly blocked chronic active BCR-induced NFκB signaling in sensitive cell lines with CD79A/B mutations, leading to downregulation of IL-6/IL-10 and subsequent suppression of JAK/STAT3 signaling. In contrast, we observed little effect of R406 in cell lines with downstream activating mutations of the NFκB pathway (such as CARD11, A20 mutations). Therefore, the molecular nature of NF-κB pathway lesions may serve as a predictive marker for R406 responsiveness in ABC subtype DLBCL cell lines. Furthermore, in exploration of a common indicator of sensitivity to R406 in both ABC and GCB subtypes, we have confirmed that response to R406 is dependent on surface expression of a functional BCR and presence of a intact BCR signaling cascade (Chen et al. 2008 Blood 111: 2230). These cells are thus characterized as “B-cell Receptor active” or BCR subtype. Work is currently underway to further elucidate the characteristics of the BCR subtype, which may serve as a general selective marker for R406's efficacy in DLBCL cell lines. In conclusion, we have demonstrated that R406 functions as a BCR antagonist and reduces viability in DLBCL through inhibition of NFκB-mediated survival signals and downregulation of MCL-1. In addition, we have confirmed in ABC subtype DLBCL cell lines, similar to other BCR-targeted agents, R406 sensitivity is correlated with CD79A/B mutations whereas A20 or CARD11 mutations render cells resistance. Eventually, we proposed that BCR classification may serve as a broader selection maker for all DLBCL cell lines. Disclosures: Zhang: AstraZeneca: Employment, Equity Ownership. Parmentier:AstraZeneca: Employment, Equity Ownership. Lai:AstraZeneca: Employment, Equity Ownership. O'Connor:AstraZeneca: Employment, Equity Ownership. Passino:AstraZeneca: Employment, Equity Ownership. Powell:AstraZeneca: Employment, Equity Ownership. Devereaux:AstraZeneca: Employment, Equity Ownership. Byth:AstraZeneca: Employment, Possible shareholder Other.
26

Fernández, Leonor, and Peter Shorett. "Lessons in Equity From the Front Lines of COVID-19 Vaccination." JAMA Health Forum 2, no. 4 (April 7, 2021): e210612. http://dx.doi.org/10.1001/jamahealthforum.2021.0612.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Turner, PG. "Fault Lines In Equity Edited by Jamie Glister and Pauline Ridge." Oxford University Commonwealth Law Journal 12, no. 2 (March 27, 2012): 393–97. http://dx.doi.org/10.1080/14729342.2012.11421351.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Gandhi, Anita K., Herve Avet-Loiseau, Michelle Waldman, Anjan Thakurta, Sharon L. Aukerman, Gengxin Chen, Derek Mendy, et al. "Detection and Quantification of Cereblon Protein and mRNA in Multiple Myeloma Cell Lines and Primary CD138+multiple Myeloma Cells." Blood 120, no. 21 (November 16, 2012): 4043. http://dx.doi.org/10.1182/blood.v120.21.4043.4043.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Abstract 4043 Background: Cereblon (CRBN), a component of the DDB1-CUL4A-Roc1 ubiquitin ligase complex, has been identified as a target of the immunomodulatory agents thalidomide, lenalidomide, and pomalidomide (Lopez-Girona et al. Leukemia. 2012; Zhu et al. Blood. 2011; Ito et al. Science. 2010.). CRBN binding by these agents mediates their anti-proliferative effects in multiple myeloma (MM) cells (Lopez-Girona et al. Leukemia. 2012; Zhu et al. Blood. 2011). However, the role of CRBN quantification as a marker for disease responsiveness or resistance to these drugs remains to be fully defined. Furthermore, it is unclear whether measuring mRNA or protein expression is the best approach for development of a quantitative CRBN expression assay. In order to define the optimal assay approach, we have studied CRBN mRNA and protein expression in MM cell lines (n=20) and MM patient samples. Methods: CRBN isoform mapping was undertaken using a nested PCR approach and Sanger sequencing. Commercially available and newly generated rabbit anti-CRBN antibodies were characterized with recombinant human CRBN protein and MM cell line extracts via western blot analysis. Results: Our data show that in addition to the transcript for full length protein (GenBank Accession NM_016302.3), in MM cells there are at least 6 alternatively spliced isoforms of CRBN as depicted in Figure 1. Five of the 6 CRBN isoforms (CRBN-003, -004, -005, -006, and -007) contain novel splice junctions not previously described. In addition, 3 of the identified transcripts (CRBN-002, -003, and -005) contain in-frame ORFs, suggesting they encode variants of CRBN protein. Of note, exon 10, which contains a portion of the IMiD-binding domain, is not present in CRBN-002. The functional consequence of CRBN-002 remains to be elucidated, but may be a marker of drug resistance. In order to measure CRBN protein levels, we developed and characterized three rabbit monoclonal antibodies to CRBN including antibody CRBN65, which has the potential to discriminate between the different CRBN protein products, including CRBN-002 by western blot analysis. Additionally, we compared 8 commercially available CRBN antibodies. Western blot analysis of cell lines with commercial and newly developed antibodies identified full length protein at 51 kD. Most commercial antibodies also identified multiple bands of other sizes which may represent CRBN protein variants; however, many are likely non-specific bands as they are larger than full-length CRBN. Conclusion: We have identified novel splice variants of CRBN from MM cell lines and primary tumor samples. The structure of the isoforms and their potential ability to be translated into several protein variants of CRBN reflect the complex regulation of the CRBN gene. These data suggest that accurate quantification of CRBN mRNA level in clinical studies may require measurement of both full-length CRBN mRNA as well as other mRNA isoforms. Currently available primers and gene expression arrays are not capable of identifying and/or resolving the complex set of CRBN isoforms present in cells. These data also demonstrate that CRBN65 is a highly specific and sensitive antibody that could be used for detection of CRBN and its key variants. Taken together, our data emphasize the importance for developing standardized reagents and assays for both mRNA and protein level measurement of CRBN before using them as markers for clinical response or resistance. Disclosures: Gandhi: Celgene Corp: Employment, Equity Ownership. Waldman:Celgene Corp: Employment, Equity Ownership. Thakurta:Celgene Corp: Employment, Equity Ownership. Aukerman:Celgene Corp: Employment, Equity Ownership. Chen:Celgene Corp: Employment, Equity Ownership. Mendy:Celgene Corp.: Employment, Equity Ownership. Rychak:Celgene Corp: Employment, Equity Ownership. Miller:Celgene Corp: Employment, Equity Ownership. Gaidarova:Celgene Corp: Employment, Equity Ownership. Gonzales:Celgene Corp: Employment, Equity Ownership. Cathers:Celgene Corp: Employment, Equity Ownership. Schafer:Celgene: Employment, Equity Ownership. Daniel:Celgene Corporation: Employment. Lopez-Girona:Celgene Corp: Employment, Equity Ownership. Chopra:Celgene Corp: Employment, Equity Ownership.
29

Lopez-Girona, Antonia, Courtney G. Havens, Gang Lu, Emily Rychak, Derek Mendy, Bonny Gaffney, Christine Surka, et al. "CC-92480 Is a Novel Cereblon E3 Ligase Modulator with Enhanced Tumoricidal and Immunomodulatory Activity Against Sensitive and Resistant Multiple Myeloma Cells." Blood 134, Supplement_1 (November 13, 2019): 1812. http://dx.doi.org/10.1182/blood-2019-124338.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Lenalidomide- and pomalidomide-based therapies are effective drugs in the treatment of patients with multiple myeloma (MM), however most patients with MM eventually relapse or become resistant. CC-92480, a novel cereblon (CRBN) E3 ligase modulator (CELMoD) with multiple activities including potent immunomodulation and single-agent antiproliferative effects, is being investigated in a phase 1 clinical trial (CC-92480-MM-001; NCT03374085) for patients with relapsed/refractory MM (RRMM). The present study investigates the preclinical data and mechanism of action of CC-92480 in MM models. CELMoD agents bound to CRBN confer differentiated substrate-degradation specificity on the CRL4CRBN E3 ubiquitin ligase. CRBN-modulator agents mediate destruction of Ikaros and Aiolos, transcription factors that contribute to myeloma cell survival. CC-92480 was found to produce rapid, deep, and sustained degradation of Ikaros and Aiolos, with superior antimyeloma activity. Accordingly, in a CRBN protein competitive binding assay, CC-92480 displaced a Cy-5-labeled CELMoD analog from CRBN with a 50% inhibitory concentration (IC50) value of 0.03 μM, whereas lenalidomide competed with an IC50 value of 1.27 μM in the same assay, demonstrating a higher binding affinity of CC-92480 for CRBN. Additionally, CC-92480 promoted the recruitment of Ikaros to the CRBN E3 ligase complex more effectively than pomalidomide in 2 orthogonal CRBN/Ikaros binding assays; it also triggered a more extensive cellular ubiquitination of Ikaros, and a faster, more efficient depletion of cellular Ikaros and Aiolos than pomalidomide. In various MM cell lines, including those with acquired resistance to lenalidomide or pomalidomide and low levels of CRBN, CC-92480 produced robust degradation of Ikaros and Aiolos followed by strong reduction of 2 additional and highly critical transcription factors, c-Myc and interferon regulatory factor 4, which are linked to the induction of apoptosis as measured by cleaved caspase-3. The tumoricidal activity of CC-92480 was shown to be CRBN dependent, since the effect was prevented by complete loss of CRBN or by the stabilization of Ikaros and Aiolos. CC-92480 displayed broad and potent antiproliferative activity across a panel of 20 MM cell lines that are either sensitive, have acquired resistance, or are refractory to lenalidomide or pomalidomide; the cell lines also contained diverse chromosomal translocations and oncogenic drivers typically found in MM patients. Approximately half of the MM cell lines evaluated were highly sensitive to CC-92480, with IC50 values for antiproliferative activity ranging from 0.04 to 5 nM; only 2 cell lines had IC50 values > 100 nM. CC-92480 inhibits cell proliferation and induces apoptosis in MM cell lines that are not sensitive to lenalidomide or pomalidomide. This panel of cell lines includes both refractory cell lines and resistant cell lines generated through continuous exposure to lenalidomide and pomalidomide that acquired low levels of CRBN protein or mutations in the CRBN gene. CC-92480 also induced deep destruction of Ikaros and Aiolos in cultures of peripheral blood mononuclear cells (PBMCs), which led to the activation of T cells and increased production of the cytokines interleukin-2 and interferon gamma. These responses occurred at the range of CC-92480 concentrations that show potent tumoricidal effect against MM cells. The T cell activation and enhanced cytokine production by CC-92480 led to the potent and effective immune-mediated killing of MM cells in co-cultures with PBMCs. CC-92480 is a potent antiproliferative and proapoptotic novel CELMoD with enhanced autonomous cell-killing activity in MM cells that are either sensitive, resistant, or have acquired resistance to lenalidomide and pomalidomide. CC-92480 has a unique and rapid degradation profile stemming from the enhanced efficiency to drive the formation of a protein-protein interaction between Ikaros and Aiolos and CRBN, inducing cytotoxic effects in a CRL4CRBN-dependent fashion that leads ultimately to the induction of apoptosis, even in the context of low or mutated CRBN protein. Additionally, similar to lenalidomide, CC-92480 conserves immunomodulatory activity against MM cells. These data support the clinical investigation of CC-92480 in patients with RRMM. Disclosures Lopez-Girona: Celgene Corporation: Employment. Havens:Pfizer: Employment, Equity Ownership; Celgene: Equity Ownership. Lu:Celgene Corporation: Employment, Equity Ownership. Rychak:Celgene Corporation: Employment, Equity Ownership. Mendy:Celgene Corporation: Employment. Gaffney:Celgene: Employment. Surka:Celgene: Employment, Equity Ownership. Lu:Celgene Corporation: Employment, Equity Ownership. Matyskiela:Celgene corporation: Employment. Khambatta:Celgene: Employment. Wong:Celgene Corporation: Employment, Equity Ownership. Hansen:Celgene Corporation: Employment. Pierce:Celgene Corporation: Employment, Equity Ownership. Cathers:Global Blood Therapeutics (GBT): Employment; Celgene Corporation: Equity Ownership. Carmichael:Celgene plc: Employment, Equity Ownership.
30

Lyons, Torrey, and Dong-ah Choi. "Transit Economic Equity Index: Developing a Comprehensive Measure of Transit Service Equity." Transportation Research Record: Journal of the Transportation Research Board 2675, no. 3 (January 7, 2021): 288–300. http://dx.doi.org/10.1177/0361198120970529.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
In this study an index is developed called the Transit Economic Equity Index, to enable quantitative assessment of transit service equity. The index measures convenience of travel for work trips for advantaged and disadvantaged populations, based on travel speed, using a multimodal network that includes transit lines, stop locations, transit schedules, and pedestrian connections via the street network. Non-peak hour service is compared with peak hour service to determine the degree to which operating resources are concentrated in times that might have greater benefits to advantaged populations. Finally, accessibility to the transit system is compared in relation to the number of transit stops in neighborhoods and employment centers, and these figures are compared between advantaged and disadvantaged locations. The scores for these three components are combined to create a single measure of transit economic equity. Disadvantage is defined using criteria established in Title VI of the Civil Rights Act of 1964. The index is constructed in a way that balances a robust and meaningful measure of transit equity that is decipherable by practitioners so that they can assess the equity of their systems as well as how potential service changes affect equity.
31

Rivera, Victor M., Justin R. Pritchard, Francois Gonzalvez, Theresa Baker, Joseph M. Gozgit, and Graeme Hodgson. "Comparative TKI Profiling Analyses to Explore Potential Mechanisms of Ponatinib-Associated Arterial Thrombotic Events." Blood 124, no. 21 (December 6, 2014): 1783. http://dx.doi.org/10.1182/blood.v124.21.1783.1783.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Background: Ponatinib is a potent pan-BCR-ABL tyrosine kinase inhibitor (TKI) indicated for patients with T315I positive or treatment-refractory CML and Ph+ ALL. To develop hypotheses regarding molecular and cellular targets of ponatinib that could contribute to arterial thrombotic events observed in some patients we are undertaking a broad comparative profiling analysis of ponatinib and other TKIs. Methods: Cellular activities of ponatinib and additional BCR-ABL (imatinib, nilotinib, dasatinib, bosutinib) and VEGFR2/multi-targeted (sunitinib, regorafenib) TKIs were examined in panels of Ba/F3 cell lines expressing activated kinase variants (N=61), tumor cell lines (N=246), and primary lines derived from human vasculature (aortic smooth muscle cells [ASMC] and umbilical vein [HUVEC], aortic [HAEC] and pulmonary artery [HPAEC] endothelial cells). Human steady-state Cave concentrations of 45 mg ponatinib (101 nM) were corrected for the functional effects of protein binding (3.6-fold) to derive the clinically-effective concentration. Results: Ponatinib inhibits the in vitro activity of multiple kinases with IC50s within 10-fold of ABL, including members of the VEGFR, PDGFR, FGFR, EPH receptor and SRC families of kinases, KIT, RET, TIE2, and FLT3. This profile was largely recapitulated in cellular assays using engineered Ba/F3 cells, with ponatinib demonstrating substantially greater potency against VEGFRs, FGFRs, TIE2, RET and FLT3 than other ABL TKIs. Across a broad panel of tumor cell lines, ponatinib inhibited viability with a median IC50 of 598 nM. Ponatinib only inhibited 16 cell lines (6.5%) with IC50s below its clinically effective concentration (28 nM) with the 5 most sensitive lines (IC50 <1 nM) all being BCR-ABL positive. Within the vasculature-derived cell panel, ponatinib inhibited viability of HUVECs grown in full serum with an IC50 of 261 nM, with all of the other ABL and non-ABL TKIs tested having IC50s >2000 nM. Effects of ponatinib on HAECs, HPAECs and ASMCs were more modest (IC50s 1533, 490 and 750 nM, respectively). Finally, ponatinib (IC50 20 nM) and other VEGFR2 inhibitors potently inhibited survival of HUVECs grown in VEGF-dependent conditions, while other BCR-ABL inhibitors, except dasatinib (IC50 14 nM), did not. Conclusions: Ponatinib is a potent BCR-ABL inhibitor that also inhibits VEGFR2 and other kinases at clinically achievable concentrations in vitro. Modest effects of ponatinib on endothelial cells have been observed that warrant further exploration in vivo. Developing a precise understanding of the mechanism by which ponatinib contributes to arterial thrombotic events should facilitate development of strategies to optimize its benefit/risk in patients. Disclosures Rivera: ARIAD Pharmaceuticals Inc: Employment, Equity Ownership. Pritchard:ARIAD Pharmaceuticals, Inc.: Employment, Equity Ownership. Gonzalvez:ARIAD Pharmaceuticals, Inc.: Employment, Equity Ownership. Baker:ARIAD Pharmaceuticals, Inc.: Employment, Equity Ownership. Gozgit:ARIAD Pharmaceuticals, Inc.: Employment, Equity Ownership. Hodgson:ARIAD Pharmaceuticals, Inc.: Employment, Equity Ownership.
32

Li, Lingna, Wenyong Tong, Megan Lau, Katherine Fells, Tong Zhu, Yingqing Sun, Ernest Kovacs, et al. "Preclinical Development of an Anti-CD38 Antibody-Drug Conjugate for Treatment of Hematological Malignancies." Blood 134, Supplement_1 (November 13, 2019): 5621. http://dx.doi.org/10.1182/blood-2019-132062.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
CD38 is a validated target for the treatment of multiple myeloma (MM). Daratumumab (Darzalex®), an anti-CD38 monoclonal antibody (mAb), has shown great clinical efficacy and has been approved for multiple myeloma treatment. However, both primary refractoriness and development of resistance to daratumumab therapy have been reported. Based on the therapeutic benefits of this CD38 antibody, we developed a CD38-targeting antibody-drug conjugate (ADC), employing a fully human anti-CD38 antibody STI-6129, identified from Sorrento's G-MAB® antibody library, and proprietary linker-toxin technology. The toxin payload is duostatin 5.2 (Duo.5.2), a microtubule inhibitor, conjugated to STI-6129 via a non-polyethylene glycol linker resulting in our lead ADC CD38-077. Cell binding studies showed that it specifically binds to CD38-positive tumor cells but not CD38-negative cell lines. The cell binding was proportional to the CD38 expression level on the cell surface. The ADC was internalized into CD38-positive cells at a rate comparable to that of the unconjugated antibody, indicating that conjugation did not change the binding characteristics of STI-6129 to its antigen. In cytotoxicity studies, CD38-077 exhibited a CD38-dependent cytotoxic activity against a panel of CD38-expressing tumor cell lines and was more potent in cells with high CD38 expression. The cytotoxic effect of CD38-077 was also examined against human PBMC cells, as it has been reported that certain types of the immune cells express CD38. The result indicated that normal PBMC cells were generally insensitive to the ADC up to 1 µM following 120 hr exposure. We investigated the anti-tumor activity of CD38-077 in xenograft animal models of Burkitt's lymphoma and two different multiple myeloma (MM) cell lines. The studies evaluated different dose levels and dosing regimens, including single dose and multiple doses at various intervals. The data showed that the ADC has a broad, potent and CD38-dependent in vivo efficacy in all three xenograft tumor models examined. In a pharmacokinetic study in naïve mice, CD38-077 was found to be stable, with T1/2 of about 7-11 days, comparable to that of the unconjugated STI-6129 antibody. In summary, CD38-077 exhibits strong anti-tumor activity in vitro and in vivo. The ADC showed specific activity towards CD38-expressing tumors but was less active against CD38-expressing normal PBMC cells, which express relatively low levels of CD38 level and where internalization was not detectable. These results warrant further development exploration of CD38-077. Disclosures Li: Concortis Biotherapeutics: Employment, Equity Ownership. Lau:Levena Biopharma: Employment, Equity Ownership. Fells:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Zhu:Levena Biopharma: Employment, Equity Ownership, Patents & Royalties. Sun:Levena Biopharma: Employment, Equity Ownership. Kovacs:Levena Biopharma: Employment, Equity Ownership. Khasanov:Levena Biopharma: Employment, Equity Ownership. Yan:Levena Biopharma: Employment, Equity Ownership. Deng:Levena Biopharma: Employment, Equity Ownership. Takeshita:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Kaufmann:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Ji:Sorrento Therapeutics Inc: Employment, Equity Ownership, Patents & Royalties; Celularity, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Li:Levena Biopharma: Employment, Equity Ownership, Patents & Royalties; Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Zhang:Concortis Biotherapeutics: Employment, Equity Ownership, Patents & Royalties.
33

Wang, Beatrice T., Thomas J. Matthew, Ling Wang, Tasnim Kothambawala, Susan E. Calhoun, Eric W. Humke, Angus M. Sinclair, and Bruce A. Keyt. "The Anti-Tumor Activity of Igm-8444, an Agonistic Death Receptor 5 (DR5) IgM Antibody, Is Sensitized in Combination with Chemotherapy and Bcl-2 Inhibitors in NHL and AML." Blood 136, Supplement 1 (November 5, 2020): 43–44. http://dx.doi.org/10.1182/blood-2020-141449.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Background: Death receptor 5 (DR5) is a member of the tumor necrosis factor (TNF) receptor superfamily that multimerizes when bound to its ligand, TNF-related apoptosis inducing ligand (TRAIL), to activate the extrinsic apoptotic pathway. DR5 is broadly expressed on solid and hematologic cancers and has been targeted with both recombinant TRAIL and agonistic antibodies in the clinic. However, these therapeutics have been unsuccessful due to lack of efficacy or hepatotoxicity. We have developed IGM-8444, a pentameric IgM with 10 binding sites specific for DR5, that multimerizes DR5 to selectively and potently induce tumor cell apoptosis while maintaining tolerability. We have previously presented the in vitro and in vivo efficacy of IGM-8444 in solid tumor models, demonstrating low picomolar potency across multiple tumor cell lines, strong tumor regressions in cell line and patient derived xenograft mouse tumor models, and dose-dependent increases in apoptotic biomarkers. Here, we evaluate the activity of IGM-8444 in hematologic malignancies in combination with chemotherapy or targeted agents including Bcl-2 inhibitors targeting the intrinsic apoptotic pathway. Methods: Human hematologic cancer cell lines and primary human hepatocytes were evaluated in vitro for dose-dependent IGM-8444-induced cytotoxicity. Cell lines were further evaluated using IGM-8444 in combination with chemotherapy or targeted agents including Bcl-2 inhibitor ABT-199. In vivo efficacy was evaluated using IGM-8444 in combination with ABT-199 in cell line-derived xenograft mouse tumor models. Results: In a previous cancer cell line screen profiling single agent IGM-8444 cytotoxicity across 190 solid and hematologic cell lines, 25 (13%) were classified as highly responsive and 75 (39%) as moderately responsive to IGM-8444 induced cell death. Here the in vitro activity of IGM-8444 was evaluated across a subset of 32 NHL and AML cell lines. 5/21 (24%) of NHL cell lines and 5/11 (45%) of AML cell lines tested were classified as highly responsive or moderately responsive to IGM-8444-induced cytotoxicity. The DOHH-2 and JEKO1 NHL cell lines were amongst the most sensitive, with growth-normalized EC50 values as low as 0.03 ng/mL (0.03 pM) for JEKO1. Combinations with chemotherapy including cytarabine and doxorubicin or targeted agents such as Bcl-2 inhibitor ABT-199 resulted in synergistic in vitro cytotoxicity in multiple cell lines, as determined by Bliss synergy scores. IGM-8444 demonstrated minimal to no in vitro cytotoxicity to primary human hepatocytes at doses several log-fold higher than efficacious doses, and this favorable in vitro safety profile was maintained in combination with chemotherapeutic agents and ABT-199. Combination of IGM-8444 with ABT-199 also resulted in synergistic in vivo efficacy. In a DOHH-2 NHL model, IGM-8444 and ABT-199 showed modest tumor growth inhibition as single agents. However the combined treatment regimen led to tumor regressions during the first 2 weeks of treatment, with 3 of 10 animals showing a partial response and 2 of 10 animals achieving a complete response. The combined treatment also extended median overall survival compared to the control group, which was a significant improvement compared to either agent alone. Collectively, these results provide a strong rationale for simultaneously targeting the extrinsic and intrinsic apoptotic pathways to achieve enhanced efficacy. Conclusions: These data support the clinical development of IGM-8444 in hematological malignancies as a single agent, in combination with standard of care chemotherapy, and in combination with targeted agents that impact the intrinsic signaling pathway such as Bcl-2 inhibitor ABT-199. Initiation of a Phase I clinical study evaluating the safety of IGM-8444 is anticipated in 2020. Disclosures Wang: IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Matthew:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Wang:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Kothambawala:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Calhoun:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Humke:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Sinclair:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company. Keyt:IGM Biosciences Inc: Current Employment, Current equity holder in publicly-traded company.
34

Bates, Jamie, Saritha Kusam, Stacey Tannheimer, Astrid Clarke, Thomas Kenney, David Breckenridge, and Daniel Tumas. "Combination of the BET Inhibitor GS-5829 and a BCL2 Inhibitor Resulted in Broader Activity in DLBCL and MCL Cell Lines." Blood 128, no. 22 (December 2, 2016): 5104. http://dx.doi.org/10.1182/blood.v128.22.5104.5104.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Introduction: GS-5829 is a potent and selective inhibitor of the bromodomain and extra-terminal (BET) family of proteins. BET proteins regulate the transcription of key oncogenes including MYC, resulting in cancer cell growth (Yang Z. et. al. Mol Cell Biol 2008; LeRoy G. et. al. Mol Cell 2008). Diffuse large B-cell lymphoma (DLBCL) is a diverse and genetically heterogeneous subtype of non-Hodgkin's lymphoma (NHL) (Lenz G. et. al. N Engl J Med 2010). Lymphomas harboring translocation of both MYC and BCL2, the so called "double hit" lymphoma, have a poor prognosis (Li S. et. al. Mod Pathol 2012). High protein expression of MYC and BCL2 together, in the absence of translocation, was also shown to be a poor prognostic indicator for DLBCL patients treated with R-CHOP which consists of rituximab, Cyclophosphamide, Hydroxydaunorubicin, Oncovin (vincristine), and Prednisone (Green T. et. al. J Clin Oncol 2012). Though double hit lymphoma occurs in 5% of DLBCL patients, 29% of DLBCL patients express high levels of both proteins-even in the absence of translocation. ABT-199 is a potent and selective BCL2 inhibitor and sensitivity to ABT-199 in DLBCL cell lines correlates with high basal BCL2 protein expression (Souers A. et. al. Nat Med 2013). We evaluated the combination of GS-5829 and ABT-199 in DLBCL and MCL cell lines in vitro. Methods: The activity of GS-5829 and ABT-199 alone or in combination in DLBCL and Mantle Cell Lymphoma (MCL) cell lines was assessed by evaluating cell growth inhibition and apoptosis. Growth inhibition was evaluated by CellTiterGlo or ATPLite. Apoptosis was assessed by Annexin V positivity by flow cytometry. The protein levels of MYC, BCL2, BIM, BCL-XL, and MCL1 were quantified by Western blotting and normalized to ACTIN levels. Analyses were performed to determine whether the expression of any of the profiled proteins correlated with the activity of GS-5829 or ABT-199. Results: DLBCL cell lines were broadly sensitive to cell growth inhibition by GS-5829, with EC50s ranging from 17 to 330 nM. Growth inhibition correlated with the reduction in MYC protein levels upon treatment with GS-5829. Treatment with GS-5829 induced apoptosis in a subset of DLBCL cell lines (58%). Cell lines with low level expression of BCL2 were more sensitive to GS-5829-induced apoptosis (p < 0.05 by T-Test). Expression levels of the other profiled proteins did not correlate with GS-5829 activity. DLBCL cell lines varied in sensitivity to ABT-199, with EC50s ranging from 1.5 to 7200 nM. Cell lines that were sensitive to ABT-199 in general had high levels of BCL2 protein. The single-agent activity of GS-5829 and ABT-199 in DLBCL cell lines had an apparent reciprocal relationship (Figure 1). The combination of GS-5829 and ABT-199 at clinically relevant concentrations resulted in broader activity and greater suppression of cell growth than either agent alone in a panel of DLBCL and MCL cell lines (Figure 2). Conclusion: The data demonstrated a reciprocal sensitivity of DLBCL cell lines to the BET inhibitor GS-5829 and the BCL2 inhibitor ABT-199. The combination of these two agents resulted in superior growth inhibition in DLBCL and MCL cell lines. These data suggest that GS-5829 and ABT-199 in combination may lead to broader, superior responses in DLBCL and MCL. Figure 1 Growth Inhibition by ABT-199 and Induction of Apoptosis by GS-5829 are Significantly Different in the High and Low Basal BCL2 Protein Expression Groups. Figure 1. Growth Inhibition by ABT-199 and Induction of Apoptosis by GS-5829 are Significantly Different in the High and Low Basal BCL2 Protein Expression Groups. Figure 2 Combination of GS-5829 and ABT-199 Resulted in More Potent Inhibition of Cell Growth in DLBCL Cell Lines at Clinically Relevant Concentrations (42 - 120 nM GS-5829 in Blue and 330 nM ABT-199). Figure 2. Combination of GS-5829 and ABT-199 Resulted in More Potent Inhibition of Cell Growth in DLBCL Cell Lines at Clinically Relevant Concentrations (42 - 120 nM GS-5829 in Blue and 330 nM ABT-199). Disclosures Bates: Gilead Sciences: Employment, Equity Ownership. Kusam:Gilead Sciences: Employment, Equity Ownership. Tannheimer:Gilead Sciences: Employment. Clarke:Gilead Sciences, Inc.: Employment, Equity Ownership. Kenney:Gilead Sciences, Inc.: Employment, Equity Ownership. Breckenridge:Gilead Sciences: Employment, Equity Ownership. Tumas:Gilead Sciences: Employment, Equity Ownership.
35

Cojocari, Dan, Sha Jin, Julie J. Purkal, Relja Popovic, Nari N. Talaty, Yu Xiao, Larry R. Solomon, Erwin Boghaert, Joel D. Leverson, and Darren C. Phillips. "5-Azacytidine Induces NOXA and PUMA Expression to Prime AML Cells for Venetoclax-Mediated Apoptosis." Blood 132, Supplement 1 (November 29, 2018): 2644. http://dx.doi.org/10.1182/blood-2018-99-111985.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Acute myeloid leukemia (AML) is a clonal hematologic malignancy characterized by genomic heterogeneity and epigenetic changes, including aberrant DNA hypermethylation. Phase-Ib clinical data in relapsed/refractory AML patients indicate that combining venetoclax with the hypomethylating agents (HMAs) 5-azacitidine (5-Aza) or decitabine results in an overall response (OR) of 62% (DiNardo et al. 2018) compared to the historical OR of 28-29% with HMAs treatment alone (Kantarjian et al. 2013; Dombret et al. 2015). Subsequently, a randomized phase-III clinical trial was initiated to evaluate venetoclax activity in combination with 5-Aza in treatment-naïve AML patients ineligible for standard induction therapy (M15-656, NCT02993523). However, the underlying mechanism for the combinational activity observed between venetoclax and 5-Aza is unknown. In this study, we demonstrate that both chronic low-dose 5-Aza treatment, which induced global DNA demethylation, and acute treatment (24 hours, non-epigenetic effects) can drive combinational activity with venetoclax in AML through distinct mechanisms. Chronic culture with a low-dose 5-Aza for one week sensitized AML cell lines to venetoclax in vitro. In contrast, acute treatment with 5-Aza, activated the integrated stress response (ISR) pathway to induce expression of the BH3-only proteins NOXA (PMAIP1) and PUMA (BBC3) in human AML cell lines, independent of DNA methylation. This resulted in an increase in the amount of NOXA and/or PUMA in complex with anti-apoptotic proteins like BCL-2, BCL-XL and MCL-1, thereby "priming" AML cells for induction of apoptosis by venetoclax treatment. Priming for apoptosis resulted in significant synergistic cell death in a panel of AML cell lines treated with venetoclax and 5-Aza in vitro. In this panel of cell lines, the level of the PMAIP1, BBC3, and DDIT3 gene induction correlated with the synergy observed between venetoclax and 5-Aza. Importantly, subsequent PMAIP1 deletion significantly impacted the kinetics and depth of apoptosis induced by 5-Aza or venetoclax alone or in combination. In accordance with the in vitro combinational activity, the venetoclax/5-Aza combination provided added benefit over either agent alone in two xenograft models of AML. Together, these data provide a rationale for an ongoing randomized phase-III clinical trial evaluating venetoclax activity in combination with 5-Aza (M15-656, NCT02993523). Disclosures: DC, SJ, JP, RP, NT, YX, EB, JL, and DP are employees of AbbVie. LS is a former employee of AbbVie and was employed during the duration of this study. The design, study conduct, and financial support for this research were provided by AbbVie and Genentech. AbbVie participated in the interpretation of data, review, and approval of the publication. Disclosures Cojocari: AbbVie Inc: Employment. Jin:AbbVie Inc: Employment, Equity Ownership. Purkal:AbbVie Inc: Employment, Equity Ownership. Popovic:AbbVie Inc: Employment, Equity Ownership. Talaty:AbbVie Inc: Employment, Equity Ownership. Xiao:AbbVie Inc: Employment, Equity Ownership. Solomon:AbbVie Inc: Equity Ownership. Boghaert:AbbVie Inc: Employment, Equity Ownership. Leverson:AbbVie Inc: Employment, Equity Ownership, Patents & Royalties. Phillips:AbbVie Inc: Employment, Equity Ownership, Patents & Royalties.
36

Pinho, Pedro, Helen Kylefjord, Vilma Rraklli, Christina Rydergård, Biljana Rizoska, Anders Eneroth, Johan Bylund, et al. "Protides of 5-Fluorotroxacitabine Display Potent Anti-Proliferative Properties and Circumvent Deoxycytidine Kinase-Mediated Resistance Associated with Cytotoxic Cytidine Analogues; A Novel Approach for Acute Myeloid Leukemia." Blood 132, Supplement 1 (November 29, 2018): 3497. http://dx.doi.org/10.1182/blood-2018-99-114440.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract The cytotoxic nucleoside cytarabine forms the backbone of AML induction and consolidation therapies, but is associated with severe toxicities that preclude its use in patients unable to tolerate aggressive chemotherapy. Options for patients that do not respond to cytarabine, or relapse post-treatment, are limited. Elderly patients and those with relapsed/refractory AML would particularly benefit from the availability of new agents to develop treatment regimens that provide increased efficacy and tolerability compared to cytarabine, and that have a decreased susceptibility to mechanisms of cytarabine resistance, such as decreased deoxycytidine kinase (dCK) and/or upregulation of cytidine deaminase (CDA). Our preclinical evaluation of potential new anti-proliferative chemotherapeutics identified 5-fluorotroxacitabine (5FTRX), a chain-terminating cytidine-based L-nucleoside, as having promising anti-proliferative activity against AML cell lines, and resistance to degradation by CDA. To understand potential mechanisms of resistance to 5FTRX, we selected a population of THP1 (THP1-R) cells resistant to 5FTRX. THP1-R cells were 66-fold resistant to 5FTRX and cross resistant to cytarabine (35-fold) with CC50 values for both nucleosides >50 μM. We discovered that THP1-R cells had decreased levels of dCK (>95% by Western blot), the kinase responsible for the phosphorylation of cytidine and cytidine analogues such as troxacitabine and cytarabine to their corresponding monophosphates. Confirming the importance of dCK in the activation of 5FTRX and cytarabine, chemical inhibition of dCK also rendered THP1 cells >90-fold resistant to 5FTRX and cytarabine. To develop molecules that overcome resistance to both high CDA and low dCK, we used protide technology to construct nucleotide monophosphate prodrugs of 5FTRX, including one potent example, MV806. MV806 was not dependent upon dCK as it maintained similar efficacy in THP1-R cells with low dCK and against THP1 cells treated with the selective dCK inhibitor. We tested MV806 and 5FTRX in a panel of AML cell lines (n=7). MV806 was more potent than 5FTRX with CC50 values ranging from 0.0020-0.19 μM, compared to 0.057-1.2 μM for 5FTRX. MV806 also demonstrated CC50s <0.1 μM against selected T- and B-cell lymphoma cell lines (e.g. MOLT4 and RAJI). Increased in vitro potency of this prodrug compared to 5FTRX correlated with elevated intracellular triphosphate levels in AML cells; MV806 generated 5-fold more triphosphate than 5FTRX in MV4-11 cells. We also tested MV806 in combination with doxorubicin or azacytidine in two AML cell lines (MV4-11 and THP-1). In both tested cell lines, strong synergy was observed (Bliss independence analysis synergy volumes >100), demonstrating future opportunities for clinical combinations. Finally, we showed that MV806 had DMPK profiles suitable for preclinical and clinical development. Leading protides were highly soluble, had a predicted half-life of >6h in human blood and demonstrated IC50 values >1 μM against major CYP isoforms (2A6, 2C9, 2D6, 3A4) with no evidence of time-dependent inhibition at 1 μM. To conclude, we used protide technology to directly deliver the active monophosphate species of 5FTRX intracellularly and thereby overcome resistance to cytarabine due to down-regulation of dCK and increased CDA expression. Taken together, our findings support the further development of protides of 5FTRX for the treatment of AML, including AML patients with reduced sensitivity to cytarabine through high CDA expression and/or low dCK expression. Disclosures Pinho: Medivir AB: Employment, Equity Ownership. Kylefjord:Medivir AB: Employment, Equity Ownership. Rraklli:Medivir AB: Employment. Rydergård:Medivir AB: Employment, Equity Ownership. Rizoska:Medivir AB: Employment, Equity Ownership. Eneroth:Medivir AB: Employment, Equity Ownership. Bylund:Medivir AB: Employment, Equity Ownership. Moses:Medivir AB: Employment, Equity Ownership. Norin:Medivir AB: Employment, Equity Ownership. Bethell:Medivir AB: Employment, Equity Ownership. Schimmer:Otsuka Pharmaceuticals: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Consultancy; Medivir AB: Research Funding. Albertella:Medivir AB: Employment, Equity Ownership. Targett-Adams:Medivir AB: Employment, Equity Ownership.
37

Klatt, Martin G., Zhiyuan Yang, Jianying Liu, Tatyana Korontsvit, Sung Soo Mun, Leila Peraro, Thomas J. Gardner, et al. "A TCR Mimic Antibody-Directed CAR T Cell Specific for Intracellular NDC80 Is Broadly Cancer Reactive and Displays High Activity Against Hematological Malignancies." Blood 136, Supplement 1 (November 5, 2020): 20–21. http://dx.doi.org/10.1182/blood-2020-140179.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Chimeric antigen receptor (CAR) T cells represent a novel class of FDA-approved drugs with high efficacy against refractory B cell derived malignancies and potentially other cancer types. However, target selection for CAR T cell therapy remains challenging as cell surface proteins are not cancer-specific and therefore often not adaptable for CAR T cell therapy. In contrast, many intracellular proteins can be highly tumor specific and are targetable after proteasomal degradation and presentation on human leukocyte antigen (HLA) complexes recognized by T cell receptor mimic antibodies. This class of antibodies recognizes peptide:HLA complexes with a similar mode of recognition as a TCR, but with the clinical versatility and applicability of an antibody. To identify a tumor specific target that is presented as a peptide in conjunction with the highly prevalent HLA allele A*02:01, we immunopurified peptide:HLA complexes from various cancer cell lines of different origins, separated HLA ligands from complexes and identified their peptide sequences via mass spectrometry. Network analysis of the resulting HLA ligand datasets identified shared biological processes among the tumor cell lines that were not present in network analyses of published datasets of healthy human tissue HLA ligandomes. Through this filtering process several potential targets were identified and an HLA ligand derived from kinetochore NDC80 protein homolog (NDC80) was selected as a target. The NDC80 derived peptide was detected in over 90% of the A*02 positive cell lines tested and never reported to be present in HLA ligand datasets of healthy human tissues. Furthermore, NDC80 has been shown to be differentially expressed in malignant compared to adjacent non-malignant tissues and is associated with poor prognosis in many cancer types. After utilizing E-ALPHA®phage library screening, one clone (NDC80-L1) was selected as the lead TCR mimic antibody. Overall, NDC80-L1 showed high specificity for the target HLA:peptide complex in both antibody and CAR T cell format in vitro and demonstrated binding primarily to the central region of the HLA ligand as determined by alanine screening assays. The exquisite specificity of NDC80-L1 was further illustrated by NDC80 knockdown experiments as well as successful immunopurification of the target peptide together with no relevant off-targets from BV173 ALL cells in mass spectrometry assays. Given the high specificity, sensitivity was assessed primarily in a potent CAR T cell format: Multiple tumor cell lines of different origin (e.g. ALL, AML, lymphoma, melanoma, mesothelioma, pancreatic and thyroid cancer) were successfully killed in vitro by NDC80-L1 CAR T cells, but no toxicity towards A*02:01 positive CAR T cells, healthy PBMCs or NDC80 target negative cell lines was observed. Interestingly, NDC80-L1 CAR T cells demonstrated highest efficacy in hematological malignancies most likely correlating with elevated expression of antigen presentation machinery and rapid cell division which leads to strong surface expression of NDC80 peptides. In summary, CAR T cells directed against peptide/HLA-A*02 derived from the NDC80 protein effectively kill multiple cancer cell lines in vitro without evidence of relevant off-target killing. However, the improved killing especially against ALL, AML and lymphomas highlights the potential of these CAR T cells to preferentially eliminate cancer cells with high proliferative capacity. Future in vivo studies with CAR T cell and antibody format will further investigate this TCR mimic antibody's potential as a tumor-agnostic therapeutic agent. Disclosures Klatt: MSKCC/EUREKA: Patents & Royalties: MSKCC AND EUREKA THERAPUETICS HAVE FILED A PATENT FOR THIS ANTIBODY/SCFV. Yang:Eureka Therapuetics: Current Employment, Current equity holder in private company, Patents & Royalties: MSKCC and Eureka have filed patent for this TCRm and ScvF. Liu:Eureka Therapue: Current Employment, Current equity holder in private company, Patents & Royalties: Eureka Therapuetics and MSKCC have filed patent on this ScFV and TCRm. Dao:Eureka Therapeutics: Consultancy. Liu:Eureka Therapeutics: Current Employment, Current equity holder in private company, Patents & Royalties: Eureka Therapuetics and MSKCC have filed patent on this ScFV and TCRm. Scheinberg:Eureka Therapeutics: Consultancy, Current equity holder in private company, Patents & Royalties: Eureka Therapuetics and MSKCC have filed patent on this ScFV and TCRm; Actinium: Consultancy, Current equity holder in private company; Sellas: Consultancy, Current equity holder in private company; Contrafect: Current equity holder in private company; Arvenas: Current equity holder in private company; Sapience: Consultancy, Current equity holder in private company; Iovance: Current equity holder in private company; Oncopep: Consultancy; Pfizer: Consultancy, Current equity holder in private company; Lantheus: Current equity holder in private company; Enscyse: Current equity holder in private company.
38

Totman, Jennifer, Dorothy Brach, Vinny Motwani, Selene Howe, Emily Deutschman, John Lampe, Thomas V. Riera, et al. "Pharmacologic Inhibition of the Histone Methyltransferase SETD2 with EZM0414 As a Novel Therapeutic Strategy in Relapsed or Refractory Multiple Myeloma and Diffuse Large B-Cell Lymphoma." Blood 138, Supplement 1 (November 5, 2021): 1142. http://dx.doi.org/10.1182/blood-2021-151147.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Introduction: SETD2 is the only known histone methyltransferase (HMT) capable of catalyzing H3K36 trimethylation (H3K36me3) in vivo. It plays an important role in several biological processes including B cell development and maturation, leading to the hypothesis that SETD2 inhibition in these settings could provide anti-tumor effects. The normal process of B cell development/maturation renders B cells susceptible to genetic vulnerabilities that can result in a dysregulated epigenome and tumorigenesis, including in multiple myeloma (MM) and diffuse large B-cell lymphoma (DLBCL). For example, 15%-20% of MM harbors the high risk (4;14) chromosomal translocation, resulting in high expression of the multiple myeloma SET domain (MMSET) gene. MMSET is an HMT that catalyzes H3K36me1 and H3K36me2 formation and extensive scientific work has established overexpressed MMSET as a key factor in t(4;14) myeloma pathogenesis. To the best of our knowledge MMSET has eluded drug discovery efforts, however, since t(4;14) results in high levels of the H3K36me2 substrate for SETD2, inhibiting SETD2 offers promise for targeting the underlying oncogenic mechanism driven by MMSET overexpression in t(4;14) MM patients. In addition, SETD2 loss of function mutations described to date in leukemia and DLBCL are always heterozygous, suggesting a haploinsufficient tumor suppressor role for SETD2. This observation points to a key role for SETD2 in leukemia and lymphoma biology and suggests that therapeutic potential of SETD2 inhibition may also exist in these or similar settings. EZM0414 is a first-in-class, potent, selective, orally bioavailable small molecule inhibitor of the enzymatic activity of SETD2. We explored the anti-tumor effects of SETD2 inhibition with EZM0414 in MM and DLBCL preclinical studies to validate its potential as a therapy in these tumor types. Methods: Cellular proliferation assays determined IC 50 values of EZM0414 in MM and DLBCL cell line panels. Cell line-derived xenograft preclinical models of MM and DLBCL were evaluated for tumor growth inhibition (TGI) in response to EZM0414. H3K36me3 levels were determined by western blot analysis to evaluate target engagement. Combinatorial potential of SETD2 inhibition with MM and DLBCL standard of care (SOC) agents was evaluated in 7-day cotreatment in vitro cellular assays. Results: Inhibition of SETD2 by EZM0414 results in potent anti-proliferative effects in a panel of MM and DLBCL cell lines. EZM0414 inhibited proliferation in both t(4;14) and non-t(4;14) MM cell lines, with higher anti-proliferative activity generally observed in the t(4;14) subset of MM cell lines. The median IC 50value for EZM0414 in t(4;14) cell lines was 0.24 μM as compared to 1.2 μM for non-t(4;14) MM cell lines. Additionally, inhibitory growth effects on DLBCL cell lines demonstrated a wide range of sensitivity with IC 50 values from 0.023 μM to &gt;10 μM. EZM0414 resulted in statistically significant potent antitumor activity compared to the vehicle control in three MM and four DLBCL cell line-derived xenograft models. In the t(4;14) MM cell line-derived xenograft model, KMS-11, robust tumor growth regressions were observed at the top two doses with maximal TGI of 95%. In addition, two non-t(4;14) MM (RPMI-8226, MM.1S) and two DLBCL xenograft models (TMD8, KARPAS422) demonstrated &gt; 75% TGI; with two additional DLBCL models (WSU-DLCL2, SU-DHL-10) exhibiting &gt; 50% TGI in response to EZM0414. In all models tested, the antitumor effects observed correlated with reductions in intratumoral H3K36me3 levels demonstrating on-target inhibition of SETD2 methyltransferase activity in vivo. In vitro synergistic antiproliferative activity was also observed when EZM0414 was combined with certain SOC agents for MM and DLBCL. Conclusions: Targeting SETD2 with a small molecule inhibitor results in significantly reduced growth of t(4;14) MM, as well as non-t(4;14) MM and DLBCL cell lines, in both in vitro and in vivo preclinical studies. In addition, in vitro synergy was observed with EZM0414 and certain SOC agents commonly used in MM and DLBCL, supporting the combination of SETD2 inhibition with current MM and DLBCL therapies. This work provides the rationale for targeting SETD2 in B cell malignancies such as MM, especially t(4;14) MM, as well as DLBCL, and forms the basis for conducting Phase 1/1b clinical studies to evaluate the safety and activity of EZM0414 in patients with R/R MM and DLBCL. Disclosures Totman: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Brach: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Motwani: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Howe: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Deutschman: Epizyme, Inc.: Divested equity in a private or publicly-traded company in the past 24 months, Ended employment in the past 24 months. Lampe: Epizyme, Inc.: Divested equity in a private or publicly-traded company in the past 24 months, Ended employment in the past 24 months. Riera: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Tang: Epizyme, Inc.: Divested equity in a private or publicly-traded company in the past 24 months, Ended employment in the past 24 months. Eckley: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Alford: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Duncan: Epizyme, Inc.: Divested equity in a private or publicly-traded company in the past 24 months, Ended employment in the past 24 months. Farrow: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Dransfield: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Raimondi: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Thomeius: Foghorn Therapeutics: Current Employment, Current equity holder in publicly-traded company. Cosmopoulos: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company. Kutok: Epizyme, Inc.: Current Employment, Current equity holder in publicly-traded company.
39

Cheung, Kam, Gloria Juan, William Wayne, Kelly Hanestad, Kathleen Keegan, Justin Huard, Patricia McElroy, et al. "AMG 900, a Potent and Highly Selective Aurora Kinase Inhibitor Shows Promising Preclinical Activity Against Acute Myeloid Leukemia Cell Lines In Vitro and In Vivo." Blood 122, no. 21 (November 15, 2013): 3823. http://dx.doi.org/10.1182/blood.v122.21.3823.3823.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Aurora kinases A and B play essential roles in multiple stages of mitosis and are frequently overexpressed in a subset of human cancers, including acute myeloid leukemia (AML) (Ikezoe T et al, 2007). AMG 900, a potent and highly selective small molecule inhibitor of aurora kinases, is currently in Phase 1 clinical testing in adult patients with AML. In this study, we report the preclinical effects of AMG 900 in AML cell lines. We show that AMG 900 inhibits the phosphorylation of Histone H3 on serine-10 (a proximal substrate of aurora-B) leading to aborted cell division, apoptosis and/or polyploidy. We evaluated the activity of AMG 900 and two other well-characterized aurora kinase inhibitors [AZD1152-hQPA (B-selective AKI) and MLN8054 (A-selective AKI)] in a panel of AML cell lines. AMG 900 inhibited proliferation in all 10 cell lines at single-digit nanomolar concentrations. At effective concentrations, AMG 900 and AZD1152-hQPA showed similar cellular phenotypes, indicating that the activity of AMG 900 may occur through inhibition of aurora-B. A subset of cell lines sensitive to AMG 900 and MLN8054 were insensitive to AZD1152-hQPA, suggesting that AMG 900 may be less susceptible to resistance mediated by drug-efflux (Grundy M et al, 2011). Two AMG 900 oral dosing schedules are being evaluated in the ongoing AML clinical trial; patients receive either 4 or 7 consecutive daily doses followed by a drug holiday in 14-day cycles. In this study, we evaluated the in vivo anti-proliferative effects of AMG 900 using the two dose schedules in the skeleton of NOD/SCID IL2γnull mice bearing MOLM-13 (AML) cells expressing luciferase. To assess tumor cell proliferation in vivo, we used 18FLT (radioactive thymidine analog) PET/CT imaging, a technique that has been used to monitor early treatment response in the bone marrow of AML patients (Vanderhoek M et al, 2011). Mice were imaged for luciferase activity and 18FLT uptake before treatment and at multiple time-points during the drug holiday phase within the 14-day cycle. While the two AMG 900 dosing schedules resulted in a similar decrease in tumor burden across study time points (as measured by luciferase activity), they differed in the timing of skeletal 18FLT responses. Mice administered AMG 900 showed an attenuated skeletal 18FLT uptake compared with the vehicle group, followed by an 18FLT flare. This 18FLT flare event is notably higher using the AMG 900 4-day schedule, although the cumulative dose is similar for both schedules. This difference in 18FLT flaring may indicate the schedules differ in the duration and/or level of target inhibition in the skeletal tumor and bone marrow cells. Mice treated with sunitinib (positive control agent) did not show a skeletal 18FLT flare during the drug holiday, suggesting its mode of action is distinct from that of AMG 900. At the end of the study, mouse bone marrow was assessed for tumor burden by flow cytometry. Mice treated with AMG 900 showed a significant decrease in tumor burden compared with the vehicle group. Interestingly, the mice administered AMG 900 7-day schedule showed the most suppression of tumor growth compared with either AMG 900 4-day schedule or sunitinib. Together, our preclinical studies demonstrate that AMG 900 is a potent inhibitor of aurora kinases that robustly suppresses the growth of AML cells in vitro and in vivo. Furthermore, we highlight the utility of in vivo imaging to monitor AMG 900 drug action, which may help to inform future dose scheduling and drug combination studies. Disclosures: Cheung: Amgen Inc: Employment, Equity Ownership. Juan:Amgen Inc.: Employment, Equity Ownership. Wayne:Amgen Inc.: Employment, Equity Ownership. Hanestad:Amgen Inc.: Employment, Equity Ownership. Keegan:Amgen Inc.: Employment, Equity Ownership. Huard:Amgen Inc.: Employment, Equity Ownership. McElroy:Amgen Inc.: Employment, Equity Ownership. Stanton:Amgen Inc.: Employment, Equity Ownership. Bush:Amgen Inc.: Employment, Equity Ownership. Kendall:Amgen Inc.: Employment, Equity Ownership. Radinsky:Amgen Inc.: Employment, Equity Ownership. Abella:Amgen Inc. : Employment, Equity Ownership. Pieslor:Amgen Inc.: Employment, Equity Ownership. Friberg:Amgen Inc.: Employment, Equity Ownership. Coxon:Amgen Inc.: Employment, Equity Ownership. Gamelin:Amgen Inc: Employment, Equity Ownership. Payton:Amgen Inc.: Employment, Equity Ownership. Off Label Use: AMG 900 is currently in phase 1 clinical development, there is no approved label.
40

Chen, Jun, Sha Jin, Paul Tapang, Stephen K. Tahir, Morey Smith, John Xue, Haichao Zhang, et al. "CDK9 Inhibition Reverses Resistance to ABT-199 (GDC-0199) By Down-Regulating MCL-1." Blood 124, no. 21 (December 6, 2014): 2161. http://dx.doi.org/10.1182/blood.v124.21.2161.2161.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract All authors are employees of AbbVie and participated in the design, conduct, and interpretation of these studies. AbbVie and Genentech provided financial support for these studies and participated in the review and approval of this publication. The BCL-2-selective inhibitor ABT-199 has demonstrated efficacy in numerous preclinical models of hematologic malignancies without causing thrombocytopenia, a dose-limiting toxicity associated with the BCL-2/BCL-XL inhibitor navitoclax (Souers et al. 2013. Nat. Med. 19, 202-208). ABT-199 has also demonstrated clinical activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin’s lymphoma (NHL) (Seymour et al. 2014. J. Clin. Oncol. 32, 448s; Davids et al. 2014. J. Clin. Oncol. 32, 544s). Despite these encouraging early clinical data, some subjects do not respond to ABT-199 or progress while on treatment. Pre-clinical models indicate that both intrinsic and acquired resistance may be a consequence of MCL-1 expression. Consequently, we have explored potent and selective small molecule inhibitors of CDK9, a kinase known to maintain the expression of MCL-1 through its role in p-TEFb-mediated transcription. Inhibition of CDK9 resulted in the rapid loss in RNA polymerase II phosphorylation (Serine 5) and MCL-1 expression that was closely followed by the induction of apoptosis in MCL-1-dependent cell lines, a cellular response that could be rescued by overexpression of BCL-2. Substantial synergy was observed between CDK9 inhibitors and ABT-199 in a number of hematologic cell lines with intrinsic or acquired resistance to ABT-199. Direct inhibition of MCL-1 with the small molecule BH3 mimetic A-1210477 was also highly synergistic with ABT-199, further validating the utility of co-inhibiting MCL-1 and BCL-2 function simultaneously in ABT-199 resistant tumors. Importantly, the CDK9 inhibitor-ABT-199 combination was well tolerated in vivo and demonstrated efficacy superior to either agent alone in xenograft models of non-Hodgkin’s lymphoma (NHL) and acute myelogenous leukemia (AML). These data indicate that CDK9 inhibitors may be highly efficacious when used in combination with ABT-199 for the treatment of hematologic malignancies. Disclosures Chen: Abbvie: Employment, Equity Ownership. Jin:Abbvie: Employment, Equity Ownership. Tapang:abbvie: Employment, Equity Ownership. Tahir:abbvie: Employment, Equity Ownership. Smith:abbvie: Employment, Equity Ownership. Xue:abbvie: Employment, Equity Ownership. Zhang:abbvie: Employment, Equity Ownership. Gao:abbvie: Employment, Equity Ownership. Tong:abbvie: Employment, Equity Ownership. Clark:abbvie: Employment, Equity Ownership. Ricker:abbvie: Employment, Equity Ownership. Penning:abbvie: Employment, Equity Ownership. Albert:abbvie: Employment, Equity Ownership. Phillips:abbvie: Employment, Equity Ownership. Souers:abbvie: Employment, Equity Ownership. Leverson:abbvie: Employment, Equity Ownership.
41

Penebre, Elayne, Kristy G. Kuplast, Christina R. Majer, L. Danielle Johnston, Nathalie Rioux, Michael Munchhof, Lei Jin, et al. "Identification of a First-in-Class PRMT5 Inhibitor with Potent in Vitro and in Vivo Activity in Preclinical Models of Mantle Cell Lymphoma." Blood 124, no. 21 (December 6, 2014): 438. http://dx.doi.org/10.1182/blood.v124.21.438.438.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Protein Arginine Methyltransferase-5 (PRMT5) has been reported to play a role in multiple diverse cellular processes including tumorigenesis. Overexpression of PRMT5 has been demonstrated in cell lines and primary patient samples derived from lymphomas, particularly Mantle Cell Lymphoma (MCL). Furthermore, knockdown of PRMT5 expression inhibits the proliferation of MCL cell lines. The mechanisms behind the oncogenic potential of PRMT5 are unclear, but the protein has been postulated to regulate processes such as cell death, cell cycle progression, and RNA processing through the dimethylation of arginine residues within a variety of cytoplasmic and nuclear target proteins. Epizyme developed small molecule inhibitors of PRMT5 enzyme activity in order to understand the role of PRMT5-mediated arginine methylation in tumorigenesis and to develop PRMT5-targeted cancer therapeutics. Here, we describe the identification and characterization of a potent and selective inhibitor of PRMT5 with anti-proliferative effects in both in vivo and in vitro models of MCL. A diverse compound library was screened for inhibitors of arginine methylation by purified recombinant PRMT5:MEP50 complex and multiple hits were identified. The inhibitors are SAM uncompetitive, peptide competitive and bind with the PRMT5:MEP50 complex in a unique binding mode not previously observed. Further optimization yielded YQ36286, an orally available inhibitor of PRMT5 with enzymatic activity in biochemical assays with an IC50 in the low nM range and broad selectivity against a panel of other histone methyltransferases. YQ36286 demonstrated potent cellular activity as measured by its ability to inhibit symmetric dimethylation of SmD3, a cytoplasmic PRMT5 substrate in a time- and concentration-dependent manner. Treatment of MCL cell lines with YQ36286 led to inhibition of SmD3 methylation and cell killing, with IC50s in the nM range. Oral dosing of YQ36286 demonstrated dose-dependent anti-tumor activity in multiple MCL xenograft models. In xenograft studies with the Z138 MCL cell line, near 95% tumor growth inhibition was observed after 21 days of dosing with a corresponding decrease in symmetrically dimethylated levels of PRMT5 substrates. In summary, we have developed the first potent and selective small molecule inhibitor of PRMT5 that has cellular activity and in vivo efficacy. MCL cells are dependent on PRMT5 activity for their survival as demonstrated with YQ36286. This small molecule represents a starting point for the development of PRMT5 inhibitors as potential cancer therapeutics. Disclosures Penebre: Company stock options: Equity Ownership; Epizyme Inc.: Employment; GSK Research Funding: Research Funding. Kuplast:GSK research funding: Research Funding; Company Stock options: Equity Ownership; Epizyme Inc.: Employment. Majer:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Johnston:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Rioux:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Munchhof:Epizyme Inc.: Employment; GSK research funding: Research Funding. Jin:Epizyme Inc.: Employment; GSK research funding: Research Funding; Company stock options: Equity Ownership. Boriak-Sjodin:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Wigle:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Jacques:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. West:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Lingaraj:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Stickland:GSK research funding: Research Funding; Company Stock options: Equity Ownership; Epizyme Inc.: Employment. Ribich:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Raimondi:Epizyme: Employment, Equity Ownership; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Porter-Scott:Company stock options: Equity Ownership; GSK research funding: Research Funding; Epizyme Inc.: Employment. Waters:Epizyme, Inc: Employment, Equity Ownership; GSK research funding: Research Funding. Pollock:Epizyme: Employment, Equity Ownership; GSK research funding: Research Funding. Smith:GSK research funding: Research Funding; Epizyme: Employment, Equity Ownership. Barbash:GlaxoSmithKline Pharmaceuticals: Employment. Kruger:GlaxoSmithKline Pharmaceuticals: Employment, Equity Ownership. Copeland:Mersana: Membership on an entity's Board of Directors or advisory committees; Epizyme, Inc: Employment, Equity Ownership; Celgene, Inc: Research Funding; Eisai Inc: Research Funding; Glaxo Smith Kline, Inc: Research Funding; Multiple Myeloma Research Foundation: Research Funding; Leukemia and Lymphoma Society: Research Funding; New Enterprise Associates: Ad hoc consultant, Ad hoc consultant Other. Moyer:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Chesworth:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding. Duncan:Epizyme Inc.: Employment; Company Stock options: Equity Ownership; GSK research funding: Research Funding.
42

Weyland, Kurt. "“Growth With Equity” in Chile's New Democracy?" Latin American Research Review 32, no. 1 (1997): 37–68. http://dx.doi.org/10.1017/s0023879100037651.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
The new democracy in Chile provides an interesting test case for two influential lines of thinking on Latin American political economy. Both these perspectives have claimed that the recently installed civilian regimes would find it exceedingly difficult to effect equity-enhancing change. One hypothesis has stressed the impediments posed by a capitalist free-market system to measures favoring the poor. The other has emphasized the obstacles presented by a transition to democracy that avoids a rupture with the preceding conservative dictatorship. Because the democratic government led by Patricio Aylwin (1990–1994) faced both types of constraints, it should have been particularly unlikely to achieve greater social fairness.
43

Rychak, Emily, Derek Mendy, Karen Miller, Jim Leisten, Rama Krishna Narla, Heather K. Raymon, Rajesh Chopra, and Antonia Lopez-Girona. "The Novel mTOR Kinase Inhibitor CC-223 Demonstrates Significant Activity In In Vitro Models Of Multiple Myeloma (MM), Both As a Single Agent and In Combination With The Approved Agents, Dexamethasone, Lenalidomide and Pomalidomide." Blood 122, no. 21 (November 15, 2013): 3160. http://dx.doi.org/10.1182/blood.v122.21.3160.3160.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Over expression of the PI3 kinase/mTOR/AKT pathway has been well documented in MM patient biopsies and human MM cell lines, suggesting this pathway plays a key role in the survival and proliferation of malignant plasma cells. Rapamycin and the rapalogs are allosteric inhibitors of the mTORC1 complex (consisting of mTOR, raptor, mLST8 and PRAS40), inducing mainly cytostatic effects but not cell death. Inhibition of mTORC1 prevents a negative feedback loop to the mTORC2 complex (consisting of mTOR, Rictor, mLST8 and Sin 1) leading to the phosphorylation of AKT. Phosphorylated AKT is a key inducer of anti-apoptosis mechanisms and cell cycle progression, which may explain the limited results of the rapalogs in the clinic. Recently developed mTOR kinase inhibitors (i.e., CC-223) target both mTORC1 and mTORC2 complexes in order to inhibit tumor growth and importantly, induce cell death. Here we evaluate the effects of CC-223 on a panel of MM cell lines, in combination with current standard of care agents in MM (the corticosteroid, dexamethasone [DEX] and the IMiD® immunomodulatory drugs, lenalidomide [LEN] and pomalidomide [POM]), as well as in the context of LEN resistance. Single agent CC-223 was shown to inhibit cell proliferation in a panel of 10 MM cell lines achieving IC50 values between 0.1-1 µM following 5 days of treatment. CC-223 also reduced cell viability reaching IC50 values between 0.4-1 µM in 5 out of 10 MM cell lines tested. CC-223 induced concentration-dependent G1 phase arrest within 24h of treatment followed by an induction of cell death by 48h. The anti-MM tumor activity of CC-223 (0.3-10 mg/kg) was further tested in SCID mice with xenotransplants of NCI-H929 grown to approximately 100-150 mm3 in size. A dose-dependent tumor growth inhibition and tumor growth delay was seen with once daily dosing of CC-223. Combination of CC-223 with standard of care therapy compounds was also evaluated in vitro. The combination of CC-223 and DEX demonstrated synergistic effects on the inhibition of cell proliferation in 6 MM cell lines (combination index: 0.0002-0.38) tested over 5 days. CC-223 also had synergistic effects on the same panel of MM cell lines when combined with LEN (combination index: 0.05-0.8). Acquisition of drug resistance in patients receiving standard of care therapies is still one of the major clinical problems in MM. POM, the next generation of IMiD® immunomodulatory agents, has shown clinically meaningful results in patients that are resistant or have relapsed to their drug regimens, including LEN. We have recently developed in vitro cellular models of LEN-resistance using the H929 MM cell line. H929 cells with acquired resistance to LEN (H929 R10-1, R10-2, R10-3 and R10-4) were shown to have one copy number loss of cereblon compared to their matched LEN-sensitive control (H929 D1). In addition to this, protein expression analysis identified that these resistant cell lines also gained the activation of signaling pathways such as PI3K/AKT/mTOR, MEK/MAPK as well as anti-apoptotic factors. For example, S473 AKT phosphorylation was highly elevated in LEN-resistant cell lines which correlated with loss of PTEN protein expression (H929 R10-3 and R10-4). Interestingly, regardless of PI3K/AKT/mTOR pathway status, all LEN-sensitive and resistant H929 cells responded to CC-223 treatment with a strong inhibition of cell proliferation (H929 D1 IC50 0.2 µM, and H929 R10 1-4 IC50 0.2-0.35 µM) and to a lesser effect, induction of cell death, over a 5 day period. Similar to the panel of MM cell lines, G1 arrest occurred after 24h treatment and cell death (Sub-G1) was increased by 72h of treatment. CC-223 treatment reduced S473 pAKT and p-4EBP1 after 1h while total AKT and 4EBP1 remained unchanged in both the sensitive and resistant MM cell lines. Combination treatment of LEN-sensitive and resistant H929 cells with CC-223 and POM had synergistic inhibitory effects on cell proliferation (combination index: 0.35-0.7) and cell viability (combination index: 0.15-0.42). In conclusion, the mTOR kinase inhibitor, CC-223 potently inhibited MM cell proliferation by inducing G1 arrest and cell death in a panel of MM cell lines and reduction of tumor volume in vivo. The combination of LEN, POM or DEX with CC-223 had synergistic effects on MM cell proliferation and viability. Therefore, CC-223 in combination with other standard of care agents could become an important clinical tool for the treatment of MM in the future. Disclosures: Rychak: Celgene Corporation: Employment, Equity Ownership. Mendy:Celgene: Employment, Equity Ownership. Miller:Celgene Corporation: Employment, Equity Ownership. Leisten:Celgene Corporation: Employment, Equity Ownership. Narla:Celgene Corporation: Employment, Equity Ownership. Raymon:Celgene Corporation: Employment, Equity Ownership. Chopra:Celgene: Employment, Equity Ownership. Lopez-Girona:Celgene: Employment, Equity Ownership.
44

McElligott, David L., Edward Kesicki, Kannan Karukarichi, Hyeseok Shim, Rui Wang, Albert S. Yu, Parisa Zolfaghari, et al. "Development of Inhibitors of PIP4K2 As a Treatment for Patients with Hematologic Malignancies." Blood 132, Supplement 1 (November 29, 2018): 213. http://dx.doi.org/10.1182/blood-2018-99-118355.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Phosphoinositide signaling is central to many cellular processes including the cell survival pathway known as autophagy. While there is evidence that autophagy can suppress tumorigenesis under certain circumstances, there is increasingly abundant evidence that autophagy promotes tumorigenesis and tumor cell survival in solid tumors and hematologic malignancies. Autophagy is a complex process that has evolved to promote survival of cells under a variety of stress or nutrient starvation conditions. There are a multitude of molecules and structures involved in initiating, promoting, and resolving the autophagy process. It is known that PI5P is an important component for the resolution of autophagy. PIP4K2α,β,γ are a family of lipid kinases that convert PI5P to PI(4,5)P2. These enzymes have recently been shown to be critical for the fusion of autophagosomes with lysosomes. This known activity, coupled with the observation that PIP4K2 activity is essential for the survival and leukemia initiating potential of human and mouse acute myeloid leukemia (AML) cells, suggest that the PIP4K2 family of enzymes may be a promising target for a new class of therapeutics for the treatment of hematologic malignancies. We have investigated the role of PIP4K2 enzymes in supporting the survival of cancer cells by developing potent and selective inhibitors of PIP4K2 enzymatic activity. A screen of human cancer cell lines show that potent and selective inhibitors of PIP4K2 are effective at inhibiting growth of a variety of hematologic cancers including leukemia- and lymphoma-derived lines. In vivo studies demonstrate that these inhibitors induce rapid regression of an AML tumor (MOLM-16) in a mouse xenograft model. These studies show a dose-dependent control of tumor growth with sustained regression of tumor volume with QD oral dosing of a prototype molecule. Body weights of the mice were stable over the course of the study suggesting that the molecule is well tolerated in this dosing protocol. A preliminary toxicity study in rats has not revealed any identifiable toxicity at doses up to 100 mg/Kg given QD/PO for 14 days. Additional in vitro safety studies suggest minimal safety concerns due to off-target activity. Exploration of the structure activity relationship of PIP4K2 inhibitors, using fragment and structure-based drug discovery, has led to highly potent and selective molecules with exceptional drug-like properties. A clinical development candidate has been selected and that candidate is currently in the late stages of preclinical studies preceding anticipated clinical entry in early 2019. Disclosures McElligott: Petra Pharma: Employment, Equity Ownership. Kesicki:Petra Pharma: Employment, Equity Ownership. Karukarichi:Petra Pharma: Employment, Equity Ownership. Shim:Petra Pharma: Employment, Equity Ownership. Wang:Petra Pharma: Employment, Equity Ownership. Yu:Petra Pharma: Employment, Equity Ownership. Zolfaghari:Petra Pharma: Employment, Equity Ownership. Linstrom:Sprint Bioscience: Employment, Equity Ownership. Persson:Sprint Bioscience: Employment, Equity Ownership. Hoglund:Sprint Bioscience: Employment, Equity Ownership. Ericsson:Sprint Bioscience: Employment, Equity Ownership. Trésaugues:Sprint Bioscience: Employment, Equity Ownership. Livendahl:Sprint Bioscience: Employment, Equity Ownership. Santangelo:Sprint Bioscience: Employment, Equity Ownership. Viklund:Sprint Bioscience: Employment, Equity Ownership. Pettersson:Sprint Bioscience: Employment, Equity Ownership. Wähling:Sprint Bioscience: Employment, Equity Ownership. Forsblom:Sprint Bioscience: Employment, Equity Ownership. Karlsson:Sprint Bioscience: Employment, Equity Ownership. Ginman:Sprint Bioscience: Employment, Equity Ownership. Braga:Sprint Bioscience: Employment, Equity Ownership. Henley:Sprint Bioscience: Employment, Equity Ownership. Talagas:Sprint Bioscience: Employment, Equity Ownership. Rahm:Sprint Bioscience: Employment, Equity Ownership. Johansson:Sprint Bioscience: Employment, Equity Ownership. Martinsson:Sprint Bioscience: Employment, Equity Ownership. Andersson:Sprint Bioscience: Employment, Equity Ownership. Cantley:Petra Pharma: Equity Ownership.
45

Csibi, Fred, Nan Ji, Bin Yang, Karen Yuan, Michele Mayo, Haojing Rong, Scott Rusin, et al. "Small Molecule-Induced, Selective STAT3 Degradation Leads to Anti-Tumor Activity in STAT3-Dependent Heme Malignancies." Blood 134, Supplement_1 (November 13, 2019): 3803. http://dx.doi.org/10.1182/blood-2019-128981.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Targeted protein degradation mediated by small molecule degraders represents a new and exciting therapeutic modality to target difficult-to-drug oncogenic proteins including transcription factors. These molecules bind to both the target protein and an E3 ligase, enabling the formation of a ternary complex that leads to ubiquitination and subsequent degradation of the target protein by the proteasome. STAT3 (signal transducers and activators of transcription 3) is a transcription factor and a member of the STAT protein family. In response to cytokines and growth factors, STAT3 is phosphorylated by receptor-associated serine/threonine kinases, and phosphylated STAT3 (pSTAT3) then forms dimers that translocate into the nucleus, binds to DNA, and regulate transcription. STAT3 is frequently mutated and activated in numerous cancers including clinically aggressive hematologic malignancies with high unmet medical need. Mechanistically, aberrant activation of STAT3 has been directly linked to the promotion of cancer cell survival, proliferation and immune evasion, thus making it a highly attractive target for oncology. Potent and selective agents specifically and directly targeting STAT3 have remained elusive, however. Herein we report the discovery of a potent and selective STAT3 heterobifunctional degrader, KYM-003, which displays strong anti-tumor activity in models of STAT3-dependent heme malignancies. KYM-003 degrades STAT3 via an E3 ligase-dependent mechanism. It strongly binds to STAT3 and a E3 ligase, leading to the formation of the a productive ternary complex, which leads to ubiquitination of STAT3 and subsequent proteasomal degradation. KYM-003 robustly degraded STAT3 in a number of primary cells or cell lines with DC50 < 100 nM. Degradation was highly selective for STAT3 vs >10,000 other detected proteins (including all other STAT family members) in cell lines and human PBMCs. Degradation of STAT3 by KYM-003 led to significant downregulation of STAT3 target genes, such as SOCS3, MYC, and PIM1. Importantly, total STAT3 and pSTAT3 levels in tumors were reduced by >90% for at least 24 hours after a single dose of KYM-003 and repeated dosing of KYM-003 showed dose-dependent antitumor activity in xenograft models of heme malignancies. Collectively, our data demonstrates that KYM-003 is a potent and selective STAT3 degrader that exhibited strong anti-tumor activity in vitro and in vivo. These data support STAT3 degraders as a new and exciting therapeutic opportunity in heme malignancies. Disclosures Csibi: kymera Therapeutics: Employment, Equity Ownership. Ji:Kymera Therapeutics: Employment, Equity Ownership. Yang:Kymera Therapeutics: Employment, Equity Ownership. Yuan:Kymera Therapeutics: Employment, Equity Ownership. Mayo:kymera Therapeutics: Employment, Equity Ownership. Rong:Kymera Therapeutics: Employment, Equity Ownership. Rusin:Kymera Therapeutics: Employment, Equity Ownership. Sharma:kymera Therapeutics: Employment, Equity Ownership. Loh:Kymera Therapeutics: Employment, Equity Ownership. Li:Kymera Therapeutics: Employment, Equity Ownership. Townson:Kymera Therapeutics: Employment, Equity Ownership. Chen:kymera therapeutics: Employment, Equity Ownership. Kamadurai:Kymera Therapeutics: Employment, Equity Ownership. Walker:Kymera Therapeutics: Employment, Equity Ownership. Gollob:Kymera Therapeutics: Employment, Equity Ownership. Mainolfi:Kymera Therapeutics: Employment, Equity Ownership.
46

Ding, Bei Bei, John Dixon Gray, Irina Krapf, Yanliang Zhang, Nan Zhang, Qingdong Mark Deng, Angel Wei, et al. "Development of a Genetically-Engineered Allogeneic Anti-CD38 T Cell Therapy Utilizing a Novel Antigen Receptor Structure." Blood 134, Supplement_1 (November 13, 2019): 4444. http://dx.doi.org/10.1182/blood-2019-131586.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Background: Autologous Chimeric Antigen Receptor (CAR) T cell therapy has shown great promise as a treatment modality for a variety of hematological malignancies. But autologous cell therapies still face several practical hurdles, including reliance on patient immune cells and manufacturing difficulties. Sorrento has pioneered an allogeneic T cell therapy approach utilizing genetic engineering of donor-derived T cells to express a Dimeric Antigen Receptor (DAR). The first DAR-T cell therapy being developed is targeted against CD38, a clinically-validated antigen in multiple myeloma. Preclinical data demonstrate potent anti-tumor activity in both in vitro assays and in vivo studies against CD38-expressing lymphoma and multiple myeloma (MM) cell lines. Methods: Anti-CD38 DAR-T cells were generated through genetic engineering of T cells derived from healthy donors inserting the anti-CD38 DAR construct into the TRAC gene locus resulting in loss of endogenous TCR expression while expressing the DAR. Three distinct DAR constructs were utilized differing only in the intracellular signaling components, namely CD28/CD3zeta, 4-1BB/CD3zeta and CD28/4-1BB/CD3zeta. The CD38 DAR-T were expanded and purified for subsequent preclinical studies. Using in vitro assays, the 3 different CD38 DAR-T cells were evaluated against multiple myeloma and lymphoma cell lines for specific cytotoxicity as well as stimulus-induced cytokine secretion and cell expansion. The in vivo anti-tumor activity was assessed using luciferase-expressing RPMI8226 cells in NSG mice in a model of disseminated disease. A single dose of anti-CD38 DAR-T cells or relevant control cells was administered and tumor burden was assessed weekly using bioluminescence imaging. Results: An anti-CD38 DAR gene was efficiently integrated into TRAC locus of T cells from healthy donors by one step knock out/knock in (KOKI) methodology with high efficiency (~40-80% CD38 DAR expression and ~90% TCR knock out). Following a CD3-depletion step, the TCR-positive T cells were less 1%. When co-cultured with CD38-positive tumor cells, anti-CD38 DAR T cells killed as effectively as retroviral anti-CD38 CAR-T cells with similar cytokine secretion profiles while no cytotoxicity was observed against CD38-negative cancer cells. Moreover, in vivo DAR-T cells showed better killing activity against multiple myeloma cell lines than CAR-T cell with anti-CD38 4-1BB/CD3zeta DAR demonstrating the best anti-tumor activity in an NSG mouse model. The anti-CD38 DAR-T cells with 41BB/CD3 zeta internal signals have been selected for clinical development. Conclusions: All tested anti-CD38 DAR-T cells exhibited potent in vitro and in vivo anti-tumor activity. Direct comparison of three different cytoplasmic signaling compositions of the DAR allowed for selection of the most potent construct, namely the anti-CD38 DAR utilizing 4-1BB and CD3zeta signaling domains. Based on these data, further development of CD38 DAR-T therapy for hematological malignancies is warranted. GMP manufacturing of the allogeneic anti-CD38 DAR-T cells has been initiated. Disclosures Ding: Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Gray:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Krapf:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Zhang:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Zhang:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Deng:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Wei:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Knight:Sorrento Therapeutics, Inc.: Employment, Equity Ownership. Zeldis:Sorrento Therapeutics Inc: Employment, Equity Ownership. Kaufmann:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties. Ji:Sorrento Therapeutics Inc: Employment, Equity Ownership, Patents & Royalties; Celularity, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Guo:Sorrento Therapeutics, Inc.: Employment, Equity Ownership, Patents & Royalties.
47

Fiore, Christopher, Michael R. McKeown, Emily Lee, Matthew L. Eaton, and Christian C. Fritz. "SY1425 (tamibarotene) Induces Profound Transcriptional Changes in AML Tumors with High Retinoic Acid Receptor Alpha." Blood 128, no. 22 (December 2, 2016): 1523. http://dx.doi.org/10.1182/blood.v128.22.1523.1523.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Retinoic acid receptor alpha (RARα) regulates myeloid differentiation and proliferation through the regulation of specific sets of genes. When unbound by a ligand, RARα is a repressive transcription factor while in its ligand-bound state it functions as a transcriptional activator. Previously, blast cells from a subset of individuals with non-APL AML were found to have a super-enhancer (SE), as revealed by H3K27 acetyl ChIP-Seq, associated with the RARA locus (hereafter called RARA-high), suggesting that tumor cell proliferation may have a dependency on RARA that can be exploited for therapeutic benefit. SEs are exceptionally large, highly active chromatin regions that are densely occupied by transcription factors and have been implicated in oncogene expression. Indeed, RARA-high non-APL AML cell lines showed >1000-fold increased sensitivity compared to RARA-low cells to the potent and selective RARα agonist SY-1425 (tamibarotene) as well as efficacy in non-APL AML patient derived xenograft models with a dependency on RARA. Since RARα is a transcription factor and the direct target of SY-1425, we investigated the change SY-1425 had on the transcriptional program of non-APL AML cell lines and the mechanism underlying those changes. Expression profiling on a panel of AML cell lines revealed that RARA-high AML cell lines had profound transcriptional changes in response to SY-1425, with 437 genes significantly changed, while RARA-low cell lines did not show significant gene expression changes. Gene set enrichment analysis (GSEA) of three RARA-high AML cell lines revealed that the genes upregulated by SY-1425 in the RARA-high cells are associated with immune signaling, interferon induction, protein secretion, and pathways associated with complement, MHC and integrin functions, all pathways indicative of more differentiated blood cells. Signatures downregulated by SY-1425 include MYC target genes. These findings are consistent with SY-1425 increasing the expression of genes involved in differentiation and decreasing those involved in proliferation. Genome-wide ChIP-Seq analysis revealed an increase in H3K27 acetylation at loci found to have strong RARα peaks as well as increased expression of those genes upon treatment with SY-1425. Together, these data support a model in which RARα binding nucleates functional enhancers in response to SY-1425 thereby upregulating proximal target gene expression and promotion of differentiation. The gene expression and epigenomic responses of RARA-high AML cell lines to SY-1425 were found to be similar to the responses of an APL cell line (NB-4) to retinoids or SY-1425. Gene sets identified in response to either retinoid treatment or genetic perturbation, such as forced expression or RAR-fusions or knockdown, matched the gene sets identified in RARA-high AML cell lines. Furthermore, the quantitative response of both NB-4 and RARA-high AML cell lines to SY-1425 was found to be similar. Across the genome, RARα binding was highly conserved between NB-4 and RARA-high AML cell lines with less overlap with the RARA-low cell lines. For example, the transcriptional and H3K27 acetylation alterations at the known PML-RARα target gene TGM2 following retinoid treatment was similar in NB-4 and the RARA- high cell lines. This locus also had a strong RARα binding site that is conserved among the cell lines and co-localized with a strong H3K27 acetylation peak. Consistent with the pattern of occupancy of RARα on the genome, the transcriptional response of the RARA enhancer-high cell lines to SY-1425 treatment was similar to the response of APL ex-vivo patient samples to retinoic acid treatment. These data support a model of a common biological response to retinoids between cells with the RARA-PML translocation in APL and cells with the RARA SE in AML. The mechanistic studies described here support the therapeutic potential of SY-1425 in myeloid leukemia patients who have a SE associated with RARA. A biomarker directed clinical trial of SY-1425, a potent and selective RARα agonist, in a subset of AML and MDS patients with an altered RARA locus (clinicaltrials.gov, NCT02807558) is supported by these data. Disclosures Fiore: Syros Pharmaceuticals: Employment, Equity Ownership. McKeown:Syros Pharmaceuticals: Employment, Equity Ownership. Lee:Syros Pharmaceuticals: Employment, Equity Ownership. Eaton:Syros Pharmaceuticals: Employment, Equity Ownership. Fritz:Syros Pharmaceuticals: Employment, Equity Ownership.
48

Lowe, Eric, Andrea R. Fan, Jing Jiang, Henry W. B. Johnson, Christopher J. Kirk, Dustin McMinn, Tony Muchamuel, Yu Qian, and Brian B. Tuch. "Blocking Protein Secretion with Novel Small Molecule Inhibitors of Sec61 Represents a Potential Treatment Strategy Against Hematologic Malignancies." Blood 134, Supplement_1 (November 13, 2019): 408. http://dx.doi.org/10.1182/blood-2019-123782.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Secreted and membrane proteins play key roles in cancer development, including as autocrine growth factors, receptors in oncogenic signaling cascades, and checkpoints in immune system evasion. The biogenesis of most secreted and transmembrane proteins involves cotranslational translocation of nascent polypeptides ("clients") into the endoplasmic reticulum (ER) through the Sec61 translocon. Multiple inhibitors of protein secretion have been described that target Sec61 and have antitumor activity but lack adequate pharmaceutical properties or tolerability to be studied clinically. Here we describe novel small molecule inhibitors of Sec61 which exhibit activity against hematologic tumor cells in vitro and in vivo. KZR-8834 (8834) was identified through a screening campaign for novel inhibitors of Sec61-dependent protein secretion, where it exhibited nanomolar potency against multiple Sec61 client proteins of therapeutic value, including several immune checkpoint proteins. The broad Sec61 client inhibition profile of 8834 led to its in vitro assessment of anti-cancer activity against a panel of 346 human cancer cell lines. 8834 displayed broad cytotoxic activity against both solid and hematologic tumor types with high potency for hematologic malignancies including acute lymphoid leukemia (mean IC50=317nM, n=8 cell lines), acute myeloid leukemia (IC50=359nM, n=14), lymphoma (IC50=250nM, n=15) and multiple myeloma (IC50=352nM, n=11). In mouse xenograft models of multiple myeloma (H929) and mantle cell lymphoma (Mino), once weekly administration of 8834 or KZR-9261 (8834 analog) resulted in &gt;90% tumor growth inhibition without significant clinical toxicity. Two multiple myeloma cell lines, H929 and U266, were chosen to assess cellular response due to their sensitivity and resistance to 8834, respectively. In H929 cells, 250nM 8834 induced activation of caspase 3/7 (&gt;15 fold) within 8 hours of exposure which corresponded with a cell viability IC50 of 98nM at 24 hours. In contrast, no caspase 3/7 activation was noted in U266 cells through 24 hours of exposure, which corresponded with minimal effects on cell viability. Gene expression profiling by RNAseq and quantitative proteomic profiling by mass spectrometry was performed on these cell lines to elucidate mechanisms of sensitivity and resistance to Sec61 inhibition. Both methods revealed rapid upregulation of ER stress response genes/proteins and activation of the unfolded protein response, which was greater in H929 cells and confirmed by immunoblot and QPCR. Gene set enrichment analysis revealed significantly higher basal levels of ER stress-related genes in H929 vs U266 cells, suggesting ER stress response capacity as a possible predictive biomarker. In summary, blockade of Sec61-dependent translocation of secreted and membrane proteins with novel small molecule inhibitors exhibits a broad antitumor profile in vitro, potentially in part through activation of proteotoxic stress. These effects translate into therapeutic activity in multiple mouse xenograft models, demonstrating a potential novel treatment for hematologic malignancies. Disclosures Lowe: Kezar Life Sciences: Employment, Equity Ownership. Fan:Kezar Life Sciences: Employment, Equity Ownership. Jiang:Kezar Life Sciences: Employment, Equity Ownership. Johnson:Kezar Life Sciences: Employment, Equity Ownership. Kirk:Kezar Life Sciences: Employment, Equity Ownership. McMinn:Kezar Life Sciences: Employment, Equity Ownership. Muchamuel:Kezar Life Sciences: Employment, Equity Ownership. Qian:Kezar Life Sciences: Employment, Equity Ownership. Tuch:Kezar Life Sciences: Consultancy.
49

Yang, Jun, Andong Guo, Xueming Li, and Tai Huang. "Study of the Impact of a High-Speed Railway Opening on China’s Accessibility Pattern and Spatial Equality." Sustainability 10, no. 8 (August 19, 2018): 2943. http://dx.doi.org/10.3390/su10082943.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
China’s high-speed rail was inaugurated in 2008; it has greatly improved accessibility, and reduced the time required to travel between cities, but at the same time, has caused an unfair distribution of accessibility levels. Therefore, this paper analyzes urban traffic roads and socio-economic statistics, using network analysis methods, accessibility coefficients of variation, and social demand indexes to explore the spatial and temporal characteristics of transport accessibility and spatial equity in China. By 2015, the national transport accessibility level will form a new pattern of “corridors” and “islands”, centered on high-speed rail lines and sites. Additionally, the opening of high-speed railways has improved, to a certain extent, the inter-regional accessibility balance, and increased accessibility from high-speed railway sites to non-site cities. Spatial equality was also analyzed using the accessibility coefficient and social demand index. In conclusion, studying accessibility and spatial equity plays an important role in the rational planning of urban land resources and transportation.
50

Aryal, Neeraj K., Anjana Sundarrajan, Scott Boiko, David Jenkins, Huayang Liu, Miika Ahdesmaki, Aurelie Bornot, et al. "Elongator Complex Regulates MCL1 Dependency Via IRE1-XBP1 Axis of the ER Stress Response Pathway in Multiple Myeloma." Blood 138, Supplement 1 (November 5, 2021): 2275. http://dx.doi.org/10.1182/blood-2021-151194.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Evasion of apoptosis is a hallmark of cancer wherein overexpression and amplification of pro-survival BCL2-family genes like MCL1 is a common observation. MCL1 is frequently amplified in many hematological cancers like Multiple Myeloma (MM) that depend on it for survival. BH3 mimetic drugs, like the BCL2-specific inhibitor Venetoclax, have been successfully used in the clinic to treat certain cancers, and MCL1-selective inhibitors are currently in clinical development. While inhibition of MCL1 displays promising preclinical activity, many cancer models display acquired or intrinsic resistance to MCL1 inhibitors (MCL1i). As MCL1-targeted therapies progress clinically, understanding mechanisms that lead to resistance will be important to not only identify therapeutically-exploitable targets to combat resistance, but to also determine if these biomarkers could stratify patients most likely to respond to an MCL1i. Here, we used a genome-wide CRISPR knock-out screen to identify mechanisms of resistance to MCL1i AZD5991 in two MM cell lines, KMS11 and KMS34. We used a sgRNA library consisting of about 118,000 sgRNAs (~6 sgRNAs/gene), and treated the cells with DMSO or 1uM AZD5991 for 16 days (5 doublings). We identified 316 genes in KMS11 and 184 genes in KMS34 with &gt;4-fold enrichment of sgRNAs; and 221 genes with &gt;2-fold enrichment of sgRNAs in both cell lines. The sgRNAs targeting BAK and BAX were the most enriched overlapping hits. Using GSEA analysis of the 221 common genes with enriched sgRNAs, we discovered that the tRNA wobble uridine modification as the most enriched pathway. The tRNA U34 mcm5s2 modification is catalyzed by the elongator complex ELP1-6 and cytosolic thiouridylase CTU1/2. Each subunit of the elongator complex is essential for its function and loss of any subunit results in destabilization of the complex. By knocking out ELP4 in five MM cell lines (KMS11, KMS34, KMS12-PE, MM.1S, and RPMI-8226), we first validated the destabilization of the complex by showing a robust decrease in the protein levels of ELP1 and ELP3 via western blot. As the elongator complex has additional functions, we also knocked-out tRNA U34 modification pathway specific CTU1 in KMS11, KMS34, and KMS12-PE cells. We showed that genetic knock-out of ELP4 and CTU1 results in increased resistance to MCL1i in all cell lines tested. We observed the highest increase in MCL1i resistance upon ELP4-KO in KMS11 and RPMI-8226 cell lines. To understand the mechanism behind elongator complex mediated regulation of MCL1 dependency, we performed RNAseq and global proteomics in KMS11 cells (Parental, non-targeting control [NTC], ELP4-KO and CTU1-KO) and RPMI-8226 cells (Parental, NTC, and ELP4-KO). We show that the elongator complex is a regulator of IRE1-XBP1 axis of the ER stress response pathway; and knockout of IRE1 also results in MCL1i-resistance in KMS11 and RPMI8226 cell lines. Mechanistically, we show that loss of elongator complex-mediated downregulation of IRE1-XBP1 axis leads to stabilization of MCL1 and upregulation of BCL-XL and NOXA expression. We further show that upon treatment with MCL1i, KMS11-ELP4-KO cells have less disruption of MCL1:Bim complex and an increase in BCL-XL:Bim complex as compared with KMS11-NTC cells. The net increase in pro-survival MCL1 and BCL-XL proteins in ELP4-KO cells resulting in lower levels of unsequestered BIM upon AZD5991 treatment suggests a reduction in apoptotic priming. The mechanistic link between the elongator complex and ER stress response pathway led us to test ER stress inducing drugs in these cell lines. We observed that ELP4-KO results in increased resistance to proteasome inhibitor Bortezomib and other ER stress inducers like Tunicamycin, Thapsigargin, and BrefeldinA as a monotherapy or in combination with AZD5991. These data are consistent with our hypothesis that ELP4-KO cells have reduced apoptotic priming and are thus multi-drug resistant. As bortezomib is used in the clinic to treat MM patients, we asked if an elongator gene signature could be used to predict response to current therapies. We show that the elongator complex components could be used as a gene signature to stratify overall survival in MM patients (MMRF CoMMpass dataset). Moreover, ER stress response gene signature has been shown to be repressed in drug-resistant MM. Taken together, an integrated elongator and IRE-XBP1 gene signature could be a strong predictor of therapy response in MM . Disclosures Aryal: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Sundarrajan: AstraZeneca: Ended employment in the past 24 months. Boiko: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Jenkins: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Liu: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Ahdesmaki: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Bornot: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Jarnuczak: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Miele: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. McDermott: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Fawell: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Drew: AstraZeneca: Current Employment, Current equity holder in publicly-traded company. Boise: AbbVie/Genentech: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Honoraria, Research Funding. Cidado: AstraZeneca: Current Employment, Current equity holder in publicly-traded company.

To the bibliography