Network


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

Hotspot


Dive into the research topics where Ragini Vuppugalla is active.

Publication


Featured researches published by Ragini Vuppugalla.


ACS Medicinal Chemistry Letters | 2015

Discovery of a Highly Selective JAK2 Inhibitor, BMS-911543, for the Treatment of Myeloproliferative Neoplasms

Honghe Wan; Gretchen M. Schroeder; Amy C. Hart; Jennifer Inghrim; James W. Grebinski; John S. Tokarski; Matthew V. Lorenzi; Dan You; Theresa McDevitt; Becky Penhallow; Ragini Vuppugalla; Yueping Zhang; Xiaomei Gu; Ramaswamy Iyer; Louis J. Lombardo; George L. Trainor; Stefan Ruepp; Jonathan Lippy; Yuval Blat; John S. Sack; Javed Khan; Kevin Stefanski; Bogdan Sleczka; Arvind Mathur; Jung-Hui Sun; Michael K. Wong; Dauh-Rurng Wu; Peng Li; Anuradha Gupta; Piramanayagam Arunachalam

JAK2 kinase inhibitors are a promising new class of agents for the treatment of myeloproliferative neoplasms and have potential for the treatment of other diseases possessing a deregulated JAK2-STAT pathway. X-ray structure and ADME guided refinement of C-4 heterocycles to address metabolic liability present in dialkylthiazole 1 led to the discovery of a clinical candidate, BMS-911543 (11), with excellent kinome selectivity, in vivo PD activity, and safety profile.


ACS Medicinal Chemistry Letters | 2015

Dimeric Macrocyclic Antagonists of Inhibitor of Apoptosis Proteins for the Treatment of Cancer

Yong Zhang; Benjamin A. Seigal; Nicholas K. Terrett; Randy Talbott; Joseph Fargnoli; Joseph G. Naglich; Charu Chaudhry; Shana Posy; Ragini Vuppugalla; Georgia Cornelius; Ming Lei; Chunlei Wang; Yingru Zhang; Robert J. Schmidt; Donna D. Wei; Michael M. Miller; Martin Patrick Allen; Ling Li; Percy H. Carter; Gregory D. Vite; Robert M. Borzilleri

A series of dimeric macrocyclic compounds were prepared and evaluated as antagonists for inhibitor of apoptosis proteins. The most potent analogue 11, which binds to XIAP and c-IAP proteins with high affinity and induces caspase-3 activation and ultimately cell apoptosis, inhibits growth of human melanoma and colorectal cell lines at low nanomolar concentrations. Furthermore, compound 11 demonstrated significant antitumor activity in the A875 human melanoma xenograft model at doses as low as 2 mg/kg on a q3d schedule.


Journal of Medicinal Chemistry | 2015

Discovery of potent heterodimeric antagonists of inhibitor of apoptosis proteins (IAPs) with sustained antitumor activity.

Heidi L. Perez; Charu Chaudhry; Stuart Emanuel; Caroline Fanslau; Joseph Fargnoli; Jinping Gan; Kyoung S. Kim; Ming Lei; Joseph G. Naglich; Sarah C. Traeger; Ragini Vuppugalla; Donna D. Wei; Gregory D. Vite; Randy Talbott; Robert M. Borzilleri

The prominent role of IAPs in controlling cell death and their overexpression in a variety of cancers has prompted the development of IAP antagonists as potential antitumor therapies. We describe the identification of a series of heterodimeric antagonists with highly potent antiproliferative activities in cIAP- and XIAP-dependent cell lines. Compounds 15 and 17 further demonstrate curative efficacy in human melanoma and lung cancer xenograft models and are promising candidates for advanced studies.


Xenobiotica | 2012

Impact of nonlinear midazolam pharmacokinetics on the magnitude of the midazolam-ketoconazole interaction in rats

Ragini Vuppugalla; Yingru Zhang; Shu-Ying Chang; Rodrigues Ad; Punit Marathe

Numerous groups have described the rat as an in vivo model for the assessment and prediction of drug–drug interactions (DDIs) in humans involving the inhibition of cytochrome P450 3A forms. Even for a well-established substrate-inhibitor pair like midazolam-ketoconazole, however, the magnitude of the DDI in rats (e.g. 1.5- to 5-fold) does not relate to what is observed clinically (e.g. 5- to 16-fold). Because nonlinear substrate pharmacokinetics (PK) may result in a weaker interaction, it was hypothesized that the lower magnitude of interaction observed in rats was due to the saturation of metabolic pathway(s) of midazolam at the doses used (10–20 mg/kg). Therefore, the inhibitory effects of ketoconazole were reevaluated at lower oral (1 and 5 mg/kg) and intravenous (IV) (1 mg/kg) doses of midazolam. In support of the hypothesis, oral exposure at 5 mg/kg dose of midazolam was 18-fold higher compared to that at 1 mg/kg. Furthermore, when the interaction was investigated at the lower midazolam dose (1 mg/kg), ketoconazole increased the IV and oral exposure of midazolam by 7-fold and 11-fold, respectively. A weaker DDI (1.5- to 1.8-fold) was observed at the higher oral midazolam dose. Collectively, these results suggest that the lower reported interaction in rats is likely due to saturation of midazolam clearance at the doses used. Therefore, when the rat is used as a DDI model to screen and differentiate compounds, or predict CYP3A inhibition in humans, it is important to use low doses of midazolam and ensure linear PK.


Regulatory Toxicology and Pharmacology | 2015

Comparison of physiologic and pharmacologic parameters in Asian and mauritius cynomolgus macaques

John Kozlosky; Jagannatha Mysore; Shawn P. Clark; Holly Burr; Jinze Li; Nelly Aranibar; Ragini Vuppugalla; Ronald C. West; Raja Mangipudy; Michael Graziano

This comparative study was conducted to assess background physiologic and pharmacologic parameters of cynomolgus macaques (Macaca fascicularis) from Cambodia, from a mixed Asian source (Cambodia, Vietnam and Indonesia), and from Mauritius. This evaluation provides a comprehensive assessment of several of these parameters in a single study. Ten male and 10 female captive-bred, age-matched macaques from each source were evaluated. Criteria for evaluation included weight gain, assessment of drug metabolizing enzyme activity, metabolomic analysis, immunologic assessments (lymphocyte subsets, TDAR, and serum Ig isotyping), clinical pathology evaluations, physical (respiratory, neurologic, cardiovascular, and ophthalmologic) examinations, pathogen screening, organ weights, and gross and microscopic pathology analyses. The results of this evaluation indicate that, compared to macaques of Asian origin, macaques from Mauritius had the lowest incidence and/or severity of spontaneous pathologic findings in several organs and tissues (lymphoid organs, stomach, kidney, urothelium, heart, arteries and lung) and better testicular maturity at a given age with minimal variability in organ weights. Although slight differences were observed in other parameters, none were considered detrimental to the use of macaques of Asian or Mauritius origin in pharmaceutical candidate safety studies with the use of a consistent source, concomitant controls, and appropriate background knowledge and screening.


ACS Medicinal Chemistry Letters | 2015

Structure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine Inhibitors.

Amy C. Hart; Gretchen M. Schroeder; Honghe Wan; James W. Grebinski; Jennifer Inghrim; James Kempson; Junqing Guo; William J. Pitts; John S. Tokarski; John S. Sack; Javed Khan; Jonathan Lippy; Matthew V. Lorenzi; Dan You; Theresa McDevitt; Ragini Vuppugalla; Yueping Zhang; Louis J. Lombardo; George L. Trainor; Ashok V. Purandare

Early hit to lead work on a pyrrolopyridine chemotype provided access to compounds with biochemical and cellular potency against Janus kinase 2 (JAK2). Structure-based drug design along the extended hinge region of JAK2 led to the identification of an important H-bond interaction with the side chain of Tyr 931, which improved JAK family selectivity. The 4,5-dimethyl thiazole analogue 18 demonstrated high levels of JAK family selectivity and was identified as a promising lead for the program.


Bioanalysis | 2016

Ultrasensitive quantitative LC–MS/MS of an inhibitor of apoptosis protein's antagonist in plasma using protein target affinity extraction

Lorell Discenza; Georgia Cornelius; Jinping Gan; Nicolas Szapiel; Randy Talbott; Charu Chaudhry; Anirban Roy; Robert M. Borzilleri; Ragini Vuppugalla; Kevin Stefanski; Robin Moore; Celia D'Arienzo; Timothy Olah; John Mehl

BACKGROUND A target protein-based affinity extraction LC-MS/MS method was developed to enable plasma level determination following ultralow dosing (0.1-3 µg/kg) of an inhibitor of apoptosis proteins molecule. Methodology & results: Affinity extraction (AE) utilizing immobilized target protein BIR2/BIR3 was used to selectively capture the inhibitor of apoptosis proteins molecule from dog plasma and enable removal of background matrix components. Pretreatment of plasma samples using protein precipitation was found to provide an additional sensitivity gain. A LLOQ of 7.8 pM was achieved by combining protein precipitation with AE. The method was used to support an ultralow dose dog toxicity study. CONCLUSION AE-LC-MS/MS, utilizing target protein, is a highly sensitive methodology for small molecule quantification with potential for broader applicability.


Archive | 2010

Anticipating and Minimizing Drug Interactions in a Drug Discovery and Development Setting: An Industrial Perspective

Ragini Vuppugalla; Sean Kim; Tatyana Zvyaga; Yong-Hae Han; Praveen Balimane; Punit Marathe; A. David Rodrigues

In the current age of polypharmacy, it is increasingly likely that a new chemical entity (NCE) will be prescribed with a second drug that demonstrates a narrow therapeutic index. As a result, one has to consider interactions involving drug-metabolizing enzymes and transporters. NCEs with drug–drug interaction (DDI) liabilities may have limited marketing potential, as they may alter the pharmacokinetic profile of a co-administered drug resulting in either unwanted side effects or loss of pharmacological activity. Within the current competitive landscape, therefore, it is highly desirable to select candidates with reduced potential for DDIs and most pharmaceutical companies spend considerable resources screening and triaging NCEs for induction and inhibition of drug-metabolizing enzymes (e.g., cytochromes P450) and transporters. Thus, the purpose of the present chapter is to provide an industrial perspective on how the existing strategies are utilized to enable the selection of suitable candidates with reduced DDI risk. Additional emphasis will be placed on in vitro tools and the challenges associated with the prediction of DDIs prior to first in man.


Experimental Cell Research | 2015

Pharmacology of smac mimetics; chemotype differentiation based on physical association with caspase regulators and cellular transport

Randy Talbott; Robert M. Borzilleri; Charu Chaudhry; Joseph Fargnoli; Henry Shen; Craig R. Fairchild; Bryan C. Barnhart; Marie Ortega; Thomas McDonagh; Ragini Vuppugalla; Gregory D. Vite; John T. Hunt; Marco M. Gottardis; Joseph G. Naglich

Cellular levels of inhibitor of apoptosis (IAP) proteins are elevated in multiple human cancers and their activities often play a part in promoting cancer cell survival by blocking apoptotic pathways, controlling signal transduction pathways and contributing to resistance. These proteins function through interactions of their BIR (baculoviral IAP repeat) protein domains with pathway components and these interactions are endogenously antagonized by Smac/Diablo (second mitochondrial activator of caspases/direct IAP binding protein with low isoelectric point). This report describes development of synthetic smac mimetics (SM) and compares their binding, antiproliferative and anti-tumor activities. All dimeric antagonists inhibit in vitro smac tetrapeptide binding to recombinant IAP proteins, rescue IAP-bound caspase-3 activity and show anti-proliferative activity against human A875 melanoma cells. One heterodimeric SM, SM3, binds tightly to IAP proteins in vitro and slowly dissociates (greater than two hours) from these protein complexes compared to the other antagonists. In addition, in vitro SM anti-proliferation potency is influenced by ABCB1 transporter (ATP-binding cassette, sub-family B; MDR1, P-gp) activities and one antagonist, SM5, does not appear to be an ABCB1 efflux pump substrate. All dimeric smac mimetics inhibit the growth of human melanoma A875 tumors implanted in athymic mice at well-tolerated doses. One antagonist, SM4, shows broad spectrum in vivo anti-tumor activity and modulates known pharmacodynamic markers of IAP antagonism. These data taken together demonstrate the range of diverse dimeric IAP antagonist activities and supports their potential as anticancer agents.


Cancer Research | 2011

Abstract DDT01-03: Discovery of BMS-911543, a highly selective JAK2 inhibitor, as a clinical candidate for the treatment of myeloproliferative disease and other malignancies

Ashok V. Purandare; Honghe Wan; Gretchen M. Schroeder; Amy C. Hart; James W. Grebinski; Jennifer Inghrim; John S. Tokarski; Dan You; Becky Penhallow; Theresa McDevitt; Ragini Vuppugalla; Yueping Zhang; Stefan Ruepp; George L. Trainor; Xia Han; Louis J. Lombardo; Francis Y. Lee; Marco M. Gottardis; Jennifer Hosbach; Elizabeth Fitzpatrick; Stuart Emanuel; Animesh Pardanani; Terra L. Lasho; Ayalew Tefferi; Matthew V. Lorenzi

Myeloproliferative diseases (MPDs) are a subset of myeloid malignancies that are characterized by the expansion of a multipotent hematopoietic stem cell. Chronic MPDs can be classified into two categories, those harboring the BCR-ABL oncogene and those that are negative. This later category of neoplasms encompasses polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). Recent discovery of activating mutations in the tyrosine kinase gene, JAK2 and constitutive activation of JAK2-STAT pathway, in large number of MPD patients has ignited considerable interest in MPD and has highlighted JAK2 as a therapeutic intervention point for drug discovery efforts. However, high-sequence homology with other JAK family members has posed a major challenge to design selective JAK2 inhibitors. Given that other JAK family members are involved in the regulation of immune function, it is important to maintain selectivity for JAK2 over these family members in order to mitigate the risks associated with undesired immunosuppression. Several JAK2 inhibitors with varying selectivity profiles are currently being evaluated in preclinical testing as well as in clinical trials for the treatment of MPD. Additionally, emerging genetic and pharmacologic evidence suggest that inhibition of the JAK2-STAT pathway may be an important therapeutic intervention point in other hematological malignancies as well as in certain solid tumors. We report here the discovery and characterization of BMS-911543, a functionally selective small molecule inhibitor of the Janus kinase family (JAK) member, JAK2. BMS-911543 is a potent and reversible inhibitor of JAK2 with a biochemical Ki of 0.48 nM. It has over 65-, 74- and 350-fold selectivity against the other JAK family members, TYK2, JAK3 and JAK1, respectively. Importantly, examination of > 450 other kinases in competition binding assays and in selected biochemical kinase assays did not reveal significant inhibitory activity for this JAK2 inhibitor, highlighting its high degree of biochemical selectivity for JAK2. Functionally, BMS-911543 displayed potent antiproliferative and pharmacodynamic (PD) effects in mutated JAK2-expressing cell lines dependent upon JAK2-STAT signaling and had little activity in cell types dependent upon other pathways such as JAK1 and JAK3. Further, single agent antiproliferative activity was not observed for BMS-911543 in a variety of solid tumor cell lines dependent upon other signaling pathways. In contrast, BMS-911543 was evaluated in colony growth assays using primary progenitor cells isolated from patients with JAK2V617F-positive myeloproliferative disease (MPD) and resulted in an increased antiproliferative response in MPD cells as compared with those from healthy volunteers. Similar to these in vitro observations, BMS-911543 was also highly active in in vivo models of JAK2-pSTAT signaling in multiple species with durable and potent pathway suppression observed after a single oral dose. Additionally, BMS-911543 was evaluated for effects in a JAK2V617F-expressing SET-2 xenograft model system and displayed a minimally effective dose of To test the hypothesis that a JAK2 selective inhibitor would have less effect on immune system function, BMS-911543 was compared to pan-JAK inhibitors in a mouse model of immunosuppression. At low dose levels active in JAK2-dependent PD models, no effects were observed on antigen-induced IgG and IgM production for BMS-911543 whereas a pan-JAK family inhibitor showed pronounced effects at all dose levels tested. The mechanistic selectivity of BMS-911543 to pan-JAK family inhibitors was extended through comparative analysis of these inhibitors in whole genome gene expression profiling experiments performed in sensitive and resistant cell types. In this comparison, BMS-911543 modulated a distinct subset of transcriptional changes as compared to pan-JAK inhibitors in clinical testing, thereby defining a minimal set of transcriptional changes underlying the pharmacologic effects of JAK2 inhibition. Collectively these results define the mechanistic basis for a differential therapeutic index between selective JAK2 and pan-JAK family inhibition pre-clinically and suggest a therapeutic rationale for the further characterization of BMS-911543 in patients with MPD and in other malignancies reliant upon constitutively active JAK2 signaling. References: Levine, R.L., et al. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders (2007). Nature Rev. Cancer, 7, 673-683. Atallah, E. and Verstovsek, S. Prospect of JAK2 inhibitor therapy in myeloproliferative neoplasms. (2009). Expert Rev. Anticancer Ther. 9, 663-670. Ghoreschi, K., et al. Janus kinases in immune cell signaling. (2009). Immunol. Rev.,228, 273-287. Mesa, R.A. and Tefferi, A. Emerging drugs for the therapy of primary and post essential thrombocythemia, post polycythemia vera myelofibrosis (2009). Expert Opin. Emerging Drugs, 14, 1-9. Roll, J.D. and Reuther, G.W. CRLF2 and JAK2 in B-progenitor acute lymphoblastic leukemia: a novel association in oncogenesis. (2010) Cancer Res, 70, 7347-7352. Rui et al., Cooperative epigenetic modulation by cancer amplicon genes (2010). Cancer Cell, 18, 590-605. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr DDT01-03. doi:10.1158/1538-7445.AM2011-DDT01-03

Collaboration


Dive into the Ragini Vuppugalla's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan You

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge