Network


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

Hotspot


Dive into the research topics where M.V. Ramana Reddy is active.

Publication


Featured researches published by M.V. Ramana Reddy.


Cell | 2016

A Small Molecule RAS-Mimetic Disrupts RAS Association with Effector Proteins to Block Signaling

Sai Krishna Athuluri-Divakar; Rodrigo Vasquez-Del Carpio; Kaushik Dutta; Stacey J. Baker; Stephen C. Cosenza; Indranil Basu; Yogesh K. Gupta; M.V. Ramana Reddy; Lynn Ueno; Jonathan R. Hart; Peter K. Vogt; David J. Mulholland; Chandan Guha; Aneel K. Aggarwal; E. Premkumar Reddy

Oncogenic activation of RAS genes via point mutations occurs in 20%-30% of human cancers. The development of effective RAS inhibitors has been challenging, necessitating new approaches to inhibit this oncogenic protein. Functional studies have shown that the switch region of RAS interacts with a large number of effector proteins containing axa0common RAS-binding domain (RBD). Because RBD-mediated interactions are essential for RAS signaling, blocking RBD association with small molecules constitutes an attractive therapeutic approach. Here, we present evidence that rigosertib, a styryl-benzyl sulfone, acts as a RAS-mimetic and interacts with the RBDs of RAF kinases, resulting in their inability to bind to RAS, disruption of RAF activation, and inhibition of the RAS-RAF-MEK pathway. We also find that ribosertib binds to the RBDs of Ral-GDS and PI3Ks. These results suggest that targeting of RBDs across multiple signaling pathways by rigosertib may represent an effective strategy for inactivation of RAS signaling.


Genes & Cancer | 2010

A Non–ATP-Competitive Dual Inhibitor of JAK2V617F and BCR-ABLT315I Kinases Elucidation of a Novel Therapeutic Spectrum Based on Substrate Competitive Inhibition

Shashidhar S. Jatiani; Stephen C. Cosenza; M.V. Ramana Reddy; Ji Hee Ha; Stacey J. Baker; Ajoy K. Samanta; Matthew J. Olnes; Loretta Pfannes; Elaine M. Sloand; Ralph B. Arlinghaus; E. Premkumar Reddy

Here we report the discovery of ON044580, an α-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 JAK2(V617F)-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-ABL(T315I) and JAK2(V617F). 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.


Organic and Biomolecular Chemistry | 2013

Hydrothiolation of benzyl mercaptan to arylacetylene: application to the synthesis of (E) and (Z)-isomers of ON 01910·Na (Rigosertib®), a phase III clinical stage anti-cancer agent

Venkat R. Pallela; Muralidhar R. Mallireddigari; Stephen C. Cosenza; Balaiah Akula; D. R. C. Venkata Subbaiah; E. Premkumar Reddy; M.V. Ramana Reddy

A stereoselective and efficient method for free radical addition of benzyl thiol to aryl acetylene in the presence of Et3B-hexane has been developed for the synthesis of (Z) and (E)-styryl benzyl sulfides where base catalyzed hydrothiolations have failed. The scope of this reaction was successfully extended for the synthesis of (E)-ON 01910·Na, a phase III clinical stage anti-cancer agent and its inactive geometrical isomer (Z)-ON 01910·Na. It is interesting to note that all the E-isomers synthesized have shown better cytotoxicity profile on cancer cells compared to the Z-isomers.


Pharmaceutical Research | 2012

Preclinical Pharmacokinetic and Pharmacodynamic Evaluation of Novel Anticancer Agents, ON01910.Na (Rigosertib, Estybon™) and ON013105, for Brain Tumor Chemotherapy

Silpa Nuthalapati; Qingyu Zhou; Ping Guo; Hua Lv; Stephen C. Cosenza; M.V. Ramana Reddy; E. Premkumar Reddy; James M. Gallo

ABSTRACTPurposeTo evaluate a mitotic inhibitor, ON01910.Na, as a potential chemotherapeutic agent for brain tumors using a series of PK/PD studies, which led to the evaluation of its structural analog, ON013105, a prodrug of the more lipophilic product, ON013100.MethodsSystemic PK characterization of ON01910 and ON013105 was completed in healthy mice. Using an orthotopic U87 glioma mouse model, brain and brain tumor distribution under steady-state conditions were evaluated for ON01910.Na and ON013105/ON013100; anticancer potential following a multiple-dose schedule of 250xa0mg/kg/day IP for 7xa0days was evaluated for ON01910.Na.ResultsON01910 exhibited low brain and brain tumor distribution with quasi-steady-state brain/plasma (Cssbrain/Cssplasma) and brain tumor/plasma (Cssbrain tumor/Cssplasma) concentration ratios of 0.03u2009±u20090.02 and 0.14u2009±u20090.08, respectively. Significant antiangiogenic potential and antiproliferative capacity of ON01910 in the intracerebral model was absent. ON013100 showed high brain and brain tumor penetration with Cssbrain/Cssplasma and Cssbrain tumor/Cssplasma ratios of 0.92u2009±u20090.26 and 1.35u2009±u20090.40, respectively; its prodrug ON013105 showed negligible brain and brain tumor penetration.ConclusionsON013105, not ON01910.Na, was identified as a potential anticancer drug candidate for further investigation in brain tumor chemotherapy based on the properties of ON013100.


Antiviral Research | 2013

A small molecule multi-kinase inhibitor reduces influenza A virus replication by restricting viral RNA synthesis

Luis Martinez-Gil; Judith G. Alamares-Sapuay; M.V. Ramana Reddy; Peter H. Goff; E. Premkumar Reddy; Peter Palese

Currently available drugs against influenza virus target the viral neuraminidase or the M2 ion channel. The emergence of viral strains resistant to these drugs has been widely described; therefore, there is an urgent need for novel antiviral drugs. Targeting of host factors required for viral replication is an attractive option for circumventing the problem of drug resistance. Several RNAi screens have demonstrated that host kinases are required for the replication of influenza virus. To determine whether compounds that inhibit these kinases can impair viral replication, we tested several kinase inhibitors for activity against influenza A virus. We demonstrate that the multi-kinase inhibitor ON108110 reduces replication of influenza A virus in a dose-dependent manner by suppressing viral RNA synthesis. In addition, ON108110 also inhibits other viruses including vesicular stomatitis virus and Newcastle disease virus, suggesting that this compound may represent a novel class of antiviral agents.


Aaps Journal | 2013

Integrated Pharmacokinetic-Driven Approach to Screen Candidate Anticancer Drugs for Brain Tumor Chemotherapy

Hua Lv; Xiaoping Zhang; Jyoti Sharma; M.V. Ramana Reddy; E. Premkumar Reddy; James M. Gallo

The goal of the study was to develop an effective screening strategy to select new agents for brain tumor chemotherapy from a series of low molecular weight anticancer agents [ON123x] by the combined use of in silico, in vitro cytotoxicity, and in vitro ADME profiling studies. The results of these studies were cast into a pipeline of tier 1 and tier 2 procedures that resulted in the identification of ON123300 as the lead compound. Of the 154 ON123xx compounds, 13 met tier 1 screening criteria based on physicochemical properties [i.e., MWu2009<u2009450xa0Da, predicted log P between 2 and 3.5] and in vitro glioma cell cytotoxicity [i.e., IC50u2009<u200910xa0μM] and were further tested in tier 2 assays. The tier 2 profiling studies consisted of metabolic stability, MDCK-MDR1 cell permeability and plasma and brain protein binding that were combined to globally assess whether favorable pharmacokinetic properties and brain penetration could be achieved in vivo. In vivo cassette dosing studies were conducted in mice for 12 compounds that permitted examination of in vitro/in vivo relationships that confirmed the suitability of the in vitro assays. A parameter derived from the in vitro assays accurately predicted the extent of drug accumulation in the brain based on the area under the drug concentration–time curve in brain measured in the cassette dosing study (r2u2009=u20090.920). Overall, the current studies demonstrated the value of an integrated pharmacokinetic-driven approach to identify potentially efficacious agents for brain tumor chemotherapy.


Investigational New Drugs | 2012

Screening candidate anticancer drugs for brain tumor chemotherapy: Pharmacokinetic-driven approach for a series of (E)-N-(substituted aryl)-3-(substituted phenyl)propenamide analogues

Hua Lv; Fan Wang; M.V. Ramana Reddy; Qingyu Zhou; Xiaoping Zhang; E. Premkumar Reddy; James M. Gallo

SummaryA pharmacokinetic [PK]-driven screening process was implemented to select new agents for brain tumor chemotherapy from a series of low molecular weight anticancer agents [ON27x] that consisted of 141 compounds. The screening procedures involved a combination of in silico, in vitro and in vivo mouse studies that were cast into a pipeline of tier 1 and tier 2 failures that resulted in a final investigation of 2 analogues in brain tumor-bearing mice. Tier 1 failures included agents with a molecular weight of > 450xa0Da, a predicted log P (log P) of either <2 or > 3.5, and a cytotoxicity IC50 value of > 2xa0uM. Next, 18 compounds underwent cassette dosing studies in normal mice that identified compounds with high systemic clearance, and low blood–brain barrier [BBB] penetration. These indices along with a derived parameter, referred to as the brain exposure index, comprised tier 2 failures that led to the administration of 2 compounds [ON27570, ON27740] as single agents [discrete dosing] to mice bearing intracerebral tumors. Comparison of ON27570’s resultant PK parameters to those obtained in the cassette dosing format suggested a drug-drug interaction most likely at the level of BBB transport, and prompted the use of the in vitro MDCK-MDR1 transport model to help assess the nature of the discrepancy. Overall, the approach was able to identify candidate compounds with suitable PK characteristics yet further revisions to the method, such as the use of in vitro metabolism and transport assays, may improve the PK-directed approach to identify efficacious agents for brain tumor chemotherapy.


Bioorganic & Medicinal Chemistry | 2016

Discovery of 2-(1H-indol-5-ylamino)-6-(2,4-difluorophenylsulfonyl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (7ao) as a potent selective inhibitor of Polo like kinase 2 (PLK2)

M.V. Ramana Reddy; Balireddy Akula; Shashidhar S. Jatiani; Rodrigo Vasquez-Del Carpio; Vinay K. Billa; Muralidhar R. Mallireddigari; Stephen C. Cosenza; D. R. C. Venkata Subbaiah; E. Vijaya Bharathi; Venkat R. Pallela; Poornima Ramkumar; Rinku Jain; Aneel K. Aggarwal; E. Premkumar Reddy

Several families of protein kinases have been shown to play a critical role in the regulation of cell cycle progression, particularly progression through mitosis. These kinase families include the Aurora kinases, the Mps1 gene product and the Polo Like family of protein kinases (PLKs). The PLK family consists of five members and of these, the role of PLK1 in human cancer is well documented. PLK2 (SNK), which is highly homologous to PLK1, has been shown to play a critical role in centriole duplication and is also believed to play a regulatory role in the survival pathway by physically stabilizing the TSC1/2 complex in tumor cells under hypoxic conditions. As a part of our research program, we have developed a library of novel ATP mimetic chemotypes that are cytotoxic against a panel of cancer cell lines. We show that one of these chemotypes, the 6-arylsulfonyl pyridopyrimidinones, induces apoptosis of human tumor cell lines in nanomolar concentrations. The most potent of these compounds, 7ao, was found to be a highly specific inhibitor of PLK2 when profiled against a panel of 288 wild type, 55 mutant and 12 lipid kinases. Here, we describe the synthesis, structure activity relationship, in vitro kinase specificity and biological activity of the lead compound, 7ao.


Journal of Pharmaceutical and Biomedical Analysis | 2011

Application of a liquid chromatography – tandem mass spectrometry (LC/MS/MS) method to the pharmacokinetics of ON01910 in brain tumor-bearing mice

Silpa Nuthalapati; Ping Guo; Qingyu Zhou; M.V. Ramana Reddy; E. Premkumar Reddy; James M. Gallo

ON01910 is a small molecular weight benzyl styryl sulfone currently under investigation as a novel anticancer agent. The purpose of the investigation was to develop a sensitive and reproducible liquid chromatography-tandem mass spectrometry (LC/MS/MS) method to quantitate levels of ON01910 in small amounts of five biological matrices; mouse plasma, feces, urine, normal brain and brain tumor. For all matrices, protein precipitation sample preparation was used that led to linear calibration curves with coefficients of determination greater than 0.99. The lower limit of quantitation (LLOQ) for all matrices was 5 ng/ml except that for mouse urine which was 10 ng/ml. The calibration standard curves were reproducible for all matrices with inter- and intra-day variability in precision and accuracy being less than 15% at all quality control concentrations except for the LLOQ in mouse plasma for which the accuracy was within 17%. The assay was successfully applied to characterize the systemic pharmacokinetics of ON01910 as well as its disposition in brain and brain tumor in mice. ON01910 exhibited a clearance of 3.61±0.85 l/h/kg and a half life of 8.66±3.30 h at 50 mg/kg dose given I.V.


Cancer Research | 2016

Dual Targeting of CDK4 and ARK5 Using a Novel Kinase Inhibitor ON123300 Exerts Potent Anticancer Activity against Multiple Myeloma

Deepak Perumal; Pei-Yu Kuo; Violetta V. Leshchenko; Zewei Jiang; Sai Krishna Athaluri Divakar; Hearn Jay Cho; Ajai Chari; Joshua Brody; M.V. Ramana Reddy; Weijia Zhang; E. Premkumar Reddy; Sundar Jagannath; Samir Parekh

Multiple myeloma is a fatal plasma cell neoplasm accounting for over 10,000 deaths in the United States each year. Despite new therapies, multiple myeloma remains incurable, and patients ultimately develop drug resistance and succumb to the disease. The response to selective CDK4/6 inhibitors has been modest in multiple myeloma, potentially because of incomplete targeting of other critical myeloma oncogenic kinases. As a substantial number of multiple myeloma cell lines and primary samples were found to express AMPK-related protein kinase 5(ARK5), a member of the AMPK family associated with tumor growth and invasion, we examined whether dual inhibition of CDK4 and ARK5 kinases using ON123300 results in a better therapeutic outcome. Treatment of multiple myeloma cell lines and primary samples with ON123300 in vitro resulted in rapid induction of cell-cycle arrest followed by apoptosis. ON123300-mediated ARK5 inhibition or ARK5-specific siRNAs resulted in the inhibition of the mTOR/S6K pathway and upregulation of the AMPK kinase cascade. AMPK upregulation resulted in increased SIRT1 levels and destabilization of steady-state MYC protein. Furthermore, ON123300 was very effective in inhibiting tumor growth in mouse xenograft assays. In addition, multiple myeloma cells sensitive to ON123300 were found to have a unique genomic signature that can guide the clinical development of ON123300. Our study provides preclinical evidence that ON123300 is unique in simultaneously inhibiting key oncogenic pathways in multiple myeloma and supports further development of ARK5 inhibition as a therapeutic approach in multiple myeloma.

Collaboration


Dive into the M.V. Ramana Reddy's collaboration.

Top Co-Authors

Avatar

E. Premkumar Reddy

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Stephen C. Cosenza

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Stacey J. Baker

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

James M. Gallo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Saikrishna A. Divakar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Samir Parekh

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aneel K. Aggarwal

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Deepak Perumal

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hearn Jay Cho

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge