68 results on '"Reddy, E. Premkumar"'
Search Results
2. Drugging the 'undruggable' cancer targets
- Author
-
Dang, Chi V., Reddy, E. Premkumar, Shokat, Kevan M., and Soucek, Laura
- Abstract
In this Viewpoint article, we asked four scientists working to target important, but so-called 'undruggable', proteins in cancer for their opinions on the most crucial advances, as well as the challenges and what the future holds for this important area of cancer research.
- Published
- 2017
- Full Text
- View/download PDF
3. Myb Permits Multilineage Airway Epithelial Cell Differentiation
- Author
-
Pan, Jie-Hong, Adair-Kirk, Tracy L., Patel, Anand C., Huang, Tao, Yozamp, Nicholas S., Xu, Jian, Reddy, E. Premkumar, Byers, Derek E., Pierce, Richard A., Holtzman, Michael J., and Brody, Steven L.
- Abstract
The epithelium of the pulmonary airway is specially differentiated to provide defense against environmental insults, but also subject to dysregulated differentiation that results in lung disease. The current paradigm for airway epithelial differentiation is a one‐step program whereby a p63+basal epithelial progenitor cell generates a ciliated or secretory cell lineage, but the cue for this transition and whether there are intermediate steps are poorly defined. Here, we identify transcription factor Myb as a key regulator that permits early multilineage differentiation of airway epithelial cells. Myb+cells were identified as p63−and therefore distinct from basal progenitor cells, but were still negative for markers of differentiation. Myb RNAi treatment of primary‐culture airway epithelial cells and Mybgene deletion in mice resulted in a p63−population with failed maturation of Foxj1+ciliated cells as well as Scbg1a1+and Muc5ac+secretory cells. Consistent with these findings, analysis of whole genome expression of Myb‐deficient cells identified Myb‐dependent programs for ciliated and secretory cell differentiation. Myb+cells were rare in human airways but were increased in regions of ciliated cells and mucous cell hyperplasia in samples from subjects with chronic obstructive pulmonary disease. Together, the results show that a p63−Myb+population of airway epithelial cells represents a distinct intermediate stage of differentiation that is required under normal conditions and may be heightened in airway disease. StemCells2014;32:3245–3256
- Published
- 2014
- Full Text
- View/download PDF
4. CDK4: A Key Player in the Cell Cycle, Development, and Cancer
- Author
-
Baker, Stacey J. and Reddy, E. Premkumar
- Abstract
The cell cycle is regulated in part by cyclins and their associated serine/threonine cyclin-dependent kinases, or CDKs. CDK4, in conjunction with the D-type cyclins, mediates progression through the G1phase when the cell prepares to initiate DNA synthesis. Although CDK4-null mutant mice are viable and cell proliferation is not significantly affected in vitrodue to compensatory roles played by other CDKs, this gene plays a key role in mammalian development and cancer. This review discusses the role that CDK4 plays in cell cycle control, normal development, and tumorigenesis as well as how small molecule inhibitors of CDK4 can be used to treat disease.
- Published
- 2012
- Full Text
- View/download PDF
5. An In Vivo Functional Screen Uncovers miR-150-Mediated Regulation of Hematopoietic Injury Response
- Author
-
Adams, Brian D., Guo, Shangqin, Bai, Haitao, Guo, Yanwen, Megyola, Cynthia M., Cheng, Jijun, Heydari, Kartoosh, Xiao, Changchun, Reddy, E. Premkumar, and Lu, Jun
- Abstract
Hematopoietic stem and progenitor cells are often undesired targets of chemotherapies, leading to hematopoietic suppression requiring careful clinical management. Whether microRNAs control hematopoietic injury response is largely unknown. We report an in vivo gain-of-function screen and the identification of miR-150 as an inhibitor of hematopoietic recovery upon 5-fluorouracil-induced injury. Utilizing a bone marrow transplant model with a barcoded microRNA library, we screened for barcode abundance in peripheral blood of recipient mice before and after 5-fluorouracil treatment. Overexpression of screen-candidate miR-150 resulted in significantly slowed recovery rates across major blood lineages, with associated impairment of bone marrow clonogenic potential. Conversely, platelets and myeloid cells from miR-150 null marrow recovered faster after 5-fluorouracil treatment. Heterozygous knockout of c-myb, a conserved target of miR-150, partially phenocopied miR-150-forced expression. Our data highlight the role of microRNAs in controlling hematopoietic injury response and demonstrate the power of in vivo functional screens for studying microRNAs in normal tissue physiology.
- Published
- 2012
- Full Text
- View/download PDF
6. Impaired adult myeloid progenitor CMP and GMP cell function in conditional c-myb-knockout mice
- Author
-
Lieu, Yen K. and Reddy, E. Premkumar
- Abstract
The differentiation of myeloid progenitors to mature, terminally differentiated cells is a highly regulated process. Here, we showed that conditional disruption of the c-mybproto-oncogene in adult mice resulted in dramatic reductions in CMP, GMP and MEP myeloid progenitors, leading to a reduction of neutrophils, basophils, monocytes and platelets in peripheral blood. In addition, c-mybplays a critical role at multiple stages of myeloid development, from multipotent CMP and bipotent GMP to unipotent CFU-G and CFU-M progenitor cells. c-mybcontrols the differentiation of these cells and is required for the proper commitment, maturation and normal differentiation of CMPs and GMPs. Specifically, c-mybregulates the precise commitment to the megakaryocytic and granulo-monocytic pathways and governs the granulocytic-monocytic lineage choice. c-mybis also required for the commitment along the granulocytic pathway for early myeloid progenitor cells and for the maturation of committed precursor cells along this pathway. On the other hand, disruption of the c-mybgene favors the commitment to the monocytic lineage, although monocytic development was abnormal with cells appearing more mature with atypical CD41 surface markers. These results demonstrate that c-mybplays a pivotal role in the regulation of multiple stages in adult myelogenesis.
- Published
- 2012
- Full Text
- View/download PDF
7. The Ins and Outs of Bcr-Abl Inhibition
- Author
-
Reddy, E. Premkumar and Aggarwal, Aneel K.
- Abstract
The development of inhibitors against Abl has changed the landscape for the treatment of chronic myelogenous leukemia (CML) and cancer in general. Beginning with the monumental discovery and approval of imatinib for CML, a second generation of inhibitors, nilotinib and dasatinib, has now gained approval for the treatment of CML. Notably, these second-generation inhibitors are active against many of the mutations in the Abl kinase that confer resistance to imatinib. However, resistance remains a major problem, and new inhibitors such as ponatinib and GNF2/GNF5 have been developed, with activity towards the common gatekeeper T315I mutation. We review here the mechanisms of Abl inhibition with an emphasis on structural elements that are important for the selectivity and design of new molecules. In particular, we focus on how changes in the conformation of the P-loop, the activation loop, the DFG motif, and other structural elements of Abl have been instrumental in developing an understanding of inhibitor binding.
- Published
- 2012
- Full Text
- View/download PDF
8. JAK/STAT Pathways in Cytokine Signaling and Myeloproliferative Disorders: Approaches for Targeted Therapies
- Author
-
Jatiani, Shashidhar S., Baker, Stacey J., Silverman, Lewis R., and Reddy, E. Premkumar
- Abstract
Hematopoiesis is the cumulative result of intricately regulated signaling pathways that are mediated by cytokines and their receptors. Studies conducted over the past 10 to 15 years have revealed that hematopoietic cytokine receptor signaling is largely mediated by a family of tyrosine kinases termed Janus kinases (JAKs) and their downstream transcription factors, termed STATs (signal transducers and activators of transcription). Aberrations in these pathways, such as those caused by the recently identified JAK2V617Fmutation and translocations of the JAK2 gene, are underlying causes of leukemias and other myeloproliferative disorders. This review discusses the role of JAK/STAT signaling in normal hematopoiesis as well as genetic abnormalities associated with myeloproliferative and myelodisplastic syndromes. This review also summarizes the status of several small molecule JAK2 inhibitors that are currently at various stages of clinical development. Several of these compounds appear to improve the quality of life of patients with myeloproliferative disorders by palliation of disease-related symptoms. However, to date, these agents do not seem to significantly affect bone marrow fibrosis, alter marrow histopathology, reverse cytopenias, reduce red cell transfusion requirements, or significantly reduce allele burden. These results suggest the possibility that additional mutational events might be associated with the development of these neoplasms, and indicate the need for combination therapies as the nature and significance of these additional molecular events is better understood.
- Published
- 2010
- Full Text
- View/download PDF
9. Cooperativity of Cdk4R24Cand Rasin melanoma development
- Author
-
Chawla, Rachna, Procknow, Judith A, Tantravahi, Ramana V, Khurana, Jasbir, Litvin, Judith, and Reddy, E. Premkumar
- Abstract
The importance of the CDK4 protein in human cancer first became evident following the identification of a germ line mutation in the Cdk4locus that predisposes humans to melanoma. This mutation results in substitution of arginine with cysteine at position 24 (R24C). In an earlier study, we introduced the R24C mutation into the Cdk4 locus of mice using Cre-loxP-mediated "knock-in" technology and observed a very low incidence of spontaneous melanomas in Cdk4R24C/R24C mice. This suggested that additional oncogenic mutations might be required for development of melanomas. Here we report an increased incidence of spontaneous cutaneous melanoma in mice expressing the oncogene HRAS(G12V) in melanocytes on a Cdk4R24Cbackground. Treatment of Tyr-HRas:Cdk4R24C/R24C mice with the carcinogen, DMBA/TPA resulted in a further increase in the number of nevi and melanomas developed when compared with Tyr-HRas:Cdk4+/+ mice. In summary, in Tyr-HRas:Cdk4R24C/R24Cmice, we observed that activated CDK4 cooperates with the oncogenic HRAS(G12V)protein to increase the susceptibility of melanoma development in vivo.
- Published
- 2010
- Full Text
- View/download PDF
10. Increased angiogenesis inCdk4R24C/R24C:Apc+/Minintestinal tumors
- Author
-
Abedin, Zahidur R., Ma, Zhongjie, and Reddy, E. Premkumar
- Abstract
Cyclin dependent kinase 4 (Cdk4) is a cell cycle regulator involved in early G1 cell cycle progression and has been indirectly implicated in angiogenesis in the Min mouse system, a mouse that harbors a mutation in the Apc gene. Apc+/Minmice when crossed with Ink4a/arf-/-mice, exhibited increased angiogenesis of colorectal tumors suggesting that dysregulation of Cdk4 (due to loss of Ink4a-mediated suppression) may contribute to enhanced angiogenesis. To demonstrate a direct role for Cdk4 in angiogenesis, we crossed mice that have an activated Cdk4, Cdk4R24C/R24Cmice, with Apc+/Minmice and examined levels of angiogenesis in intestinal tumors formed. Our results show an increase in the percentage of highly vascularized tumors in Cdk4R24C/R24C:ApcMin/+and Cdk4+/R24C:ApcMin/+mice compared to Cdk4+/+:ApcMin/+mice. In addition immunohistochemical analysis showed an increase in CD-31 staining localized to endothelial cells of Cdk4R24C/R24C:ApcMin/+mouse tumors, supporting the hypothesis of increased vasculature in these tumors. Further analysis showed an increase in the expression of the E2F1 target proteins Vegf-b and Cyclin A in Cdk4R24C/R24C:Apc+/Min intestinal tumors. Together these data suggest that the dysregulated Cdk4 gene plays an important role in angiogenesis during intestinal tumor formation and may in part act via increasing E2F1 target proteins. This is the first report to show that Cdk4 has a direct role in angiogenesis in vivo and may be an important drug target to reduce or prevent angiogenesis during intestinal tumor formation.
- Published
- 2010
- Full Text
- View/download PDF
11. A Non–ATP-Competitive Dual Inhibitor of JAK2V617Fand BCR-ABLT315IKinases: Elucidation of a Novel Therapeutic Spectrum Based on Substrate Competitive Inhibition
- Author
-
Jatiani, Shashidhar S., Cosenza, Stephen C., Reddy, M.V. Ramana, Ha, Ji Hee, Baker, Stacey J., Samanta, Ajoy K., Olnes, Matthew J., Pfannes, Loretta, Sloand, Elaine M., Arlinghaus, Ralph B., and Reddy, E. Premkumar
- Abstract
Here we report the discovery of ON044580, an a-benzoyl styryl benzyl sulfide that possesses potent inhibitory activity against two unrelated kinases, JAK2 and BCR-ABL, and exhibits cytotoxicity to human tumor cells derived from chronic myelogenous leukemia (CML) and myelodysplasia (MDS) patients or cells harboring a mutant JAK2 kinase. This novel spectrum of activity is explained by the non–ATP-competitive inhibition of JAK2 and BCR-ABL kinases. ON044580 inhibits mutant JAK2 kinase and the proliferation of JAK2V617F-positive leukemic cells and blocks the IL-3–mediated phosphorylation of JAK2 and STAT5. Interestingly, this compound also directly inhibits the kinase activity of both wild-type and imatinib-resistant (T315I) forms of the BCR-ABL kinase. Finally, ON044580 effectively induces apoptosis of imatinib-resistant CML patient cells. The apparently unrelated JAK2 and BCR-ABL kinases share a common substrate, STAT5, and such substrate competitive inhibitors represent an alternative therapeutic strategy for development of new inhibitors. The novel mechanism of kinase inhibition exhibited by ON044580 renders it effective against mutant forms of kinases such as the BCR-ABLT315Iand JAK2V617F. Importantly, ON044580 selectively reduces the number of aneuploid cells in primary bone marrow samples from monosomy 7 MDS patients, suggesting another regulatory cascade amenable to this agent in these aberrant cells. Data presented suggest that this compound could have multiple therapeutic applications including monosomy 7 MDS, imatinib-resistant CML, and myeloproliferative neoplasms that develop resistance to ATP-competitive agents.
- Published
- 2010
- Full Text
- View/download PDF
12. Destabilization of Bcr-Abl/Jak2 Network by a Jak2/Abl Kinase Inhibitor ON044580 Overcomes Drug Resistance in Blast Crisis Chronic Myelogenous Leukemia (CML)
- Author
-
Samanta, Ajoy K., Chakraborty, Sandip N., Wang, Yan, Schlette, Ellen, Reddy, E. Premkumar, and Arlinghaus, Ralph B.
- Abstract
Bcr-Abl is the predominant therapeutic target in chronic myeloid leukemia (CML), and tyrosine kinase inhibitors (TKIs) that inhibit Bcr-Abl have been successful in treating CML. With progression of CML disease especially in blast crisis stage, cells from CML patients become resistant to imatinib mesylate (IM) and other TKIs, resulting in relapse. Because Bcr-Abl is known to drive multiple signaling pathways, the study of the regulation of stability of Bcr-Abl in IM-resistant CML cells is a critical issue as a possible therapeutic strategy. Here, we report that a new dual-kinase chemical inhibitor, ON044580, induced apoptosis of Bcr-Abl+ IM-sensitive, IM-resistant cells, including the gatekeeper Bcr-Abl mutant, T315I, and also cells from blast crisis patients. In addition, IM-resistant K562-R cells, cells from blast crisis CML patients, and all IM-resistant cell lines tested had reduced ability to form colonies in soft agar in the presence of 0.5 µM ON044580. In in vitrokinase assays, ON044580 inhibited the recombinant Jak2 and Abl kinase activities when the respective Jak2 and Abl peptides were used as substrates. Incubation of the Bcr-Abl+ cells with ON044580 rapidly reduced the levels of the Bcr-Abl protein and also reduced the expression of HSP90 and its client protein levels. Lysates of Bcr-Abl+ cell lines were found to contain a large signaling network complex composed of Bcr-Abl, Jak2, HSP90, and its client proteins as detected by a gel filtration column chromatography, which was rapidly disrupted by ON044580. Therefore, targeting Jak2 and Bcr-Abl kinases is an effective way to destabilize Bcr-Abl and its network complex, which leads to the onset of apoptosis in IM-sensitive and IM-resistant Bcr-Abl+ cells. This inhibitory strategy has potential to manage all types of drug-resistant CML cells, especially at the terminal blast crisis stage of CML, where TKIs are not clinically useful.
- Published
- 2010
- Full Text
- View/download PDF
13. Requirement of Cdk4 for v-Ha-ras–Induced Breast Tumorigenesis and Activation of the v-ras–Induced Senescence Program by the R24C Mutation
- Author
-
Reddy, Haritha K.D.L., Graña, Xavier, Dhanasekaran, Danny N., Litvin, Judith, and Reddy, E. Premkumar
- Abstract
Activating mutations in CDK4 and inactivation of its key kinase inhibitor, p16INK4A, have been implicated in the genesis and progression of human cancer. Previous work has demonstrated that CDK4 expression is required for Neu-induced but not Wnt-induced breast tumorigenesis in mice. However, the role that CDK4 plays in ras-mediated breast tumor development is not well defined. To gain an understanding of the role of Cdk4 in ras-induced breast tumorigenesis, MMTV-v-Ha-rastransgenic mice were bred with Cdk4(+/neo)and Cdk4(R24C/R24C)mice to generate Cdk4(neo/neo):MMTV-v-Ha-ras, Cdk4(+/+):MMTV-v-Ha-ras, and Cdk4(R24C/R24C):MMTV-v-Ha-rasmice. The studies presented here demonstrate that Cdk4 expression is essential for Ras-mediated breast tumorigenesis. Surprisingly, the results also show that coexpression of mutant rasand Cdk4R24Cgenes in breast epithelial cells leads to an activation of senescent pathways that delay tumorigenesis. Analysis of the phosphorylated form of H2AX, a marker for DNA damage, indicated its increased presence in the tumors of Cdk4(R24C/R24C):MMTV-v-Ha-rasmice. These observations indicate that the increased apoptosis and senescence seen in breast tumors of these mice might be due to increased DNA damage response in cells expressing activated forms of rasand Cdk4(R24C).
- Published
- 2010
- Full Text
- View/download PDF
14. Rigosertib in Combination with Azacitidine Impacts Metabolic and Differentiation Pathways in the MDS-L Cell Line
- Author
-
Rai, Richa, Patel, Foramben, Melana, Stella, Feld, Jonathan, Navada, Shyamala C., Odchimar-Reissig, Rosalie, Demakos, Erin P., Reddy, E. Premkumar, and Silverman, Lewis R.
- Abstract
BackgroundMyelodysplastic Syndrome (MDS) is characterized by ineffective clonal hematopoiesis with peripheral blood cytopenias, leading to death from infection or bleeding. Azacitidine (AZA), a hypomethylating agent (HMA) is the standard of care for treatment of MDS patients (pts) with higher-risk MDS [Silverman LR, The Myelodysplastic Syndrome in Cancer Medicine, Editors: R.J. Bast, et al. 2017]. Responses to AZA occur in 50% of pts with significant effects on hematopoiesis ranging from improvement in a single lineage to complete restoration of blood counts and transfusion independence [Silverman LR, et al. Leukemia, 1993]. AZA treatment is associated with global DNA hypomethylation, including human endogenous retroviruses (HEV) which further activates innate immune signaling [Chiappinelli KB, et al.Cell, 2015]. The exact mechanism by which AZA improves hematopoiesis is unknown. AZA improves overall survival of pts, yet despite this, 100% of pts ultimately fail treatment with worsening cytopenias or transformation to leukemia [Silverman LR, et al. B. J Clin Oncol, 2002; Cancer, 2011]. Thus, understanding the mechanism of resistance and identification of targets which can reverse HMA failure and improve hematopoiesis in MDS pts is critical. Our clinical data demonstrate that AZA combined with Rigosertib (RIGO), a novel Ras mimetic that inhibits Ras/Raf signaling [Athuluri-Divakar SK, et al.Cell, 2016], yields a response rate of 54% of pts who were HMA failures [Navada SC, et al. EHA 2019]. The response was associated with significant improvement in hematopoiesis and represents a critical observation in overcoming the epigenetic clinical resistance phenotype. The precise mechanism that leads to reversal of the resistance phenotype is poorly understood.
- Published
- 2020
- Full Text
- View/download PDF
15. Inhibition of tumor angiogenesis in vivo by a monoclonal antibody targeted to domain 5 of high molecular weight kininogen
- Author
-
Song, James S., Sainz, Irma M., Cosenza, Stephen C., Isordia-Salas, Irma, Bior, Abdel, Bradford, Harlan N., Guo, Yan-Lin, Pixley, Robin A., Reddy, E. Premkumar, and Colman, Robert W.
- Abstract
We have shown that human high molecular weight kininogen is proangiogenic due to release of bradykinin. We now determined the ability of a murine monoclonal antibody to the light chain of high molecular weight kininogen, C11C1, to inhibit tumor growth compared to isotype-matched murine IgG. Monoclonal antibody C11C1 efficiently blocks binding of high molecular weight kininogen to endothelial cells in a concentration-dependent manner. The antibody significantly inhibited growth of human colon carcinoma cells in a nude mouse xenograft assay and was accompanied by a significant reduction in the mean microvascular density compared to the IgG control group. We also showed that a hybridoma producing monoclonal antibody C11C1 injected intramuscularly exhibited markedly smaller tumor mass in a syngeneic host compared to a hybridoma producing a monoclonal antibody to the high molecular weight kininogen heavy chain or to an unrelated plasma protein. In addition, tumor inhibition by purified monoclonal antibody C11C1 was not due to direct antitumor effect because there was no decrease of tumor cell growth in vitro in contrast to the in vivo inhibition. Our results indicate that monoclonal antibody C11C1 inhibits angiogenesis and human tumor cell growth in vivo and has therapeutic potential for treatment of human cancer. (Blood. 2004;104:2065-2072)
- Published
- 2004
- Full Text
- View/download PDF
16. Mechanisms associated with IL-6–induced up-regulation of Jak3 and its role in monocytic differentiation
- Author
-
Mangan, James K., Rane, Sushil G., Kang, Anthony D., Amanullah, Arshad, Wong, Brian C., and Reddy, E. Premkumar
- Abstract
We report here that Janus kinase 3 (Jak3) is a primary response gene for interleukin-6 (IL-6) in macrophage differentiation, and ectopic overexpression of Jak3 accelerates monocytic differentiation of normal mouse bone marrow cells stimulated with cytokines. Furthermore, we show that incubation of normal mouse bone marrow cells with a JAK3-specific inhibitor results in profound inhibition of myeloid colony formation in response to granulocyte-macrophage colony-stimulating factor or the combination of stem cell factor, IL-3, and IL-6. In addition, mutagenesis of the Jak3 promoter has revealed that Sp1 binding sites within a -67 to -85 element and a signal transducer and activator of transcription (Stat) binding site at position -44 to -53 are critical for activation of Jak3 transcription in murine M1 myeloid leukemia cells stimulated with IL-6. Electrophoretic mobility shift assay (EMSA) analysis has demonstrated that Sp1 can bind to the -67 to -85 element and Stat3 can bind to the -44 to -53 STAT site in IL-6-stimulated M1 cells. Additionally, ectopic overexpression of Stat3 enhanced Jak3 promoter activity in M1 cells. This mechanism of activation of the murine Jak3 promoter in myeloid cells is distinct from a recently reported mechanism of activation of the human JAK3 promoter in activated T cells.
- Published
- 2004
- Full Text
- View/download PDF
17. Mechanisms associated with IL-6–induced up-regulation of Jak3and its role in monocytic differentiation
- Author
-
Mangan, James K., Rane, Sushil G., Kang, Anthony D., Amanullah, Arshad, Wong, Brian C., and Reddy, E. Premkumar
- Abstract
We report here that Janus kinase 3 (Jak3)is a primary response gene for interleukin-6 (IL-6) in macrophage differentiation, and ectopic overexpression of Jak3 accelerates monocytic differentiation of normal mouse bone marrow cells stimulated with cytokines. Furthermore, we show that incubation of normal mouse bone marrow cells with a JAK3-specific inhibitor results in profound inhibition of myeloid colony formation in response to granulocyte-macrophage colony-stimulating factor or the combination of stem cell factor, IL-3, and IL-6. In addition, mutagenesis of the Jak3promoter has revealed that Sp1 binding sites within a -67 to -85 element and a signal transducer and activator of transcription (Stat) binding site at position -44 to -53 are critical for activation of Jak3transcription in murine M1 myeloid leukemia cells stimulated with IL-6. Electrophoretic mobility shift assay (EMSA) analysis has demonstrated that Sp1 can bind to the -67 to -85 element and Stat3 can bind to the -44 to -53 STAT site in IL-6-stimulated M1 cells. Additionally, ectopic overexpression of Stat3 enhanced Jak3promoter activity in M1 cells. This mechanism of activation of the murine Jak3promoter in myeloid cells is distinct from a recently reported mechanism of activation of the human JAK3promoter in activated T cells.
- Published
- 2004
- Full Text
- View/download PDF
18. Payoff of Molecular Oncology Research
- Author
-
Reddy, E. Premkumar
- Abstract
NA
- Published
- 2003
- Full Text
- View/download PDF
19. Activation of the Jak3 pathway is associated with granulocytic differentiation of myeloid precursor cells
- Author
-
Rane, Sushil G., Mangan, James K., Amanullah, Arshad, Wong, Brian C., Vora, Renu K., Liebermann, Dan A., Hoffman, Barbara, Graña, Xavier, and Reddy, E. Premkumar
- Abstract
Jak3, a member of the Janus kinase family of cytoplasmic tyrosine kinases, is expressed at low levels in immature hematopoietic cells and its expression is dramatically up-regulated during the terminal differentiation of these cells. To better understand the role of Jak3 in myeloid cell development, we have investigated the role of Jak3 in myeloid cell differentiation using the 32Dcl3 cell system. Our studies show that Jak3 is a primary response gene for granulocyte colony-stimulating factor (G-CSF) and the accumulation of tyrosine phosphorylated Jak3 correlated with cell growth inhibition and terminal granulocytic differentiation in response to G-CSF. Ectopic overexpression of Jak3 in 32Dcl3 cells resulted in an acceleration of the G-CSF–induced differentiation program that was preceded by G1 cell cycle arrest, which was associated with the up-regulation of the cyclin-dependent kinase inhibitor p27Kip1 and down-regulation of Cdk2, Cdk4, Cdk6, and Cyclin E. In addition, ectopic overexpression of Jak3 appears to result in the inactivation of PKB/Akt and Stat3-mediated proliferative pathways in the presence of G-CSF. Similarly, overexpression of Jak3 in primary bone marrow cells resulted in an acceleration of granulocytic differentiation in the presence of granulocyte-macrophage colony-stimulating factor, which was associated with their growth arrest in the G1 phase of the cell cycle. Taken together, these results indicate that Jak3-mediated signals play an important role in myeloid cell differentiation.
- Published
- 2002
- Full Text
- View/download PDF
20. Germ Line Transmission of the Cdk4R24CMutation Facilitates Tumorigenesis and Escape from Cellular Senescence
- Author
-
Rane, Sushil G., Cosenza, Stephen C., Mettus, Richard V., and Reddy, E. Premkumar
- Abstract
ABSTRACTMutations in CDK4 and its key kinase inhibitor p16INK4ahave been implicated in the genesis and progression of familial human melanoma. The importance of the CDK4locus in human cancer first became evident following the identification of a germ line CDK4-Arg24Cys (R24C) mutation, which abolishes the ability of CDK4 to bind to p16INK4a. To determine the role of the Cdk4R24Cgerm line mutation in the genesis of other cancer types, we introduced the R24C mutation in the Cdk4locus of mice by using Cre-loxP-mediated “knock-in” technology. Cdk4R24C/R24Cmouse embryo fibroblasts (MEFs) displayed increased Cdk4 kinase activity resulting in hyperphosphorylation of all three members of the Rb family, pRb, p107, and p130. MEFs derived from Cdk4R24C/R24Cmice displayed decreased doubling times, escape from replicative senescence, and escape sensitivity to contact-induced growth arrest. These MEFs also exhibited a high degree of susceptibility to oncogene-induced transformation, suggesting that the Cdk4R24Cmutation can serve as a primary event in the progression towards a fully transformed phenotype. In agreement with the in vitro data, homozygous Cdk4R24C/R24Cmice developed tumors of various etiology within 8 to 10 months of their life span. The majority of these tumors were found in the pancreas, pituitary, brain, mammary tissue, and skin. In addition, Cdk4R24C/R24Cmice showed extraordinary susceptibility to carcinogens and developed papillomas within the first 8 to 10 weeks following cutaneous application of the carcinogens 9,10-di-methyl-1,2-benz[a]anthracene (DMBA) and 12-O-tetradecanoylphorbol-13-acetate (TPA). This report formally establishes that the activation of Cdk4 is sufficient to promote cancer in many tissues. The observation that a wide variety of tumors develop in mice harboring the Cdk4R24Cmutation offers a genetic proof that Cdk4activation may constitute a central event in the genesis of many types of cancers in addition to melanoma.
- Published
- 2002
- Full Text
- View/download PDF
21. Combination of Ras Modulator and Azacitidine Impacts Innate Immune Signaling Pathway in MDS-L Cell Line
- Author
-
Rai, Richa, Patel, Foramben, Feld, Jonathan, Melana, Stella, Navada, Shyamala C., Odchimar-Reissig, Rosalie, Demakos, Erin P., Reddy, E. Premkumar, and Silverman, Lewis R.
- Abstract
Background:Myelodysplastic syndrome (MDS) is a clinically heterogenous disease of hematopoietic stem cells (HSC) characterized by ineffective hematopoiesis, uni/multi-lineage dysplasia and a high tendency to transform into acute myeloid leukemia. Aberrant chromosomal and genetic lesions contribute to MDS pathogenesis which has been associated with chronic activation of the innate immune response and a hyperinflammatory microenvironment (Barryero L, et al.Blood, 2018). Dysfunction of Toll like receptors (TLR) and downstream effectors has been associated with the loss of progenitor function and differentiation of bone marrow (BM) cells in MDS patients. Azacitidine (AZA), a hypomethylating agent (HMA), is the mainstay of therapy for patients with higher-risk MDS (Silverman LR, The Myelodysplastic Syndrome in Cancer Medicine, Editors: R.J. Bast, et al. 2017) and carries an overall response rate (ORR) of 50% in patients with significant effects on hematopoiesis, ranging from improvement in a single lineage to complete restoration of blood counts and transfusion independence with survival benefits (Silverman LR, et al., Leukemia, 1993). The response to AZA is not durable and all patients relapse with worsening bone marrow failure.
- Published
- 2021
- Full Text
- View/download PDF
22. Differential regulation of the retinoblastoma family of proteins during cell proliferation and differentiation
- Author
-
GARRIGA, Judit, LIMÓN, Ana, MAYOL, Xavier, RANE, Sushil G., ALBRECHT, Jeffrey H., REDDY, E. Premkumar, ANDRÉS, Vicente, and GRAÑA, Xavier
- Abstract
In the present study we have analysed the regulation of pocket protein expression and post-transcriptional modifications on cell proliferation and differentiation, both in vivo and in vitro. There are marked changes in pocket protein levels during these transitions, the most striking differences being observed between p130 and p107. The mechanisms responsible for regulating pocket protein levels seem to be dependent on both cell type and pocket protein, in addition to their dependence on the cell growth status. Changes in retinoblastoma protein and p107 levels are independent of their state of phosphorylation. However, whereas p130 phosphorylation to forms characteristic of quiescent/differentiated cells results in the accumulation of p130 protein, phosphorylation of p130 to one or more forms characteristic of cycling cells is accompanied by down-regulation of its protein levels. We also show here that the phosphorylation status and protein levels of p130 and p107 are regulated in vivo as in cultured cells. In vivo, changes in p130 forms are correlated with changes in E2F complexes. Moreover, the modulation of p130 and p107 status during cell differentiation in vitro is consistent with the patterns of protein expression and phosphorylation status found in mouse tissues. Thus in addition to the direct disruption of pocket protein/E2F complexes induced by cyclin/cyclin-dependent kinase, the results we report here indicate that the differential modulation of pocket protein levels constitutes a major mechanism that regulates the pool of each pocket protein that is accessible to E2F and/or other transcription factors.
- Published
- 1998
- Full Text
- View/download PDF
23. Cellular genes analogous to retroviral onc genes are transcribed in human tumour cells
- Author
-
Eva, Alessandra, Robbins, Keith C., Andersen, Philip R., Srinivasan, Alagarsamy, Tronick, Steven R., Reddy, E. Premkumar, Ellmore, Nelson W., Galen, Angela T., Lautenberger, James A., Papas, Takis S., Westin, Eric H., Wong-Staal, Flossie, Gallo, Robert C., and Aaronson, Stuart A.
- Abstract
Polyadenylated RNAs of certain human tumour cell lines are shown to contain transcripts related to the cell-derived transforming onc genes of molecularly cloned primate, murine or avian transforming retrovirus genomes. Thus, analogues of retroviral transforming genes are both present and frequently expressed in human neoplastic cells.
- Published
- 1982
- Full Text
- View/download PDF
24. Cellular Factors Binding to a Novel cis-Acting Element Mediate Steroid Hormone Responsiveness of Mouse Mammary Tumor Virus Promoter (∗)
- Author
-
Lee, Kyong-Il, Reddy, E. Premkumar, and Reddy, C. Damodara
- Abstract
Steroid hormone receptors regulate mouse mammary tumor virus (MMTV) gene expression by binding to hormone response DNA elements present in the long terminal repeat. Tissue-specific expression of MMTV is unlikely to be regulated by steroid hormone-receptor complex alone, and mammary cell-specific factors might play a role in the hormone-induced transcriptional activation. In this report we have investigated the function of a novel cis-acting element designated Kil (−204 to −188) which is located adjacent to the distal glucocorticoid response element, in steroid hormone-induced transcription of MMTV. Electrophoretic mobility shift assays indicate that cellular factors bind to the Kil element, and dexamethasone stimulation results in alterations in the binding pattern of proteins in this region. By transient transfection assays using wild type and deletion mutants of the Kil element, we show that this novel cis-acting element is necessary for hormone-induced transcription of MMTV and functions in mammary tumor cells but not in NIH/3T3 cells. Mutagenesis of the Kil sequence suggests that the entire Kil element functioning as one unit is necessary for hormone-induced transcription of MMTV. When placed in the context of heterologous promoters, neither Kil element nor glucocorticoid response element is able to induce significant hormone-induced transcription of MMTV. The presence of both the DNA elements in tandem results in optimal induction of transcription in the presence of steroid hormones. Our results also indicate that the Kil element functions in human breast carcinoma cell lines such as T47D and MCF-7. These results suggest that Kil element in combination with distal glucocorticoid response element functions as a mammary cell-specific enhancer to regulate MMTV transcription.
- Published
- 1995
- Full Text
- View/download PDF
25. Translational Products of Moloney Murine Sarcoma Virus RNA: Identification of Proteins Encoded by the Murine Sarcoma Virus srcGene
- Author
-
Cremer, Kenneth, Reddy, E. Premkumar, and Aaronson, Stuart A.
- Abstract
In vitro translation of virion RNA of Moloney murine sarcoma virus (MSV) strain 124 yielded major products having molecular weights of 63,000 (63K), 43K, 40K, 31K, and 24K daltons. A molecularly cloned subgenomic fragment of Moloney MSV comprised of the cellular insertion (src) region was utilized in hybridization arrest translation as a means of identifying products of the MSV srcgene. MSV srcDNA specifically inhibited synthesis of the 43K, 40K, 31K, and 24K proteins, implying that each of these proteins was coded within the MSV srcgene. The MSV src-specific nature of this family of proteins was further confirmed by partial purification of MSV src-containing RNAs from MSV non-producer cells. In vitro translation of enriched cellular RNAs yielded products with molecular weights identical to those of the 43K family of proteins synthesized from virion RNA. Nucleotide sequence analysis of the MSV transforming region has revealed a long open reading frame which includes five methionine codons (Reddy et al., Proc. Natl. Acad. Sci. U.S.A. 77:5234-5238, 1980). The molecular weights of the four largest proteins that could be synthesized within this open reading frame corresponded closely to the molecular weights of the 43K family of proteins. Partial cyanogen bromide cleavage of each of the three largest proteins resulted in an uncleaved fragment having a molecular weight equal to that of the smallest (24K) protein. These findings provide direct biochemical evidence that the 43K, 40K, 31K, and 24K proteins are related in their carboxy-terminal regions, as well as information concerning the MSV srcgene coding sequences from which each protein originates:
- Published
- 1981
- Full Text
- View/download PDF
26. The C-Terminal Domain of B-Myb Acts As a Positive Regulator of Transcription and Modulates Its Biological Functions
- Author
-
Oh, Il-Hoan and Reddy, E. Premkumar
- Abstract
ABSTRACTThe mybgene family consists of three members, named A-, B-, and c-myb. All three members of this family encode nuclear proteins that bind DNA in a sequence-specific manner and function as regulators of transcription. In this report, we have examined the biochemical and biological activities of murine B-myband compared these properties with those of murine c-myb. In transient transactivation assays, murine B-mybexhibited transactivation potential comparable to that of c-myb. An analysis of deletion mutants of B-myband c-mybshowed that while the C-terminal domain of c-Myb acts as a negative regulator of transcriptional transactivation, the C-terminal domain of B-Myb functions as a positive enhancer of transactivation. To compare the biological activities of c-myband B-myb, the two genes were overexpressed in 32Dcl3 cells, which are known to undergo terminal differentiation into granulocytes in the presence of granulocyte colony-stimulating factor (G-CSF). We observed that c-mybblocked the G-CSF-induced terminal differentiation of 32Dcl3 cells, resulting in their continued proliferation in the presence of G-CSF. In contrast, ectopic overexpression of B-mybblocked the ability of 32D cells to proliferate in the presence of G-CSF and accelerated the G-CSF-induced granulocytic differentiation of these cells. Similar studies with B-myb–c-mybchimeras showed that only chimeras that contained the C-terminal domain of B-Myb were able to accelerate the G-CSF-induced terminal differentiation of 32Dcl3 cells. These studies show that c-myband B-mybdo not exhibit identical biological activities and that the carboxyl-terminal regulatory domain of B-Myb plays a critical role in its biological function.
- Published
- 1998
- Full Text
- View/download PDF
27. Murine A-mybGene Encodes a Transcription Factor, Which Cooperates with Ets-2 and Exhibits Distinctive Biochemical and Biological Activities from c-myb*
- Author
-
Oh, Il-Hoan and Reddy, E. Premkumar
- Abstract
The mybgene family consists of three members, named A-, B-, and c-myb, which encode nuclear proteins that bind to DNA and function as regulators of transcription. Our results show that murine A-mybis a poor transactivator of transcription compared with murine c-myb. Deletion of the COOH-terminal domain of A-Myb, or co-expression with Ets-2 resulted in increased transactivation potential. While ectopic overexpression of c-mybin 32Dcl3 cells results in a block to the ability of these cells to undergo terminal differentiation resulting in indefinite growth in granulocyte-colony-stimulating factor (G-CSF), similar overexpression of A-mybresults in growth arrest and concomitant terminal differentiation of 32D cells into granulocytes. Co-expression of A-myband ets-2in these cells results in the restoration of the proliferative activity of the cells in G-CSF, but fails to induce a block to G-CSF-induced terminal differentiation. However, overexpression of the COOH-terminal deletion mutant of A-mybresults in a block to G-CSF-induced differentiation of 32D cells, suggesting that the distinctive biological phenotypes produced by A-myband c-mybgenes are mediated by their COOH-terminal domains.
- Published
- 1997
- Full Text
- View/download PDF
28. Acquisition of transforming properties by alternative point mutations within c-bas/has human proto-oncogene
- Author
-
Yuasa, Yasuhito, Srivastava, Shiv K., Dunn, Claire Y., Rhim, Johng S., Reddy, E. Premkumar, and Aaronson, Stuart A.
- Abstract
The transforming gene of a human lung carcinoma-derived cell line, Hs242, has been cloned in biologically active form, and identified as c-bas/has (otherwise known as c-Ha-ras). The genetic lesion responsible for the transforming activity of the Hs242 oncogene has been localized to a point mutation in the second exon which results in the substitution of leucine for glutamine as amino acid 61 of the predicted protein. No changes were observed in the first exon, the region of c-bas/has in which a point mutation is responsible for activation of the T24 and EJ bladder carcinoma oncogenes.
- Published
- 1983
- Full Text
- View/download PDF
29. Multi-Unit Anti-BCR-ABL Ribozyme Therapy in Chronic Myelogenous Leukemia
- Author
-
Leopold, Lance, Shore, Scott, and Reddy, E. Premkumar
- Abstract
In this review, we summarize and update our data on the development of a multi-unit anti-BCR/ABL ribozyme. In vitro studies comparing several anti-BCR/ABL ribozymes demonstrated that a tripleunit ribozyme is the most efficient. Detailed kinetic analysis revealed this ribozyme to have a lower Kcat, most likely due to non homologous bases at restriction enzyme sites used in ribozyme construction. Delivery of this ribozyme to a BCR/ABL transformed cell line by a novel vehicle targeting the folate receptor resulted in a 3 log reduction in BCR/ABL mRNA when analyzed by RT-PCR. This delivery strategy reversed the IL-3 independence of this cell line. Retroviral vectors containing genes coding for the multi-unit ribozyme have been constructed and their use to effect BCR/ABL transformed cell biology is discussed.
- Published
- 1996
- Full Text
- View/download PDF
30. Abrogation of Interleukin-3 Dependence of Myeloid Cells by the v-srcOncogene Requires SH2 and SH3 Domains Which Specify Activation of STATs
- Author
-
Chaturvedi, Priya, Sharma, Sunita, and Reddy, E. Premkumar
- Abstract
The v-srconcogene encodes a nonreceptor tyrosine kinase. When this gene was expressed in the myeloblastic cell line 32Dcl3, it was found to abrogate interleukin-3 (IL-3) dependence of this cell line and to block its ability to terminally differentiate into granulocytes in response to granulocyte colony-stimulating factor (GCSF). In contrast, a highly related tyrosine kinase gene, v-fgr, fails to render this cell line IL-3 independent for growth or to block its ability to undergo terminal differentiation in the presence of GCSF. The active structural domains of v-srcthat are responsible for the abrogation of IL-3 dependence of myeloid cells and the mechanisms by which v-srctransforms these cells are at present unclear. To identify the domains in v-srcwhich are responsible for this activity, we constructed several chimeric recombinants between the v-srcand the related Src family member v-fgrby replacing portions of v-srcwith corresponding domains of v-fgr. These chimeric DNAs were transfected into 32Dcl3 cells and examined for their abilities to render this cell line IL-3 independent. Our results show that only chimeras containing both the SH3 and the SH2 domains of v-srcwere capable of rendering the 32Dcl3 cell line IL-3 independent. To understand the possible mechanisms underlying the IL-3-independent growth of v-src-transformed 32Dcl3 cells, we examined the phosphorylation status of JAK-1, JAK-2, and JAK-3 kinases in the v-src- and v-fgr-transformed 32Dcl3 cells. Our results show that none of the JAK kinases are constitutively phosphorylated by v-srcor v-fgr. We then examined the phosphorylation status of the STAT (signal transducers and activators of transcription) family of transcription factors. Our results show that STAT1, STAT3, and STAT5 exist in a constitutively phosphorylated state in v-src-transformed 32Dcl3 cells, while such constitutive phosphorylation is not seen in v-fgr-transformed cell lines. Our results also show that STAT3 coimmunoprecipitates with v-Src, suggesting that the activation of STAT3 occurs due to direct association with v-Src. However, STAT1 and STAT5, which also exist in a constitutively phosphorylated state in v-src-transformed 32Dcl3 cells, do not coimmunoprecipitate with v-Src, suggesting that these proteins either interact weakly with v-Src or are phosphorylated by a mechanism distinctive from that of STAT3.
- Published
- 1997
- Full Text
- View/download PDF
31. A point mutation is responsible for the acquisition of transforming properties by the T24 human bladder carcinoma oncogene
- Author
-
Reddy, E. Premkumar, Reynolds, Roberta K., Santos, Eugenio, and Barbacid, Mariano
- Abstract
The genetic change that leads to the activation of the oncogene in T24 human bladder carcinoma cells is shown to be a single point mutation of guanosine into thymidine. This substitution results in the incorporation of valine instead of glycine as the twelfth amino acid residue of the T24 oncogene-encoded p21 protein. Thus, a single amino acid substitution appears to be sufficient to confer transforming properties on the gene product of the T24 human bladder carcinoma oncogene.
- Published
- 1982
- Full Text
- View/download PDF
32. Nucleotide sequence of chicken c-myb complementary DNA and implications for myb oncogene activation
- Author
-
Rosson, Dan and Reddy, E. Premkumar
- Abstract
Avian myeloblastosis virus (AMV), like other acute transforming viruses, arose by recombination between its helper virus and host cellular sequences. The latter sequences, termed v-myb, are responsible for the oncogenic properties of the virus. AMY causes acute myeloblastic leukaemia in chickens and transforms a specific class of haematopoietic cells in vitro, but does not induce morphological transformation of cultured fibroblasts, suggesting that only a restricted target-cell population is responsive to its transforming gene product1,2. The normal cellular counterpart of v-myb, c-myb, is highly conserved and is present in all vertebrate and some invertebrate species examined3,4. DNA rearrangements and altered expression of the myb oncogene have been reported in mouse lymphoid tumours5–7and human myeloid8and colon tumours9. The mechanism of activation of the cellular proto-oncogenes is thought to involve the structural alteration of the coding regions that result in either the synthesis of an altered gene product or the enhanced expression of a proto-oncogene caused by alterations in its regulatory elements. To distinguish between these two mechanisms, we have cloned and sequenced the chicken c-myb complementary DNA and compared it with that of v-myb sequences. We demonstrate that during the transduction of the cellular sequences and/or viral passage a substantial portion of the coding region of the c-myb gene has been lost from both the 5′ and 3′ ends, resulting in the generation of a truncated gene product that mediates the transforming function of the virus.
- Published
- 1986
- Full Text
- View/download PDF
33. A cDNA sequence encoding a rabbit heavy chain variable region of the VHa2 allotype showing homologies with human heavy chain sequences
- Author
-
Bernstein, K. E., Reddy, E. Premkumar, Alexander, C. B., and Mage, R. G.
- Abstract
There are only four positions in the amino acid sequences of immunoglobulin variable region domains of all species at which the same amino acid is always found1. The compiled sequences of rabbit heavy chain variable (VH) domains have 48 invariant positions1,2and at 14 additional positions in the first and third framework segments the different amino acids found correlate with the rabbits' VHa allotype3–5. This VHallotypic system with three common ‘alleles’ VHa1, VHa2 and VHa3 inherited in simple mendelian fashion3,4poses the question of how gene families with distinct sets of conserved codons have evolved and been maintained in the midst of information leading to hypervariability. Here we report a cDNA sequence that encodes a protein characteristic of the rabbit VHa2 allotype and we predict the allotype-associated codons that may be found in VHa1 and VHa3 genes. We have also found within the second hypervariable, complementarity-determining region (CDR2) encoded by our cDNA clone, a DNA sequence highly homologous to a human DHminigene6. This observation and a similar one of Wu and Kabat7, who identified a DHminigene sequence in the CDR2 of a cloned human VHgene8, lends support to their idea that D minigene information has contributed to CDR2 either during evolution or ontogeny7. Gene conversion9,10, minigene assortment11,12or insertion13mechanisms may be involved both in the generation of hypervariability and the conservation of allotype codons.
- Published
- 1982
- Full Text
- View/download PDF
34. The Sequenced Combination of Rigosertib and Azacitidine Has Modulatory Effects on CXCL8, RIG-I like Receptor (RLR) and Wnt/ß-Catenin Signaling and Downstream Hematopoiesis Pathways in an in Vitro Model of the Myelodysplastic Syndrome
- Author
-
Rai, Richa, Melana, Stella, Navada, Shyamala C., Odchimar-Reissig, Rosalie, Demakos, Erin P., Reddy, E. Premkumar, and Silverman, Lewis R.
- Abstract
Navada: Onconova Therapeutics Inc: Research Funding. Reddy:Onconova Therapeutics Inc: Equity Ownership, Research Funding. Silverman:Medimmune: Research Funding; Onconova Therapeutics Inc: Patents & Royalties, Research Funding; Celgene: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
35. Sequential Treatment with Rigosertib Followed By Azacitidine Maximizes the Effects on the Interferon Signaling Pathway in Hematopoietic Cells in Myelodysplastic Syndrome (MDS)
- Author
-
Rai, Richa, Melana, Stella, Navada, Shyamala C., Odchimar-Reissig, Rosalie, Demakos, Erin P., Reddy, E. Premkumar, and Silverman, Lewis R.
- Abstract
Navada: Onconova: Research Funding. Silverman:Mount Sinai School of Medicine: Employment; Onconova Therapeutics Inc.: Patents & Royalties, Research Funding; Celgene: Research Funding; Medimmune: Research Funding; Johnson and Johnson: Research Funding; Bayer: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
36. Effects of Rigosertib (RIGO) Alone or in Combination with Azacitidine or Vorinostat on Epigenetic Reprogramming of CD34+ Cells in the Myelodysplastic Syndrome
- Author
-
Silverman, Lewis R, Chaurasia, Pratima, Melana, Stella, Odchimar-Reissig, Rosalie, Demakos, Erin P., Navada, Shyamala C., and Reddy, E. Premkumar
- Abstract
Rigosertib (RIG) which functions as a RAS-mimetic, binds to the RAS-binding domain (RBD) impacting many pathways that involve RBD-mediated interactions, including the RAS-Raf and AKT/PI3-kinase pathways (Divakar et al., Cell 2016). Recently, we demonstrated that RIG also functions as an epigenetic modifier in MDS. RIG alone or in combination with Azacitidine (AZA) exerted epigenetic effects on the global histone post-translational modifications (PTMs), chromatin remodelers, HDACs, and DNMTs and caused enhanced apoptosis in MDS (MDS-L) and AML (BW-90) cell lines as well as in bone marrow cells (BM) derived from pts with MDS (Chaurasia et al ASCO 2016). In clinical trials, the combination of RIG and AZA demonstrated an overall response rate of 76% in all MDS patients; 62% response in pts who had failed prior hypomethyating agent (HMA) (Navada et al ASH 2016) and 85% in HMA naive pts. The growing evidence suggests that combined therapy may be more effective than either agent alone. To further study these effects, we explored activity of RIG in combination with Vorinostat (VOR), a histone deacetylase inhibitor, on the cell lines. Combined treatment in sequential order VOR/RIG and RIG/VOR potentiates far greater effects on the global PTMs, HDACs, DNMTs, chromatin remodelers, cell cycle checkpoint proteins and PI3/AKT pathway than RIG/AZA or AZA/RIG.
- Published
- 2017
- Full Text
- View/download PDF
37. Stage-Specific Human Induced Pluripotent Stem Cells Map the Progression of Myeloid Transformation to Transplantable Leukemia
- Author
-
Kotini, Andriana G., Chang, Chan-Jung, Chow, Arthur, Yuan, Han, Ho, Tzu-Chieh, Wang, Tiansu, Vora, Shailee, Solovyov, Alexander, Husser, Chrystel, Olszewska, Malgorzata, Teruya-Feldstein, Julie, Perumal, Deepak, Klimek, Virginia M., Spyridonidis, Alexandros, Rampal, Raajit K., Silverman, Lewis, Reddy, E. Premkumar, Papaemmanuil, Elli, Parekh, Samir, Greenbaum, Benjamin D., Leslie, Christina S., Kharas, Michael G., and Papapetrou, Eirini P.
- Abstract
Myeloid malignancy is increasingly viewed as a disease spectrum, comprising hematopoietic disorders that extend across a phenotypic continuum ranging from clonal hematopoiesis to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). In this study, we derived a collection of induced pluripotent stem cell (iPSC) lines capturing a range of disease stages encompassing preleukemia, low-risk MDS, high-risk MDS, and secondary AML. Upon their differentiation, we found hematopoietic phenotypes of graded severity and/or stage specificity that together delineate a phenotypic roadmap of disease progression culminating in serially transplantable leukemia. We also show that disease stage transitions, both reversal and progression, can be modeled in this system using genetic correction or introduction of mutations via CRISPR/Cas9 and that this iPSC-based approach can be used to uncover disease-stage-specific responses to drugs. Our study therefore provides insight into the cellular events demarcating the initiation and progression of myeloid transformation and a new platform for testing genetic and pharmacological interventions.
- Published
- 2017
- Full Text
- View/download PDF
38. Rigosertib Blocks RAS Signaling By Acting As a Small Molecule RAS Mimetic That Binds to the RAS-Binding Domains of RAS Effector Proteins
- Author
-
Reddy, E. Premkumar, Divakar, Sai Krishna, Vasquez-Del Carpio, Rodrigo, Dutta, Kaushik, Baker, Stacey J, Reddy, Ramana, and Aggarwal, Aneel K
- Abstract
Reddy: Onconova Therapeutics Inc: Research Funding. Divakar:Onconova Therapeutics Inc: Research Funding. Vasquez-Del Carpio:Onconova Therapeutics Inc: Research Funding. Dutta:Onconova Therapeutics Inc: Research Funding. Baker:Onconova Therapeautics Inc: Consultancy. Reddy:Onconova Therapeutics Inc: Consultancy. Aggarwal:Onconova Therapeutics Inc: Research Funding.
- Published
- 2014
- Full Text
- View/download PDF
39. Weighted Gene Co-Expression Network Analysis (WGCNA) Identifies Highly Proliferative Myeloma Subgroup Responsive to CDK4/ARK5 Inhibition
- Author
-
Perumal, Deepak, Leshchenko, Violetta V., Kuo, Pei-Yu, Jiang, Zewei, Readhead, Ben, Eden, Caroline, Athaluri Divakar, Sai Krishna, Zhang, Weijia, Cho, Hearn Jay, Chari, Ajai, Reddy, M.V.Ramana, Reddy, E. Premkumar, Dudley, Joel, Jagannath, Sundar, and Parekh, Samir
- Abstract
Chari: Array BioPharma: Scientific Advisory Board Other; Celgene Corporation: Scientific Advisory Board, Scientific Advisory Board Other; Millennium Pharmaceuticals, Inc: Scientific Advisory Board, Scientific Advisory Board Other; Onyx Pharmaceuticals, Inc.: Scientific Advisory Board, Scientific Advisory Board Other. Reddy:Onconova Therapeutics, Inc.: Consultancy, Patents & Royalties. Reddy:Onconova Therapeutics, Inc.: Consultancy, Patents & Royalties. Dudley:Ecoeos, Inc: Consultancy, Equity Ownership; GNS Healthcare: Consultancy; GlaxoSmithKline: Consultancy; Ayasdi, Inc: Equity Ownership; NuMedii, Inc: Equity Ownership; Ubalo, Inc.: Equity Ownership. Jagannath:Onyx Pharmaceuticals, Inc.: Consultancy; Celgene Corporation: Scientific Advisory Board, Scientific Advisory Board Other; Millennium Pharmaceuticals, Inc. : Scientific Advisory Board, Scientific Advisory Board Other.
- Published
- 2014
- Full Text
- View/download PDF
40. Predictors Of Response To Rigosertib In Patients With a Myelodysplastic Syndrome (MDS) Or Acute Myeloid Leukemia (AML) Relapsing After Or Refractory To Hypomethylating Agents
- Author
-
Navada, Shyamala C., Odchimar-Reissig, Rosalie, Reddy, E. Premkumar, Demakos, Erin P, Holland, James F, Wilhelm, Francois, and Silverman, Lewis R.
- Abstract
Rigosertib is a small molecule anti-cancer agent which inhibits the PI-3K and PLK pathways, promotes G2/M arrest, and selectively induces apoptosis in cancer cells. We have previously reported results of a phase I/II study of rigosertib in patients (pts) with MDS and AML who had relapsed or were refractory to hypomethylating agents (HMA), a population for which there are currently no approved second line therapies. Rigosertib appeared to be well tolerated in this pt population and to have biologic activity with reduction or stabilization of bone marrow (BM) blasts and improvement in the peripheral blood (PB) counts in few treated pts. Reduction in BM blasts by rigosertib was associated with increased survival. In the current analysis, we evaluate pt characteristics that may predict for response.We analyzed the results of a phase I/II study of Rigosertib that was conducted in pts with MDS and AML. In the phase I component, pts were entered in cohorts of escalating doses in a classic 3+3 design ranging from 650 up to 1700 mg/m2/d continuous IV infusion (CIV) administered for 72 to 144 hours. A MTD of 1375 mg/m2 was identified for the phase II component, and subsequent pts were treated with this dose as a CIV for 72 hours. BMs were performed at baseline, week 4, 8, and then q3 months.Twenty-two pts with MDS or AML refractory or relapsing to a HMA have been treated with rigosertib. The study cohort comprised pts with a diagnosis of int-2 MDS (2), high risk MDS (6), CMMOL (1), and AML (13 pts all with antecedent MDS). Responses according to IWG 2006 criteria were observed in the BM and PB: marrow CR (4), marrow PR (2). Two pts also had hematologic improvement of the erythroid (1) and platelet (1) lineages. Four pts had stable disease (SD) after treatment but their courses were complicated by infections requiring hospitalization and removal from study. Three pts were deemed to be inevaluable because they received < 2 cycles of treatment or did not have a follow-up BM evaluation. Thus, 10/19 evaluable pts (53%) demonstrated either a BM/PB response (6) or SD (4).The median overall survival (OS) of pts with marrow CR+PR (n=6) was 12 months versus 1.8 months for those without a BM response (n=9) (p=0.0159, log-rank test). Age was not a predictor of response. 1 out of 6 responders had a major elimination in the size of the clonal population. Non-responders did not have a change in the magnitude of the clone. Prior response to HMA was not a predictor for response to rigosertib. Those who did have a marrow CR or PR responded early with median time to response of 2-4 cycles. Those with higher blast counts were less likely to respond. At study entry, the median blast percentage of responders was 16% versus 44% for non-responders. Less than 20% blasts at study entry was a positive predictor of response (p=0.047). Of those pts who did not respond or were inevaluable, the majority (75%) had AML, many with a proliferative course. Nine of 19 pts developed cystitis manifested by dysuria and/or hematuria as a side-effect of therapy. Among responding patients, 5 of 6 had cystitis [grade (GR) 1 (2); GR 2 (1); GR 3 (2)] compared with 3 of 9 non-responders [GR 1(1); GR 2 (1); GR 3(1)] (p=0.08). In one responding pt with grade 3 cystitis, a cystoscopy was performed which revealed polypoid inflammatory changes of the mucosa with hemorrhage. Biopsy showed neutrophilic inflammation without malignant cells. Upon resolution of symptoms, treatment was restarted at 50% of the original dose without complications. Pts who developed symptomatic cystitis were treated with sodium bicarbonate with improvement. The relationship between cystitis and response is being investigated.Rigosertib appears to have biologic activity with reduction in BM blasts associated with increased survival and improvement in the PB counts in a subset of treated pts. Pts with <20% blasts at study entry had a greater likelihood of response. Pts with proliferative disease with rapidly rising or high wbc did not respond. Age, cytogenetic profiles, and response to prior therapy were not predictors of response. Cystitis may be a response related biomarker and requires further analysis. A phase III multicenter randomized trial is underway to compare rigosertib to best supportive care with a primary endpoint of OS in pts with higher risk MDS who have failed, progressed, or relapsed after treatment with HMA, and can be used to validate the observations reported here in a larger study.Reddy: Onconova: Equity Ownership, Research Funding. Holland:Onconova: Research Funding. Wilhelm:Onconova Therapeutics: Employment, Equity Ownership. Silverman:Onconova: Research Funding.
- Published
- 2013
- Full Text
- View/download PDF
41. Evaluation of Rigosertib in Patients with a Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) Relapsed or Refractory to Hypomethylating Agents: A Phase I/II Study
- Author
-
Navada, Shyamala C., Odchimar-Reissig, Rosalie, Reddy, E. Premkumar, Holland, James F, Wilhelm, Francois, and Silverman, Lewis R.
- Abstract
Rigosertib is a small molecule anti-cancer agent with a multi-targeted mechanism of action. It is a multi-kinase/PI3 kinase inhibitor that promotes G2/M arrest and selectively induces apoptosis in cancer cells. Leukemic cells exhibit significantly higher levels of sensitivity to rigosertib compared to normal marrow progenitors and increasing cytotoxicity upon prolonged and repetitive exposure (Chen Proc AACR 2008). Azacitidine is first-line therapy for patients (pts) with higher-risk MDS and produces a response rate of 50%. Pts relapsed or refractory to hypomethylating agents have a short life expectancy of approximately 4 to 6 months (Jabbour 2010, Prebet 2011). There are no approved second line therapies for this patient population.A phase I/II study of Rigosertib is being conducted in pts with MDS and AML. Pts with higher-risk disease had to have failed a hypomethylating agent. In the phase I component, pts were entered in cohorts of escalating doses in a classic 3+3 design ranging from 650 up to 1700 mg/m2/d continuous IV infusion (CIV) for durations from 72 hours to 144 hours every 2 weeks (1 cycle) for 4 cycles of treatment during the induction phase. Subsequent treatments were administered every 3 to 4 weeks. A maximum tolerated dose of 1375 mg/m2 was identified for the phase II component, and subsequent pts were treated with this dose as a CIV for 72 hours. A CBC was performed weekly and a bone marrow (BM) was performed at baseline and week 4, 8, and then q3 months afterwards.Twenty-one patients with MDS or AML refractory/relapsed to a hypomethylating agent have been treated with rigosertib. The study cohort comprised pts with a diagnosis of intermediate-2 MDS (2 pts), high risk MDS (5 pts), chronic myelomonocytic leukemia (1 pt), and AML (13 pts) (all AML had an antecedent MDS). The median age was 79 years. 86% of pts were male. Patients received between 1–19 cycles of treatment. Their cytogenetic profiles were diverse with the most recurrent abnormalities including a complex karyotype (5 pts), normal (5 pts), monosomy 7 (4 pts), and trisomy 8 (2 pts). Responses according to IWG 2006 criteria were observed in the BM and peripheral blood: marrow CR (4), hematologic improvement (HI) (2); erythroid (1) platelet (1). Time to response was 2–4 cycles. An additional 2 pts had a >50% BM blast decrease from baseline but not to < 5%. Three pts had stable disease after treatment but their courses were complicated by infections requiring hospitalization and removal from the study. Three pts were deemed to be inevaluable because they received less than 2 cycles of treatment or did not have a follow-up bone marrow evaluation. Thus, 9/18 evaluable pts (50%) demonstrated either a bone marrow/peripheral blood response (6) or stable disease (3). The median overall survival of those with marrow CR+PR was 10.1 months versus 2 months for those without a bone marrow response (p=0.0011, log-rank test). Of those pts who did not respond or were inevaluable, the majority (83%) had AML, many with a proliferative course. The most frequent grade 1–2 side-effects included dysuria, hematuria, fatigue, anorexia, nausea, and diarrhea. Possibly related grade 3 side-effects included fatigue, hematuria, and dyspnea, each in one pt. Six of 21 pts developed cystitis manifested by dysuria and/or hematuria. Among responding or stable patients, 5 of 9 had cystitis compared with 1 of 12 non-responders. Patients who developed symptomatic cystitis were treated with sodium bicarbonate with improvement. The relationship between dysuria and/or cystitis and response is being investigated.Rigosertib appears to be safe and well tolerated in patients with refractory or relapsed MDS and AML. It has biologic activity with reduction in BM blasts and improvement in the peripheral blood counts in a subset of treated pts, and these effects are associated with increased survival. Dysuria/cystitis may be a response related biomarker and requires further analysis. Data regarding pharmacokinetics and pharmacodynamics will be presented and correlated to response. Given promising initial results, a phase III multicenter randomized trial is underway to compare rigosertib to best supportive care with a primary endpoint of overall survival in patients with higher risk MDS who have failed, progressed, or relapsed after treatment with hypomethylating agents.Reddy: Onconova: Research Funding. Holland:Onconova: Research Funding. Wilhelm:Onconova: Employment, Equity Ownership. Silverman:Onconova: Research Funding.
- Published
- 2012
- Full Text
- View/download PDF
42. An In Vivo Functional Screen Identifies miRNA-150 As a Regulator of Hematopoietic Regeneration Post Chemotherapeutic Injury
- Author
-
Adams, Brian D., Guo, Shangqin, Bai, Haitao, Xiao, Changchun, Reddy, E. Premkumar, and Lu, Jun
- Abstract
No relevant conflicts of interest to declare.
- Published
- 2011
- Full Text
- View/download PDF
43. An In VivoFunctional Screen Identifies miRNA-150 As a Regulator of Hematopoietic Regeneration Post Chemotherapeutic Injury
- Author
-
Adams, Brian D., Guo, Shangqin, Bai, Haitao, Xiao, Changchun, Reddy, E. Premkumar, and Lu, Jun
- Abstract
Abstract 2333
- Published
- 2011
- Full Text
- View/download PDF
44. Evaluation of ON01910.Na In Patients with a Myelodysplastic Syndrome (MDS) or Acute Myeloid Leukemia (AML) Relapsed or Refractory to Hypomethylating Agents: A Phase I Study
- Author
-
Silverman, Lewis R, Navada, Shyamala C., Odchimar-Reissig, Rosalie, Najfeld, Vesna, Ohnuma, Takao, Wilhelm, Francois, Reddy, E. Premkumar, and Holland, James F.
- Abstract
ON 01910.Na a novel benzyl styryl sulfone derivative is under clinical development in hematologic malignancies. It is a multi-kinase/PI3 kinase inhibitor that promotes G2/M arrest and selectively induces apoptosis in cancer cells. Leukemic cells exhibit significantly higher levels of sensitivity to ON 01910.Na compared to normal marrow progenitors and increasing cytotoxicity upon prolonged and repetitive exposure (Skidan Proc AACR 2006; Chen Proc AACR 2008). Azacitidine (AzaC), is first line therapy for patients (pts) with higher-risk MDS and produces a response rate of 50%. Pts relapsed or refractory to hypomethylating based therapies have a poor prognosis and there are no accepted effective second line treatments, thus a need for new agents.A phase I/II study of ON 01910.Na is being conducted in pts with hematological malignancies. In the phase I component pts are entered in cohorts of escalating doses in a classic 3+3 design in doses ranging from 650 up to 1700 mg/m2/d continuous IV infusion (CIV) for durations from 72 hours up to 144 hours every 2 weeks (1 cycle) for 4 cycles of treatment during the induction phase. Subsequent treatments are administered every 3 to 4 weeks. A CBC is performed weekly and a bone marrow (BM) is performed at baseline and week 4, 8, and then q3 months thereafter. Pts with higher-risk disease had to have failed a hypomethylating agent.Ten pts with MDS or AML relapsed/refractory to a hypomethylating agent have been treated with ON 01910.Na thus far (table 1). The study cohort comprised pts with a diagnosis (Dx) RAEB-2 (4 pts), RAEB-T (1 pt), and AML (5 pts) (median age of 75 years). Their cytogenetic profile included 1 pt with normal, 2 with intermediate (+8), and 7 pts with poor risk cytogenetics (monosomy 7 and/or complex). Patients were treated between 5 and 70 weeks. Responses according to IWG 2006 criteria were observed in the BM and peripheral blood: Marrow CR (3), hematologic improvement (HI-P) (2); erythroid (1) platelet (1). An additional 2 pts had a >50% BM blast decrease from baseline but not to <5%. Thus, 5/10 (50%) demonstrate a bone marrow response. Survival of these pts was 7.3, 15.7, and 16.4 months; one patient remains on study 5+ months. Four of the five responders had MDS at the initiation of treatment: RAEB-2 (3), CMMoL (1), AML (1). Responders had monosomy 7 (2), trisomy 8 (1) and complex cytogenetics (2). One pt had an elimination of the MDS clone and the others had persistence of the abnormal karyotype throughout their treatment course. Five pts had SD without HI at 4 weeks, 2 pts progressed to AML. All 5 non-responders had AML; 4 with a proliferative course. These latter received only 2 (2) or 3 (3) cycles before succumbing to disease related infectious complications. Survival for these patients ranged from 1.3 – 2 months with a median duration on study of 42 days. The most frequent side effects grade2 2 for all pts included fatigue, anorexia, nausea, and dysuria in patients receiving extended duration infusions. One pt had a grade 3 urinary frequency. No hematologic toxicities occurred and no bone marrow toxicity or hypoplasia was noted. Pharmacokinetic studies are ongoing, data to date demonstrate no evidence of drug accumulation in patients who are treated repeatedly.ON 01910.Na appears to be safe and well tolerated in patients with refractory or relapsed MDS and AML. ON 01910.Na has biologic activity with reduction in BM blasts, eradication of the MDS clone and improvement in the peripheral blood counts in some pts. These effects are associated with increased survival albeit in limited numbers of pts treated thus far. Further study of ON 01910.Na is warranted to better define biologic activity, appropriate target populations and to define mechanism of action.Silverman: Onconova Therapeutics Inc: Research Funding, Research support of Clinical Trial. Wilhelm:Onconova Therapeutics Inc: Employment, Equity Ownership.
- Published
- 2010
- Full Text
- View/download PDF
45. Synergistic Effects of a Novel Water-Soluble Small Molecule, ON 013105, and Rituximab on Mantle Cell Lymphoma In Vitro and In Vivo
- Author
-
Prasad, Anil, Shrivastava, Ashutosh, Reddy, Ramana, Gillum, Amanda M., Reddy, E. Premkumar, and Groopman, Jerome E.
- Abstract
No relevant conflicts of interest to declare.
- Published
- 2010
- Full Text
- View/download PDF
46. Editorial
- Author
-
Reddy, E. Premkumar
- Published
- 2010
- Full Text
- View/download PDF
47. Effects of a Novel Benzyl Styryl Sulfone Derivative ON 01910.Na On the Myelodysplastic Syndrome (MDS) Derived Clone in Patients Relapsing Following Response to Azacitidine (AzaC) Therapy.
- Author
-
Silverman, Lewis R., Odchimar-Reissig, Rosalie, Navada, Shyamala C., Ohnuma, Takao, Najfeld, Vesna, Wilhelm, Francois, Reddy, E. Premkumar, and Holland, James F
- Abstract
ON 01910.Na, a novel benzyl styryl sulfone derivative is currently under clinical development as an anticancer agent in solid tumors and hematologic malignancies. ON 01910.Na selectively targets tumor cells at the mitotic phase of the cell cycle, arrests leukemia cells at the G2/M stage and induces apoptosis in a time and dose dependent manner. ON 01910.Na affects multiple cellular pathways, has multikinase activity and has been shown to target multiple genetic pathways, including those that are regulated by cyclin dependent kinases. Our studies indicate that leukemic cells exhibit significantly higher levels of sensitivity of ON01910.Na compared to normal marrow progenitors and increasing cytotoxicity upon prolonged and repetitive exposure (Skidan Proc AACR 2006; Chen Proc AACR 2008). AzaC alters the natural history of higher risk MDS with effects on marrow function, reduction in the risk of leukemic transformation and increased overall survival (Silverman JCO 2002, 2006; Fenaux Lancet 2009). However, 50% of patients (pts) do not respond to AzaC and upon progression after treatment their prognosis is poor, thus, a need for alternative agents.A phase I/II study of ON 01910.Na is being conducted in pts with heme malignancies. In the phase I component pts are entered in cohorts of escalating doses in a classic 3+3 design in doses ranging from 650 up to 1700 mg/m2/d continuous IV infusion (CI) for durations from 72 hours up to 144 hours every 2 weeks (1 cycle) for 4 cycles of treatment during the induction phase. Subsequent treatments are administered every 3 to 4 weeks.This report details two pts with progressive disease following treatment for MDS with AzaC at a dose of 75mg/m2/d x 7 days SC every 4 weeks. Upon progression they were treated with ON 01910.Na at 1050 mg/m2/d CI x 72h to 144 hours. Patient 002 presented with MDS int-2 with myelofibrosis and mild splenomegaly, 8% marrow blasts, normal karyotype, and RBC transfusion dependence. AzaC therapy was initiated in 9/2004. A hematologic improvement (HI) was achieved according to modified IWG criteria with marrow myeloblasts < 5%, RBC transfusion independence and a persistence of mild splenomegaly. A stable HI was maintained for 4 years on monthly AzaC therapy until relapse. At progression (10/2008), marrow myeloblasts increased to 11%, RBC transfusion requirement resumed, platelets fell to 20 × 109/L, the spleen enlarged and a new cytogenetically abnormal clone emerged, 46,XY,del(20)(q11q13),i(21)(q10),der(21)t(1;21)(q12;p11) resulting in trisomy for 1q. Administration of ON 01910.Na 1050 mg/m2/d x 72 hours CI was initiated 3 months after the last dose of AzaC. After 2 cycles of treatment evaluation demonstrated a reduction in: marrow myeloblast to 1%, spleen size by 50% and the MDS clone by 74% by standard cytogenetic analysis. Following the 4th and 7th cycles of treatment with ON 01910.Na, marrow myeloblasts ranged from 1 to 3%, palpable splenomegaly resolved, RBC transfusions decreased by 33% and platelets rose to 80 × 109/L. Although the conventional karyotype normalized after the 4th cycle, all 3 abnormalites were detected by FISH in 30% of cells following the 4th and 7th cycles suggesting a loss of proliferative capacity of the MDS clone. Patient 004 presented with high risk MDS, 19% marrow blasts, pancytopenia and a complex karyotype. AzaC treatment was initiated in 9/2002 leading to a stable HI, with marrow myeloblasts < 5%, RBC transfusion independence, and persistence of the MDS clone with no change in the complex karyotype, including deletion of the MLL locus. The HI was maintained for 6.5 years with monthly AzaC therapy until progression. At relapse marrow myeloblasts increased to 19%, and the complex karyotype persisted unchanged from initial diagnosis. ON 01910.Na treatment started (3/2009) and evaluations at the 2nd, 4th and 7th cycles revealed: a) reduction in % marrow myeloblasts to 12%, 8% and 7% respectively; b) persistence of abnormal karyotype in over 90% of metaphase and interphase cells, irrespective of the decreased % blasts; c) increased platelets from 20 to 40 × 109/L. Patient 002 and 004 remain on treatment with ON 01910.Na at 8+ and 5+ months, respectively.Preliminary data suggest that ON 01910.Na has an anti-proliferative modulatory effect on the MDS clone in pts progressing after AzaC therapy and should be explored in a broader MDS population.Silverman: Onconova Therapeutics: Research Funding. Off Label Use: ON01910Na is investigational and not FDA approved. Wilhelm:Onconova: Employment. Reddy:Onconova Therapeutics Inc.: Consultancy, Equity Ownership, Grantee, Membership on an entity's Board of Directors or advisory committees. Holland:Onconova Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding.
- Published
- 2009
- Full Text
- View/download PDF
48. Single Cell Network Profiling (SCNP) to Evaluate the Mechanism of Action of ON 01910.Na, A Novel Clinical Trial Stage Compound.
- Author
-
Soper, David M., Huang, Ying-Wen, Wilhelm, Francois, Cosenza, S. C., Reddy, E. Premkumar, Cesano, Alessandra, Greenberg, Peter L, and Fantl, Wendy J.
- Abstract
ON 01910.Na, a small molecule multikinase inhibitor, promotes G2/M arrest and apoptosis. Key targets for this inhibitor include Plk1 (polo-like kinase, a cell cycle regulator), Cdk1, (cyclin dependent kinase, a mitotic regulator) and the PI-3 kinase pathway (Ramana Reddy et al. J. Med. Chem. 2008, Park et al, Oncogene, 2007, Gumireddy et al., Cancer Cell, 2005). The drug has been shown to have anti-tumor activity in in vitro and in vivo models. Phase I studies in >100 advanced cancer patients revealed that the drug is well tolerated. Further, in several ongoing Phase 1 clinical trials in patients with myelodysplastic syndromes (MDS), positive effects on hematological indicators have been noted (Sloand et al, ASH 2008). Based on these data, a Phase 2 single-arm study is in progress to assess the efficacy and safety of the drug in IPSS Intermediate-2 and High risk MDS patients. Single Cell Network Profiling (SCNP) using flow cytometry is a platform that measures multiple fluorescent parameters (up to 10) in each cell, including both surface markers and intracellular signaling proteins in response to extracellular network inputs. By simultaneously measuring the effects of drug exposure on several pathways within each cell type in a heterogeneous patient tissue sample, valuable data can be gained about drug interactions with specific cellular pathways and cell type selectivity. This information has potential implications for dose/schedule optimization and development of patient stratification biomarkers.Studies were designed to evaluate the in vitro effects of ON 01910.Na, at clinically relevant concentrations, on intracellular pathways in the human GM-CSF-dependent erythroblastic TF-1 cell line using SCNP in order to monitor transitional changes in the cell cycle, with a focus on the G2-M phase and to perform dose-dependent titrations of drug using these cell cycle readouts.The reagents chosen to measure cell cycle readouts were fluorochrome-conjugated antibodies that recognize cyclin B1, p-histone H3(S28) and p-Cdk1(Y15) and 4'6'-diamino-2-phenylindole (DAPI), a fluorescent dye that binds strongly to DNA. The phosphorylation status of p-histone H3(S28) and p-Cdk1(Y15), and the level of cyclin B1 expression are all determinants of the G2-M and/or M phase of the cell cycle. Dose dependent titrations of ON 01910.Na and its inactive analog ON 01911 were performed over a dose range starting at 10-5 M and decreasing to 10-10 M (dose range which includes pharmacologically achievable concentrations in humans) with 3-fold serial dilutions for eleven points after an exposure to the drug for either 24 or 48 hrs. Cells were processed for multiparameter flow cytometry by fixation, permeabilization and incubation with fluorchrome-conjugated antibodies.The data showed that at 24 hours after ON 01910.Na exposure there was a simultaneous increase in phosphorylation of histone H3(S28), a decrease in phosphorylation of Cdk-1(Y15), and accumulation of cyclin B1. These data suggest that ON 01910 exposure disrupted the G2/M cell cycle transition leading to mitotic arrest with subsequent apoptosis. TF-1 cell DNA content measured by DAPI verified this to be the case as increases in G2/M and sub-G1 (a measure of apoptotic cell death) were simultaneously observed. No significant effects on G2/M targets were observed when TF-1 cells were exposed to ON 01911, indicating the effects of ON 01910.Na on the cell cycle were specific to the drug. Maximal effects of ON-01910.Na on cell cycle signaling molecules were observed at a drug concentration of 0.37 mM and no further changes were seen at higher concentrations. These effects were also observed at 48 hours, although with more cell death.These data indicate that intracellular phosphorylation changes of histone H3(S28) and Cdk-1(Y15), in addition to accumulation of cyclin B1 with subsequent apoptosis, reflect possible mechanisms of action of ON 01910.Na. The assay will be used in ongoing clinical trials to measure the pharmacodynamic activity of the drug in MDS patient samples pre- and post-treatment.Soper: Nodality Inc.: Employment, Equity Ownership. Huang:Nodality Inc.: Employment, Equity Ownership. Wilhelm:Onconova Therapeutics Inc: Employment. Cosenza:Onconova Therapeutics Inc.: Consultancy. Reddy:Onconova Therapeutics Inc.: Consultancy, Equity Ownership, Grantee, Membership on an entity's Board of Directors or advisory committees. Cesano:Nodality Inc.: Employment, Equity Ownership. Greenberg:Nodality Inc.: Research Funding; Onconova Therapeutics Inc.: Research Funding. Fantl:Nodality, Inc.: Employment, Equity Ownership.
- Published
- 2009
- Full Text
- View/download PDF
49. Evaluation of Novel Cell Cycle Inhibitors in Mantle Cell Lymphoma.
- Author
-
Park, In-Woo, Reddy, M.V. Ramana, Reddy, E. Premkumar, and Groopman, Jerome E.
- Abstract
Signature abnormalities in the cell cycle and apoptotic pathway have been identified in mantle cell lymphoma (MCL), affording the opportunity to develop targeted therapies. Here, we report on a novel class of kinase inhibitors, styryl sulfones, that differ from prior cell cycle inhibitors in that they are not related to purines or pyrimidines. We observed that two closely related compounds, ON013100 and ON01370, altered the growth and cell cycle status of MCL lines and potently inhibited the expression of several important molecules, including CDK4, p53, MDM2, cyclin D, and cyclin B. Using both TUNEL and PARP assays, we found that these compounds caused apoptosis in MCL cells. In addition, using molecular analyses, we observed the modulation of caspase-3 activity but not the expression of Bcl family molecules in these cells. Next, we investigated the cytotoxicity of the MCL lines upon treatment with styryl sulfone compounds in combination with other currently used chemotherapeutic agents, such as doxorubicin (DOX) or vincristine (VCR). We found that the combination of DOX plus styryl sulfone or VCR plus styryl sulfone increased cytotoxicity by one log scale, compared with the single styryl sulfone compound. Thus styryl sulfones alone, or in combination with chemotherapeutic agents, present attractive opportunities for new drug development in MCL.
- Published
- 2006
- Full Text
- View/download PDF
50. Sequential Reduction and Dehydration of Phenacyl‐(E)‐styryl Sulfones to Unsymmetrical (E,E)‐Bis(styryl) Sulfones.
- Author
-
Mallireddigari, Muralidhar Reddy, Pallela, Venkat R., Reddy, E. Premkumar, and Reddy, M. V. Ramana
- Abstract
ChemInform is a weekly Abstracting Service, delivering concise information at a glance that was extracted from about 200 leading journals. To access a ChemInform Abstract, please click on HTML or PDF.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.