Network


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

Hotspot


Dive into the research topics where Prasoon Chaturvedi is active.

Publication


Featured researches published by Prasoon Chaturvedi.


Cancer Discovery | 2013

Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M-Mediated Resistance in NSCLC

Annette O Walter; Robert Tjin Tham Sjin; Henry J Haringsma; Kadoaki Ohashi; Jing Sun; Kwangho Lee; Aleksander Dubrovskiy; Matthew T. Labenski; Zhendong Zhu; Zhigang Wang; Michael Sheets; Thia St Martin; Russell Karp; Dan van Kalken; Prasoon Chaturvedi; Deqiang Niu; M. Nacht; Russell C. Petter; William F. Westlin; Kevin Lin; Sarah S. Jaw-Tsai; Mitch Raponi; Terry Van Dyke; Jeff Etter; Zoe Weaver; William Pao; Juswinder Singh; Andrew Simmons; Thomas Harding; Andrew E. Allen

UNLABELLED Patients with non-small cell lung cancer (NSCLC) with activating EGF receptor (EGFR) mutations initially respond to first-generation reversible EGFR tyrosine kinase inhibitors. However, clinical efficacy is limited by acquired resistance, frequently driven by the EGFR(T790M) mutation. CO-1686 is a novel, irreversible, and orally delivered kinase inhibitor that specifically targets the mutant forms of EGFR, including T790M, while exhibiting minimal activity toward the wild-type (WT) receptor. Oral administration of CO-1686 as single agent induces tumor regression in EGFR-mutated NSCLC tumor xenograft and transgenic models. Minimal activity of CO-1686 against the WT EGFR receptor was observed. In NSCLC cells with acquired resistance to CO-1686 in vitro, there was no evidence of additional mutations or amplification of the EGFR gene, but resistant cells exhibited signs of epithelial-mesenchymal transition and demonstrated increased sensitivity to AKT inhibitors. These results suggest that CO-1686 may offer a novel therapeutic option for patients with mutant EGFR NSCLC. SIGNIFICANCE We report the preclinical development of a novel covalent inhibitor, CO-1686, that irreversibly and selectively inhibits mutant EGFR, in particular the T790M drug-resistance mutation, in NSCLC models. CO-1686 is the fi rst drug of its class in clinical development for the treatment of T790M-positive NSCLC, potentially offering potent inhibition of mutant EGFR while avoiding the on-target toxicity observed with inhibition of the WT EGFR.


Journal of Pharmacology and Experimental Therapeutics | 2013

Inhibition of Btk with CC-292 Provides Early Pharmacodynamic Assessment of Activity in Mice and Humans

Erica Evans; Richland Wayne Tester; Sharon Aslanian; Russell Karp; Michael Sheets; Matthew T. Labenski; Steven Richard Witowski; Heather Lounsbury; Prasoon Chaturvedi; Hormoz Mazdiyasni; Zhendong Zhu; M. Nacht; Martin I. Freed; Russell C. Petter; Alex Dubrovskiy; Juswinder Singh; William F. Westlin

Targeted therapies that suppress B cell receptor (BCR) signaling have emerged as promising agents in autoimmune disease and B cell malignancies. Bruton’s tyrosine kinase (Btk) plays a crucial role in B cell development and activation through the BCR signaling pathway and represents a new target for diseases characterized by inappropriate B cell activity. N-(3-(5-fluoro-2-(4-(2-methoxyethoxy)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide (CC-292) is a highly selective, covalent Btk inhibitor and a sensitive and quantitative assay that measures CC-292-Btk engagement has been developed. This translational pharmacodynamic assay has accompanied CC-292 through each step of drug discovery and development. These studies demonstrate the quantity of Btk bound by CC-292 correlates with the efficacy of CC-292 in vitro and in the collagen-induced arthritis model of autoimmune disease. Recently, CC-292 has entered human clinical trials with a trial design that has provided rapid insight into safety, pharmacokinetics, and pharmacodynamics. This first-in-human healthy volunteer trial has demonstrated that a single oral dose of 2 mg/kg CC-292 consistently engaged all circulating Btk protein and provides the basis for rational dose selection in future clinical trials. This targeted covalent drug design approach has enabled the discovery and early clinical development of CC-292 and has provided support for Btk as a valuable drug target for B-cell mediated disorders.


Nature Chemical Biology | 2011

Selective irreversible inhibition of a protease by targeting a noncatalytic cysteine

Margit Hagel; Deqiang Niu; Thia St. Martin; Michael Sheets; Lixin Qiao; Hugues Bernard; Russell Karp; Zhendong Zhu; Matthew T. Labenski; Prasoon Chaturvedi; Mariana Nacht; William F. Westlin; Russell C. Petter; Juswinder Singh

Designing selective inhibitors of proteases has proven problematic, in part because pharmacophores that confer potency exploit the conserved catalytic apparatus. We developed a fundamentally different approach by designing irreversible inhibitors that target noncatalytic cysteines that are structurally unique to a target in a protein family. We have successfully applied this approach to the important therapeutic target HCV protease, which has broad implications for the design of other selective protease inhibitors.


Molecular Cancer Therapeutics | 2014

In vitro and In vivo Characterization of Irreversible Mutant-Selective EGFR Inhibitors that are Wild-type Sparing

Robert Tjin Tham Sjin; Kwangho Lee; Annette O. Walter; Aleksandr Dubrovskiy; Michael Sheets; Thia St Martin; Matthew T. Labenski; Zhendong Zhu; Richland Wayne Tester; Russell Karp; Aravind Prasad Medikonda; Prasoon Chaturvedi; Yixuan Ren; Henry J. Haringsma; Jeff Etter; Mitch Raponi; Andrew Simmons; Thomas C. Harding; Deqiang Niu; M. Nacht; William F. Westlin; Russell C. Petter; Andrew M. Allen; Juswinder Singh

Patients with non–small cell lung carcinoma (NSCLC) with activating mutations in epidermal growth factor receptor (EGFR) initially respond well to the EGFR inhibitors erlotinib and gefitinib. However, all patients relapse because of the emergence of drug-resistant mutations, with T790M mutations accounting for approximately 60% of all resistance. Second-generation irreversible EGFR inhibitors are effective against T790M mutations in vitro, but retain affinity for wild-type EGFR (EGFRWT). These inhibitors have not provided compelling clinical benefit in T790M-positive patients, apparently because of dose-limiting toxicities associated with inhibition of EGFRWT. Thus, there is an urgent clinical need for therapeutics that overcome T790M drug resistance while sparing EGFRWT. Here, we describe a lead optimization program that led to the discovery of four potent irreversible 2,4-diaminopyrimidine compounds that are EGFR mutant (EGFRmut) selective and have been designed to have low affinity for EGFRWT. Pharmacokinetic and pharmacodynamic studies in H1975 tumor–bearing mice showed that exposure was dose proportional resulting in dose-dependent EGFR modulation. Importantly, evaluation of normal lung tissue from the same animals showed no inhibition of EGFRWT. Of all the compounds tested, compound 3 displayed the best efficacy in EGFRL858R/T790M-driven tumors. Compound 3, now renamed CO-1686, is currently in a phase I/II clinical trial in patients with EGFRmut-advanced NSCLC that have received prior EGFR-directed therapy. Mol Cancer Ther; 13(6); 1468–79. ©2014 AACR.


Journal of Medicinal Chemistry | 2013

Discovery of a potent and isoform-selective targeted covalent inhibitor of the lipid kinase PI3Kα.

Mariana Nacht; Lixin Qiao; Michael Sheets; Thia St. Martin; Matthew T. Labenski; Hormoz Mazdiyasni; Russell Karp; Zhendong Zhu; Prasoon Chaturvedi; Deepa Bhavsar; Deqiang Niu; William F. Westlin; Russell C. Petter; Aravind Prasad Medikonda; Juswinder Singh

PI3Kα has been identified as an oncogene in human tumors. By use of rational drug design, a targeted covalent inhibitor 3 (CNX-1351) was created that potently and specifically inhibits PI3Kα. We demonstrate, using mass spectrometry and X-ray crystallography, that the selective inhibitor covalently modifies PI3Kα on cysteine 862 (C862), an amino acid unique to the α isoform, and that PI3Kβ, -γ, and -δ are not covalently modified. 3 is able to potently (EC(50) < 100 nM) and specifically inhibit signaling in PI3Kα-dependent cancer cell lines, and this leads to a potent antiproliferative effect (GI(50) < 100 nM). A covalent probe, 8 (CNX-1220), which selectively bonds to PI3Kα, was used to investigate the duration of occupancy of 3 with PI3Kα in vivo. This is the first report of a PI3Kα-selective inhibitor, and these data demonstrate the biological impact of selectively targeting PI3Kα.


Cancer Research | 2011

Abstract 4482: Discovery of an irreversible PI3Kα-specific Inhibitor

Lixin Qiao; Mariana Nacht; Michael Sheets; Thia St. Martin; Matthew T. Labenski; Hormoz Mazdiyasni; Zhendong Zhu; Prasoon Chaturvedi; Deepa Bhavsar; Deqiang Niu; William F. Westlin; Russell C. Petter; Juswinder Singh

The PI3K pathway, which regulates cell growth, proliferation and survival, is activated in many types of human tumors by mutational activation of PI3Kα, PTEN inactivation, or activation of upstream receptor tyrosine kinases. Several PI3K inhibitors are currently in clinical development, but most are pan-PI3K inhibitors. Tumor biology data suggests that targeting PI3Kα specifically should be efficacious, and there may be advantages to not disrupting other members of the complex PI3K signaling cascade. Using structure-based drug design (SBDD), we have discovered a series of small molecules that selectively inhibit PI3Kα via the formation of an irreversible covalent bond. In addition to the enhanced pharmacodynamics achieved through permanent silencing of targets, covalent inhibitors also exhibit key advantages in terms of isoform selectivity and translational biomarker opportunities. SBDD was used to design small molecules which bond to an amino acid present uniquely in the α-isoform of PI3K. Mass spectrometry verified covalent bond formation to PI3Kα but not to the other isoforms. PI3K enzyme activity was measured using an HTRF assay. PI3Kα inhibition was evaluated in SKOV-3 cells by measuring P-AktSer473 levels. Washout experiments were performed to assess prolonged duration of action in cells. Effects on cell proliferation were assessed in cell lines bearing different mutations, such as SKOV-3 (PIK3CA H1047R), PC3 (PTEN-null) and others. SKOV-3 xenograft studies were performed in mice to measure P-AktSer473 inhibition and tumor growth inhibition in vivo. A biotinylated covalent probe molecule specific for PI3Kα was used to verify and quantitate target occupancy by the covalent inhibitor, both in vitro and ex vivo. Using trypsin digestion and MS-MS analysis, we confirmed that the designed inhibitors covalently bonded to the protein at the desired amino acid that is unique to PI3Kα. The covalent bonding resulted in potent inhibition of the PI3Kα enzyme activity and inhibition of P-AktSer473 in cells (EC50 Selective irreversible inhibitors of PI3Kα demonstrate prolonged duration of action and activity in vivo. This targeted approach should yield a first-in-class selective covalent PI3Kα inhibitor with clinical advantages. 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 4482. doi:10.1158/1538-7445.AM2011-4482


Blood | 2011

Clinical Development of AVL-292; A Potent, Selective Covalent Btk Inhibitor for the Treatment of B Cell Malignancies,

Erica K. Evans; Richland Wayne Tester; Sharon Aslanian; Prasoon Chaturvedi; Hormoz Mazdiyasni; Sabine Ponader; Bethany Tesar; Michael Sheets; Mariana Nacht; Kathryn Stiede; Steve Witowski; Heather Lounsbury; Russell C. Petter; Jennifer R. Brown; Jan A. Burger; Juswinder Singh; William F. Westlin


Clinical Lymphoma, Myeloma & Leukemia | 2011

2.23 Covalent Inhibition of Btk with Clinical Development Compound AVL-292 Disrupts Signaling That Maintains the Microenvironment Necessary for Chronic Lymphocytic Leukemia Growth

Erica Evans; S. Ponader; Russell Karp; Richland Wayne Tester; Michael Sheets; S. Aslanian; T. St. Martin; M. Nacht; Zhiming Zhu; Prasoon Chaturvedi; S. Witowski; H. Lounsbury; K. Stiede; Jan A. Burger; Russell C. Petter; Juswinder Singh; William F. Westlin


Blood | 2015

A Subcutaneously Administered Investigational RNAi Therapeutic (ALN-CC5) Targeting Complement C5 for Treatment of PNH and Complement-Mediated Diseases: Interim Phase 1 Study Results

Anita Hill; Jorg Taubel; Jim Bush; Anna Borodovsky; Noriyuki Kawahata; Helen Mclean; Christine Powell; Prasoon Chaturvedi; Garvin Warner; Pushkal Garg; Benny Sorensen


Journal of Hepatology | 2010

764 AVL-181, A POTENT AND SELECTIVE IRREVERSIBLE HCV PROTEASE INHIBITOR, REQUIRES A HELPER AMINO ACID IN THE BINDING MICROENVIRONMENT TO FORM A COVALENT BOND WITH CYS-159

Deqiang Niu; Margit Hagel; Lixin Qiao; T. St. Martin; Michael Sheets; Prasoon Chaturvedi; Matthew T. Labenski; M. Nacht; William F. Westlin; Russell C. Petter; Juswinder Singh

Collaboration


Dive into the Prasoon Chaturvedi's collaboration.

Researchain Logo
Decentralizing Knowledge