Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Vladimir Beljanski is active.

Publication


Featured researches published by Vladimir Beljanski.


Journal of Pharmacology and Experimental Therapeutics | 2010

Pharmacology and Antitumor Activity of ABC294640, a Selective Inhibitor of Sphingosine Kinase-2

Kevin J. French; Yan Zhuang; Lynn W. Maines; Peng Gao; Wenxue Wang; Vladimir Beljanski; John J. Upson; Cecelia L. Green; Staci N. Keller; Charles D. Smith

Sphingolipid-metabolizing enzymes control the dynamic balance of the cellular levels of important bioactive lipids, including the apoptotic compound ceramide and the proliferative compound sphingosine 1-phosphate (S1P). Many growth factors and inflammatory cytokines promote the cleavage of sphingomyelin and ceramide leading to rapid elevation of S1P levels through the action of sphingosine kinases (SK1 and SK2). SK1 and SK2 are overexpressed in a variety of human cancers, making these enzymes potential molecular targets for cancer therapy. We have identified an aryladamantane compound, termed ABC294640 [3-(4-chlorophenyl)-adamantane-1-carboxylic acid (pyridin-4-ylmethyl)amide], that selectively inhibits SK2 activity in vitro, acting as a competitive inhibitor with respect to sphingosine with a Ki of 9.8 μM, and attenuates S1P formation in intact cells. In tissue culture, ABC294640 suppresses the proliferation of a broad panel of tumor cell lines, and inhibits tumor cell migration concomitant with loss of microfilaments. In vivo, ABC294640 has excellent oral bioavailability, and demonstrates a plasma clearance half-time of 4.5 h in mice. Acute and chronic toxicology studies indicate that ABC294640 induces a transient minor decrease in the hematocrit of rats and mice; however, this normalizes by 28 days of treatment. No other changes in hematology parameters, or gross or microscopic tissue pathology, result from treatment with ABC294640. Oral administration of ABC294640 to mice bearing mammary adenocarcinoma xenografts results in dose-dependent antitumor activity associated with depletion of S1P levels in the tumors and progressive tumor cell apoptosis. Therefore, this newly developed SK2 inhibitor provides an orally available drug candidate for the treatment of cancer and other diseases.


Journal of Pharmacology and Experimental Therapeutics | 2010

A NOVEL SPHINGOSINE KINASE INHIBITOR INDUCES AUTOPHAGY IN TUMOR CELLS

Vladimir Beljanski; Christian Knaak; Charles D. Smith

The sphingolipids ceramide, sphingosine, and sphingosine 1-phosphate (S1P) regulate cell signaling, proliferation, apoptosis, and autophagy. Sphingosine kinase-1 and -2 (SK1 and SK2) phosphorylate sphingosine to form S1P, shifting the balanced activity of these lipids toward cell proliferation. We have previously reported that pharmacological inhibition of SK activity delays tumor growth in vivo. The present studies demonstrate that the SK2-selective inhibitor 3-(4-chlorophenyl)-adamantane-1-carboxylic acid (pyridin-4-ylmethyl)amide (ABC294640) induces nonapoptotic cell death that is preceded by microtubule-associated protein light chain 3 cleavage, morphological changes in lysosomes, formation of autophagosomes, and increases in acidic vesicles in A-498 kidney carcinoma cells. ABC294640 caused similar autophagic responses in PC-3 prostate and MDA-MB-231 breast adenocarcinoma cells. Simultaneous exposure of A-498 cells to ABC294640 and 3-methyladenine, an inhibitor of autophagy, switched the mechanism of toxicity to apoptosis, but decreased the potency of the SK2 inhibitor, indicating that autophagy is a major mechanism for tumor cell killing by this compound. Induction of the unfolded protein response by the proteasome inhibitor N-(benzyloxycarbonyl)leucinylleucinylleucinal Z-Leu-Leu-Leu-al (MG-132) or the heat shock protein 90 inhibitor geldanamycin synergistically increased the cytotoxicity of ABC294640 in vitro. In severe combined immunodeficient mice bearing A-498 xenografts, daily administration of ABC294640 delayed tumor growth and elevated autophagy markers, but did not increase terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling-positive cells in the tumors. These data suggest that ABC294640 promotes tumor cell autophagy, which ultimately results in nonapoptotic cell death and a delay of tumor growth in vivo. Consequently, ABC294640 may effectively complement anticancer drugs that induce tumor cell apoptosis.


Cancer Biology & Therapy | 2011

Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts

Vladimir Beljanski; Clayton S. Lewis; Charles D. Smith

The balance between the pro-apoptotic lipids ceramide and sphingosine and the pro-survival lipid sphingosine 1-phosphate (S1P) is termed the “sphingosine rheostat”. Two isozymes, sphingosine kinase 1 and 2 (SK1 and SK2), are responsible for phosphorylation of pro-apoptotic sphingosine to form pro-survival S1P. We have previously reported the antitumor properties of an SK2 selective inhibitor, ABC294640, alone or in combination with the multikinase inhibitor sorafenib in mouse models of kidney carcinoma and pancreatic adenocarcinoma. Here we evaluated the combined antitumor effects of the aforementioned drug combination in two mouse models of hepatocellular carcinoma. Although combining the SK2 inhibitor, ABC294640, and sorafenib in vitro only afforded additive drug-drug effects, their combined antitumor properties in the mouse model bearing HepG2 cells mirrored effects previously observed in animals bearing kidney carcinoma and pancreatic adenocarcinoma cells. Combining ABC294640 and sorafenib led to a decrease in the levels of phosphorylated ERK in SK-HEP-1 cells, indicating that the antitumor effect of this drug combination is likely mediated through a suppression of the MAPK pathway in hepatocellular models. We also measured levels of S1P in the plasma of mice treated with two different doses of ABC294640 and sorafenib. We found decreases in the levels of S1P in plasma of mice treated daily with 100 mg/kg of ABC294640 for 5 weeks, and this decrease was not affected by co-administration of sorafenib. Taken together, these data support combining ABC294640 and sorafenib in clinical trials in HCC patients. Furthermore, monitoring levels of S1P may provide a pharmacodynamic marker of ABC294640 activity. See commentary: Sphingosine kinase: A promising cancer therapeutic target


Investigational New Drugs | 2011

Combined anticancer effects of sphingosine kinase inhibitors and sorafenib

Vladimir Beljanski; Christian Knaak; Yan Zhuang; Charles D. Smith

SummaryThe pro-apoptotic lipid sphingosine is phosphorylated by sphingosine kinases 1 and 2 (SK1 and SK2) to generate the mitogenic lipid sphingosine-1-phosphate (S1P). We previously reported that inhibition of SK activity delays tumor growth in a mouse mammary adenocarcinoma model. Because SK inhibitors and the multikinase inhibitor sorafenib both suppress the MAP kinase pathway, we hypothesized that their combination may provide enhanced inhibition of tumor growth. Therefore, we evaluated the effects of two SK inhibitors, ABC294640 (a SK2-specific inhibitor) and ABC294735 (a dual SK1/SK2 inhibitor), alone and in combination with sorafenib on human pancreatic adenocarcinoma (Bxpc-3) and kidney carcinoma (A-498) cells in vitro and in vivo. Exposure of either Bxpc-3 or A-498 cells to combinations of ABC294640 and sorafenib or ABC294735 and sorafenib resulted in synergistic cytotoxicity, associated with activation of caspases 3/7 and DNA fragmentation. Additionally, strong decreases in ERK phosphorylation were observed in Bxpc-3 and A-498 cells exposed to either the sorafenib/ABC294640 or the sorafenib/ABC294735 combination. Oral administration of either ABC294640 or ABC294735 to mice led to a delay in tumor growth in both xenograft models without overt toxicity to the animals. Tumor growth delay was potentiated by co-administration of sorafenib. These studies show that combination of an SK inhibitor with sorafenib causes synergistic inhibition of cell growth in vitro, and potentiates antitumor activity in vivo. Thus, a foundation is established for clinical trials evaluating the efficacy of combining these signaling inhibitors.


Molecular and Cellular Biology | 2007

Skittish Abca2 Knockout Mice Display Tremor, Hyperactivity, and Abnormal Myelin Ultrastructure in the Central Nervous System†

Jody T. Mack; Vladimir Beljanski; Athena Soulika; Danyelle M. Townsend; Carol B Brown; Warren Davis; Kenneth D. Tew

ABSTRACT The ATP-binding cassette transporter 2 (ABCA2) is an endolysosomal protein most highly expressed in the central and peripheral nervous system tissues and macrophages. Previous studies indicated its role in cholesterol/steroid (estramustine, estradiol, and progesterone) trafficking/sequestration, oxidative stress response, and Alzheimers disease. Developmental studies have shown its expression during macrophage and oligodendrocyte differentiation, processes requiring membrane growth. To determine the in vivo function(s) of this transporter, we generated a knockout mouse from a gene-targeted disruption of the murine ABCA2 gene. Knockout males and females are viable and fertile. However, a non-Mendelian inheritance pattern was shown among male progeny of heterozygous crosses. Compared to wild-type and heterozygous littermates, knockout mice displayed a tremor without ataxia, hyperactivity, and reduced body weight; the latter two phenotypes were more marked in females than in males. This sexual disparity suggests a role for ABCA2 in hormone-dependent neurological and/or developmental pathways. Myelin sheath thickness in the spinal cords of knockout mice was greatly increased compared to that in wild-type mice, while a significant reduction in myelin membrane periodicity (compaction) was observed in both spinal cords and cerebra of knockout mice. Loss of ABCA2 function in vivo resulted in abnormal myelin compaction in spinal cord and cerebrum, an ultrastructural defect that we propose to be the cause of the phenotypic tremor.


Molecular Cancer Therapeutics | 2009

Abstract B251: Combined anticancer effects of sphingosine kinase inhibitors and sorafenib

Vladimir Beljanski; Christian Knaak; Yan Zhuang; Charles D. Smith

Purpose: The pro‐apoptotic lipid sphingosine is phosphorylated by sphingosine kinases 1 and 2 (SK1 and SK2) to generate the pro‐survival lipid sphingosine‐1‐phosphate (S1P). We previously reported that inhibition of SK activity delays tumor growth in a mouse mammary adenocarcinoma tumor model. Because SK inhibitors and the multikinase inhibitor sorafenib both suppress the MAP kinase pathway, we hypothesized that the combination may provide enhanced tumor growth inhibition. Therefore, we evaluated the effects of two SK inhibitors, ABC294640 (a SK2‐specific inhibitor) and ABC294735 (a SK1/SK2 inhibitor), alone and in combination with sorafenib on human pancreatic adenocarcinoma (Bxpc‐3) and kidney carcinoma (A‐498) cells in vitro and in vivo . Experimental Design: Effects of the compounds as single agents or in combination in cell culture were assessed using the SRB assay, and CalcuSyn software was used to calculate synergistic or additive growth inhibition. Western blotting measured effects of compounds on signaling pathways. SCID mice bearing tumors of either Bxpc‐3 or A‐498 cells were treated with the agents and tumor growth was monitored. Results: Exposure of either Bxpc‐3 or A‐498 cells to ABC29464/sorafenib or ABC294735/sorafenib combinations resulted in synergistic cytotoxicity, associated with increases in caspases 3/7 activation and apoptotic DNA fragmentation. In addition, strong decreases in ERK phosphorylation were observed in Bxpc‐3 and A‐498 cells exposed to the either sorafenib/ABC29464 or sorafenib/ABC294735 combinations. An increase in the phosphorylation of inhibitory site of c‐Raf (S259) was also observed in the drug‐treated cells. Oral administration of either ABC294640 or ABC294735 to mice led to a delay in tumor growth in both xenograft models without overt toxicity to the animals. Tumor growth delay was potentiated by co‐administration of sorafenib. Conclusions: These studies show that combination of an SK inhibitor with sorafenib causes synergistic inhibition of cell growth in vitro and potentiates antitumor activity in vivo. Thus, a foundation is established for clinical trials evaluating the efficacy of combining these signaling inhibitors. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B251.


xPharm: The Comprehensive Pharmacology Reference | 2008

Angiostatin 4.5

Vladimir Beljanski

For tumors to grow and metastasize, they must induce the generation of new blood vessels, a process referred to as angiogenesis. Angiogenesis begins when a fibrin clot forms on the external surface of an existing blood vessel, followed by growing of new capillaries. The initial phase begins with increased vascular permeability and local degradation of the vessel wall. The endothelial cells enter the tumor stroma, migrate toward a stimulus such as VEGF or FGF, and proliferate behind the leading edge. At this time, the cells may be most vulnerable to agents that interfere with their proliferation, since they lack protection from other cell types. The next step in vessel formation is recruitment of pericytes, followed by smooth muscle cells. Inhibition of angiogenesis thus provides an important new therapeutic target. The angiogenesis inhibitor angiostatin is a kringle-containing internal fragment of plasminogen, which inhibits cancer growth in numerous animal models …


Current Drug Metabolism | 2007

The ABCA2 transporter: intracellular roles in trafficking and metabolism of LDL-derived cholesterol and sterol-related compounds.

Jody T. Mack; Danyelle M. Townsend; Vladimir Beljanski; Kenneth D. Tew


Biomedicine & Pharmacotherapy | 2006

The ATP-binding cassette transporter ABCA2 as a mediator of intracellular trafficking

Jody T. Mack; Vladimir Beljanski; Kenneth D. Tew; Danyelle M. Townsend


in Vivo | 2005

Characterization of the ATPase Activity of Human ATP-binding Cassette Transporter-2 (ABCA2)

Vladimir Beljanski; Athena Soulika; Jody M. Tucker; Danyelle M. Townsend; Warren Davis; Kenneth D. Tew

Collaboration


Dive into the Vladimir Beljanski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danyelle M. Townsend

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Kenneth D. Tew

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Christian Knaak

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jody T. Mack

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Yan Zhuang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Warren Davis

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carol B Brown

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge