Network


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

Hotspot


Dive into the research topics where Ashvinikumar V. Gavai is active.

Publication


Featured researches published by Ashvinikumar V. Gavai.


Clinical Cancer Research | 2006

Preclinical antitumor activity of BMS-599626, a pan-HER kinase inhibitor that inhibits HER1/HER2 homodimer and heterodimer signaling.

Tai W. Wong; Francis Y. Lee; Chiang Yu; Feng R. Luo; Simone Oppenheimer; Hongjian Zhang; Richard Smykla; Harold Mastalerz; Brian E. Fink; John T. Hunt; Ashvinikumar V. Gavai; Gregory D. Vite

Purpose: The studies described here are intended to characterize the ability of BMS-599626, a small-molecule inhibitor of the human epidermal growth factor receptor (HER) kinase family, to modulate signaling and growth of tumor cells that depend on HER1 and/or HER2. Experimental Design: The potency and selectivity of BMS-599626 were assessed in biochemical assays using recombinant protein kinases, as well as in cell proliferation assays using tumor cell lines with varying degrees of dependence on HER1 or HER2 signaling. Modulation of receptor signaling was determined in cell assays by Western blot analyses of receptor autophosphorylation and downstream signaling. The ability of BMS-599626 to inhibit receptor heterodimer signaling in tumor cells was studied by receptor coimmunoprecipitation. Antitumor activity of BMS-599626 was evaluated using a number of different xenograft models that represent a spectrum of human tumors with HER1 or HER2 overexpression. Results: BMS-599626 inhibited HER1 and HER2 with IC50 of 20 and 30 nmol/L, respectively, and was highly selective when tested against a broad panel of diverse protein kinases. Biochemical studies suggested that BMS-599626 inhibited HER1 and HER2 through distinct mechanisms. BMS-599626 abrogated HER1 and HER2 signaling and inhibited the proliferation of tumor cell lines that are dependent on these receptors, with IC50 in the range of 0.24 to 1 μmol/L. BMS-599626 was highly selective for tumor cells that depend on HER1/HER2 and had no effect on the proliferation of cell lines that do not express these receptors. In tumor cells that are capable of forming HER1/HER2 heterodimers, BMS-599626 inhibited heterodimerization and downstream signaling. BMS-599626 had antitumor activity in models that overexpress HER1 (GEO), as well as in models that have HER2 gene amplification (KPL4) or overexpression (Sal2), and there was good correlation between the inhibition of receptor signaling and antitumor activity. Conclusions: BMS-599626 is a highly selective and potent inhibitor of HER1 and HER2 kinases and inhibits tumor cell proliferation through modulation of receptor signaling. BMS-599626 inhibits HER1/HER2 receptor heterodimerization and provides an additional mechanism of inhibiting tumors in which receptor coexpression and heterodimerization play a major role in driving tumor growth. The preclinical data support the advancement of BMS-599626 into clinical development for the treatment of cancer.


ACS Medicinal Chemistry Letters | 2015

Discovery of Clinical Candidate BMS-906024: A Potent Pan-Notch Inhibitor for the Treatment of Leukemia and Solid Tumors.

Ashvinikumar V. Gavai; Claude A. Quesnelle; Derek J. Norris; Wen-Ching Han; Patrice Gill; Weifang Shan; Aaron Balog; Ke Chen; Andrew J. Tebben; Richard Rampulla; Dauh-Rurng Wu; Yingru Zhang; Arvind Mathur; Ronald E. White; Anne Rose; Haiqing Wang; Zheng Yang; Asoka Ranasinghe; Celia D’Arienzo; Victor R. Guarino; Lan Xiao; Ching Su; Gerry Everlof; Vinod Arora; Ding Ren Shen; Mary Ellen Cvijic; Krista Menard; Mei-Li Wen; Jere E. Meredith; George L. Trainor

Structure-activity relationships in a series of (2-oxo-1,4-benzodiazepin-3-yl)-succinamides identified highly potent inhibitors of γ-secretase mediated signaling of Notch1/2/3/4 receptors. On the basis of its robust in vivo efficacy at tolerated doses in Notch driven leukemia and solid tumor xenograft models, 12 (BMS-906024) was selected as a candidate for clinical evaluation.


Clinical Cancer Research | 2011

Antitumor and Antiangiogenic Activities of BMS-690514, an Inhibitor of Human EGF and VEGF Receptor Kinase Families

Tai Wai Wong; Francis Lee; Stuart Emanuel; Craig R. Fairchild; Joseph Fargnoli; Brian E. Fink; Ashvinikumar V. Gavai; Amy Hammell; Benjamin Henley; Christine Hilt; John T. Hunt; Bala Krishnan; Daniel Kukral; Anne Lewin; Harold Malone; Derek J. Norris; Simone Oppenheimer; Gregory D. Vite; Chiang Yu

Purpose: The extensive involvement of the HER kinases in epithelial cancer suggests that kinase inhibitors targeting this receptor family have the potential for broad spectrum antitumor activity. BMS-690514 potently inhibits all three HER kinases, and the VEGF receptor kinases. This report summarizes data from biochemical and cellular pharmacology studies, as well as antitumor activity of BMS-690514. Experimental Design: The potency and selectivity of BMS-690514 was evaluated by using an extensive array of enzymatic and binding assays, as well as cellular assays that measure proliferation and receptor signaling. Antitumor activity was evaluated by using multiple xenograft models that depend on HER kinase signaling. The antiangiogenic properties of BMS-690514 were assessed in a matrigel plug assay, and effect on tumor blood flow was measured by dynamic contrast-enhanced MRI. Results: BMS-690514 is a potent and selective inhibitor of epidermal growth factor receptor (EGFR), HER2, and HER4, as well as the VEGF receptor kinases. It inhibits proliferation of tumor cells with potency that correlates with inhibition of receptor signaling, and induces apoptosis in lung tumor cells that have an activating mutation in EGFR. Antitumor activity was observed with BMS-690514 at multiple doses that are well tolerated in mice. There was evidence of suppression of tumor angiogenesis and endothelial function by BMS-690514, which may contribute to its efficacy. Conclusions: By combining inhibition of two receptor kinase families, BMS-690524 is a novel targeted agent that disrupts signaling in the tumor and its vasculature. Clin Cancer Res; 17(12); 4031–41. ©2011 AACR.


Journal of Medicinal Chemistry | 2009

Discovery and preclinical evaluation of [4-[[1-(3-fluorophenyl)methyl]-1H-indazol-5-ylamino]-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yl]carbamic acid, (3S)-3-morpholinylmethyl ester (BMS-599626), a selective and orally efficacious inhibitor of human epidermal growth factor receptor 1 and 2 kinases.

Ashvinikumar V. Gavai; Brian E. Fink; David J. Fairfax; Gregory Scott Martin; Lana M. Rossiter; Christian L. Holst; Soong-Hoon Kim; Kenneth J. Leavitt; Harold Mastalerz; Wen-Ching Han; Derek J. Norris; Bindu Goyal; Shankar Swaminathan; Bharat P. Patel; Arvind Mathur; Dolatrai M. Vyas; John S. Tokarski; Chiang Yu; Simone Oppenheimer; Hongjian Zhang; Punit Marathe; Joseph Fargnoli; Francis Y. Lee; Tai W. Wong; Gregory D. Vite

Structure-activity relationships in a series of 4-[1H-indazol-5-ylamino]pyrrolo[2,1-f][1,2,4]triazine-6-carbamates identified dual human epidermal growth factor receptor (HER)1/HER2 kinase inhibitors with excellent biochemical potency and kinase selectivity. On the basis of its favorable pharmacokinetic profile and robust in vivo activity in HER1 and HER2 driven tumor models, 13 (BMS-599626) was selected as a clinical candidate for treatment of solid tumors.


Bioorganic & Medicinal Chemistry Letters | 2001

Beta 3 agonists. Part 1: evolution from inception to BMS-194449.

William N. Washburn; Philip M. Sher; K.M. Poss; Ravindar N Girotra; P.J. McCann; Ashvinikumar V. Gavai; Amarendra B. Mikkilineni; Arvind Mathur; Peter T. W. Cheng; Tamara Dejneka; Chongqing Sun; Tammy C. Wang; Timothy W. Harper; Anita D. Russell; Dorothy Slusarchyk; S. Skwish; G.T. Allen; D.E. Hillyer; B.H. Frohlich; B.E. Abboa-Offei; Michael Cap; Thomas L. Waldron; R.J. George; B. Tesfamariam; Carl P. Ciosek; Denis E. Ryono; D.A. Young; Kenneth E.J. Dickinson; A.A. Seymour; C.M. Arbeeny

Screening of the BMS collection identified 4-hydroxy-3-methylsulfonanilidoethanolamines as full beta 3 agonists. Substitution of the ethanolamine nitrogen with a benzyl group bearing a para hydrogen bond acceptor promoted beta(3) selectivity. SAR elucidation established that highly selective beta(3) agonists were generated upon substitution of C(alpha) with either benzyl to form (R)-1,2-diarylethylamines or with aryl to generate 1,1-diarylmethylamines. This latter subset yielded a clinical candidate, BMS-194449 (35).(1)


Journal of Pharmaceutical Sciences | 2010

Preclinical Pharmacokinetics and In Vitro Metabolism of BMS-690514, a Potent Inhibitor of EGFR and VEGFR2

Punit Marathe; Yuwei Tang; Bogdan Sleczka; David Rodrigues; Ashvinikumar V. Gavai; Tai Wong; Lisa J. Christopher; Hongjian Zhang

BMS-690514, a potent inhibitor of human epidermal growth factor receptor (HER) 1 (EGFR), 2, and 4, and vascular endothelial growth factor receptors (VEGFR) 1-3, is currently under investigation as an oral agent for the treatment of solid tumors. In vitro and in vivo studies were conducted to characterize the pharmacokinetics and metabolism. Through integration of in vitro and in vivo pharmacokinetic data and antitumor efficacy in nude mice, human pharmacokinetics and efficacious doses were projected for BMS-690514. The oral bioavailability of BMS-690514 was 78% in mice, approximately 100% in rats, 8% in monkeys, and 29% in dogs. The low oral bioavailability in monkeys could be attributed to high systemic clearance in that species, which was also consistent with predicted clearance using in vitro data from monkey liver microsomes. Permeability of BMS-690514 in Caco-2 cells was in the intermediate range with a moderate potential to be a P-gp substrate. Experiments using recombinant human CYP enzymes and human liver microsomes suggested that CYP2D6 and CYP3A4 are likely to play a key role in the metabolic clearance of BMS-690514; in addition, direct glucuronidation of BMS-690514 was also observed in human hepatocytes. BMS-690514 was able to cross the blood-brain barrier with a brain-to-plasma ratio of approximately 1. The preclinical ADME properties of BMS-690514 suggest good oral bioavailability in humans and metabolism by multiple pathways including oxidation and glucuronidation. Based on the efficacious AUC in nude mice and predicted human pharmacokinetics, the human efficacious QD dose is predicted to be in the range of 100-200 mg.


Bioorganic & Medicinal Chemistry Letters | 2001

BMS-196085: A potent and selective full agonist of the human β3 adrenergic receptor

Ashvinikumar V. Gavai; Philip M. Sher; Amarendra B. Mikkilineni; K.M. Poss; P.J. McCann; Ravindar N Girotra; Liesl G. Fisher; Ginger Wu; Mark S. Bednarz; Arvind Mathur; Tammy C. Wang; Chongqing Sun; Dorothy Slusarchyk; S. Skwish; G.T. Allen; D.E. Hillyer; B.H. Frohlich; B.E. Abboa-Offei; Michael Cap; Thomas L. Waldron; R.J. George; B. Tesfamariam; Timothy W. Harper; Carl P. Ciosek; D.A. Young; Kenneth E.J. Dickinson; A.A. Seymour; C.M. Arbeeny; William N. Washburn

A series of 4-hydroxy-3-methylsulfonanilido-1,2-diarylethylamines were prepared and evaluated for their human beta(3) adrenergic receptor agonist activity. SAR studies led to the identification of BMS-196085 (25), a potent beta(3) full agonist (K(i)=21 nM, 95% activation) with partial agonist (45%) activity at the beta(1) receptor. Based on its desirable in vitro and in vivo properties, BMS-196085 was chosen for clinical evaluation.


Bioorganic & Medicinal Chemistry Letters | 2011

Novel pyrrolo[2,1-f][1,2,4]triazin-4-amines: Dual inhibitors of EGFR and HER2 protein tyrosine kinases

Brian E. Fink; Derek J. Norris; Harold Mastalerz; Ping Chen; Bindu Goyal; Yufen Zhao; Soong-Hoon Kim; Gregory D. Vite; Francis Y. Lee; Hongjian Zhang; Simone Oppenheimer; John S. Tokarski; Tai W. Wong; Ashvinikumar V. Gavai

A novel series of 5-((4-aminopiperidin-1-yl)methyl)-pyrrolo[2,1-f][1,2,4]triazin-4-amines with small aniline substituents at the C4 position were optimized for dual EGFR and HER2 protein tyrosine kinase inhibition. Compound 8l exhibited promising oral efficacy in both EGFR and HER2-driven human tumor xenograft models.


Bioorganic & Medicinal Chemistry Letters | 2010

Design and synthesis of 4-[3,5-dioxo-11-oxa-4,9-diazatricyclo[5.3.1.02,6]undec-4-yl]-2-trifluoromethyl-benzonitriles as androgen receptor antagonists

Hai-Yun Xiao; Aaron Balog; Ricardo M. Attar; David J. Fairfax; Linda Fleming; Christian L. Holst; Gregory Scott Martin; Lana M. Rossiter; Jing Chen; Mary-Ellen Cvjic; Janet Dell-John; Jieping Geng; Marco M. Gottardis; Wen-Ching Han; Andrew Nation; Mary T. Obermeier; Cheryl A. Rizzo; Liang Schweizer; Thomas Spires; Weifang Shan; Ashvinikumar V. Gavai; Mark E. Salvati; Gregory D. Vite

A novel series of 4-[3,5-dioxo-11-oxa-4,9-diazatricyclo[5.3.1.0(2,6)]undec-4-yl]-2-trifluoromethyl-benzonitriles has been synthesized. The ability of these compounds to act as antagonists of the androgen receptor was investigated and several were found to have potent activity in vitro and in vivo.


ACS Combinatorial Science | 2009

Design and Synthesis of a G-Protein-Coupled Receptor Antagonist Library of Aryloxyalkanolamines Using a Polymer-Supported Acyclic Acetal Linker

Jacques Y. Roberge; Lalgudi S. Harikrishnan; Muthoni G. Kamau; Zheming Ruan; Katy Van Kirk; Yalei Liu; Christopher B. Cooper; Michael A. Poss; John K. Dickson; Ashvinikumar V. Gavai; Sam T. Chao; Leslie Leith; Mark S. Bednarz; Arvind Mathur; Ramesh Kakarla; Dora M. Schnur; Roy J. Vaz; R. Michael Lawrence

A G-Protein-coupled receptor-targeted library of aryloxypropanolamines and aryloxybutanolamines was efficiently executed using a novel, polymer-supported acyclic acetal linker, producing compounds in good yields and purities.

Collaboration


Dive into the Ashvinikumar V. Gavai's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge