Network


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

Hotspot


Dive into the research topics where Chakrapani Subramanyam is active.

Publication


Featured researches published by Chakrapani Subramanyam.


Journal of Medicinal Chemistry | 2010

Discovery of CP-690,550: A Potent and Selective Janus Kinase (JAK) Inhibitor for the Treatment of Autoimmune Diseases and Organ Transplant Rejection

Mark Edward Flanagan; Todd Andrew Blumenkopf; Matthew Frank Brown; Jeffrey M. Casavant; Chang Shang-Poa; Jonathan L. Doty; Eileen A. Elliott; Michael B. Fisher; Michael Hines; Craig R. Kent; Elizabeth M. Kudlacz; Brett M. Lillie; Kelly S. Magnuson; Sandra P. McCurdy; Michael John Munchhof; Bret D. Perry; Perry S. Sawyer; Timothy J. Strelevitz; Chakrapani Subramanyam; Jianmin Sun; David A. Whipple; Paul S. Changelian

There is a critical need for safer and more convenient treatments for organ transplant rejection and autoimmune disorders such as rheumatoid arthritis. Janus tyrosine kinases (JAK1, JAK3) are expressed in lymphoid cells and are involved in the signaling of multiple cytokines important for various T cell functions. Blockade of the JAK1/JAK3-STAT pathway with a small molecule was anticipated to provide therapeutic immunosuppression/immunomodulation. The Pfizer compound library was screened against the catalytic domain of JAK3 resulting in the identification of a pyrrolopyrimidine-based series of inhibitors represented by CP-352,664 (2a). Synthetic analogues of 2a were screened against the JAK enzymes and evaluated in an IL-2 induced T cell blast proliferation assay. Select compounds were evaluated in rodent efficacy models of allograft rejection and destructive inflammatory arthritis. Optimization within this chemical series led to identification of CP-690,550 1, a potential first-in-class JAK inhibitor for treatment of autoimmune diseases and organ transplant rejection.


Journal of Medicinal Chemistry | 2014

Discovery of Cytotoxic Dolastatin 10 Analogues with N-Terminal Modifications

A Maderna; M Doroski; Chakrapani Subramanyam; A Porte; C.A Leverett; Beth Cooper Vetelino; Zecheng Chen; H Risley; K Parris; J Pandit; A.H Varghese; S Shanker; C Song; S.C Sukuru; K.A Farley; M.M Wagenaar; M.J Shapiro; Sylvia Musto; M.H Lam; Frank Loganzo; C.J. O'Donnell

Auristatins, synthetic analogues of the antineoplastic natural product Dolastatin 10, are ultrapotent cytotoxic microtubule inhibitors that are clinically used as payloads in antibody-drug conjugates (ADCs). The design and synthesis of several new auristatin analogues with N-terminal modifications that include amino acids with α,α-disubstituted carbon atoms are described, including the discovery of our lead auristatin, PF-06380101. This modification of the peptide structure is unprecedented and led to analogues with excellent potencies in tumor cell proliferation assays and differential ADME properties when compared to other synthetic auristatin analogues that are used in the preparation of ADCs. In addition, auristatin cocrystal structures with tubulin are being presented that allow for the detailed examination of their binding modes. A surprising finding is that all analyzed analogues have a cis-configuration at the Val-Dil amide bond in their functionally relevant tubulin bound state, whereas in solution this bond is exclusively in the trans-configuration. This remarkable observation shines light onto the preferred binding mode of auristatins and serves as a valuable tool for structure-based drug design.


Journal of Medicinal Chemistry | 2011

Metabolism-Directed Design of Oxetane-Containing Arylsulfonamide Derivatives as γ-Secretase Inhibitors

Antonia F. Stepan; Kapil Karki; W. Scott McDonald; Peter Hans Dorff; Jason K. Dutra; Kenneth J. DiRico; Annie Won; Chakrapani Subramanyam; Ivan Viktorovich Efremov; Christopher J. O’Donnell; Charles E. Nolan; Stacey L. Becker; Leslie R. Pustilnik; Blossom Sneed; Hao Sun; Yasong Lu; Ashley Robshaw; David Riddell; Theresa J. O'Sullivan; Evelyn Sibley; Steven Capetta; Kevin Atchison; Andrew J. Hallgren; Emily Miller; Anthony Wood; R. Scott Obach

A metabolism-based approach toward the optimization of a series of N-arylsulfonamide-based γ-secretase inhibitors is reported. The lead cyclohexyl analogue 6 suffered from extensive oxidation on the cycloalkyl motif by cytochrome P450 3A4, translating into poor human liver microsomal stability. Knowledge of the metabolic pathways of 6 triggered a structure-activity relationship study aimed at lowering lipophilicity through the introduction of polarity. This effort led to several tetrahydropyran and tetrahydrofuran analogues, wherein the 3- and 4-substituted variants exhibited greater microsomal stability relative to their 2-substituted counterparts. Further reduction in lipophilicity led to the potent γ-secretase inhibitor and 3-substituted oxetane 1 with a reduced propensity toward oxidative metabolism, relative to its 2-substituted isomer. The slower rates of metabolism with 3-substituted cyclic ethers most likely originate from reductions in lipophilicity and/or unfavorable CYP active site interactions with the heteroatom. Preliminary animal pharmacology studies with a representative oxetane indicate that the series is generally capable of lowering Aβ in vivo. As such, the study also illustrates the improvement in druglikeness of molecules through the use of the oxetane motif.


Journal of Medicinal Chemistry | 2010

Design of selective, ATP-competitive inhibitors of Akt.

Kevin Daniel Freeman-Cook; Christopher Autry; Gary Borzillo; Deborah Gordon; Elsa G. Barbacci-Tobin; Vincent Bernardo; David Briere; Tracey Clark; Matthew Corbett; John Jakubczak; Shefali Kakar; Elizabeth Knauth; Blaise Lippa; Michael Joseph Luzzio; Mahmoud N. Mansour; Gary J. Martinelli; Matthew A. Marx; Kendra Louise Nelson; Jayvardhan Pandit; Francis Rajamohan; Shaughnessy Robinson; Chakrapani Subramanyam; Liuqing Wei; Martin James Wythes; Joel Morris

This paper describes the design and synthesis of novel, ATP-competitive Akt inhibitors from an elaborated 3-aminopyrrolidine scaffold. Key findings include the discovery of an initial lead that was modestly selective and medicinal chemistry optimization of that lead to provide more selective analogues. Analysis of the data suggested that highly lipophilic analogues would likely suffer from poor overall properties. Central to the discussion is the concept of optimization of lipophilic efficiency and the ability to balance overall druglike propeties with the careful control of lipophilicity in the lead series. Discovery of the nonracemic amide series and subsequent modification produced an advanced analogue that performed well in advanced preclinical assays, including xenograft tumor growth inhibition studies, and this analogue was nominated for clinical development.


Bioorganic & Medicinal Chemistry Letters | 2011

Optimization of the physicochemical and pharmacokinetic attributes in a 6-azauracil series of P2X7 receptor antagonists leading to the discovery of the clinical candidate CE-224,535

Allen J. Duplantier; Mark A. Dombroski; Chakrapani Subramanyam; Aimee M. Beaulieu; Shang-Poa Chang; Christopher A. Gabel; Crystal K. Jordan; Amit S. Kalgutkar; Kenneth G. Kraus; Jeff M. Labasi; Christopher Mussari; David G. Perregaux; Rick Shepard; Timothy J. Taylor; Kristen A. Trevena; Carrie Whitney-Pickett; Kwansik Yoon

High throughput screening (HTS) of our compound file provided an attractive lead compound with modest P2X(7) receptor antagonist potency and high selectivity against a panel of receptors and channels, but also with high human plasma protein binding and a predicted short half-life in humans. Multi-parameter optimization was used to address the potency, physicochemical and pharmacokinetic properties which led to potent P2X(7)R antagonists with good disposition properties. Compound 33 (CE-224,535) was advanced to clinical studies for the treatment of rheumatoid arthritis.


Proteins | 2009

Structural comparison of chromosomal and exogenous dihydrofolate reductase from Staphylococcus aureus in complex with the potent inhibitor trimethoprim

Holly Heaslet; Melissa S. Harris; Kelly Fahnoe; Ronald W. Sarver; Henry Putz; Jeanne Chang; Chakrapani Subramanyam; Gabriela Barreiro; J. Richard Miller

Dihydrofolate reductase (DHFR) is the enzyme responsible for the NADPH‐dependent reduction of 5,6‐dihydrofolate to 5,6,7,8‐tetrahydrofolate, an essential cofactor in the synthesis of purines, thymidylate, methionine, and other key metabolites. Because of its importance in multiple cellular functions, DHFR has been the subject of much research targeting the enzyme with anticancer, antibacterial, and antimicrobial agents. Clinically used compounds targeting DHFR include methotrexate for the treatment of cancer and diaminopyrimidines (DAPs) such as trimethoprim (TMP) for the treatment of bacterial infections. DAP inhibitors of DHFR have been used clinically for >30 years and resistance to these agents has become widespread. Methicillin‐resistant Staphylococcus aureus (MRSA), the causative agent of many serious nosocomial and community acquired infections, and other gram‐positive organisms can show resistance to DAPs through mutation of the chromosomal gene or acquisition of an alternative DHFR termed “S1 DHFR.” To develop new therapies for health threats such as MRSA, it is important to understand the molecular basis of DAP resistance. Here, we report the crystal structure of the wild‐type chromosomal DHFR from S. aureus in complex with NADPH and TMP. We have also solved the structure of the exogenous, TMP resistant S1 DHFR, apo and in complex with TMP. The structural and thermodynamic data point to important molecular differences between the two enzymes that lead to dramatically reduced affinity of DAPs to S1 DHFR. These differences in enzyme binding affinity translate into reduced antibacterial activity against strains of S. aureus that express S1 DHFR. Proteins 2009.


Bioorganic & Medicinal Chemistry Letters | 2013

Novel quinoline derivatives as inhibitors of bacterial DNA gyrase and topoisomerase IV.

Mark J. Mitton-Fry; Steven J. Brickner; Judith C. Hamel; Lori Brennan; Jeffrey M. Casavant; Michael Chen; Tao Chen; Xiaoyuan Ding; James P. Driscoll; Joel R. Hardink; Thuy Hoang; Erbing Hua; Michael D. Huband; Meghan Maloney; Anthony Marfat; Sandra P. McCurdy; Dale McLeod; Michael Plotkin; Usa Reilly; Shaughn Robinson; John Schafer; Richard M. Shepard; James F. Smith; Gregory G. Stone; Chakrapani Subramanyam; Kwansik Yoon; Wei Yuan; Richard P. Zaniewski; Christopher Zook

A structurally novel set of inhibitors of bacterial type II topoisomerases with potent in vitro and in vivo antibacterial activity was developed. Dual-targeting ability, hERG inhibition, and pharmacokinetic properties were also assessed.


Bioorganic & Medicinal Chemistry Letters | 2012

Design and synthesis of dihydrobenzofuran amides as orally bioavailable, centrally active γ-secretase modulators.

Martin Pettersson; Douglas S. Johnson; Chakrapani Subramanyam; Kelly R. Bales; Christopher W. am Ende; Benjamin Adam Fish; Michael Eric Green; Gregory W. Kauffman; Ricardo Lira; Patrick B. Mullins; Thayalan Navaratnam; Subas M. Sakya; Cory Michael Stiff; Tuan P. Tran; Beth Cooper Vetelino; Longfei Xie; Liming Zhang; Leslie R. Pustilnik; Kathleen M. Wood; Christopher J. O’Donnell

We report the discovery and optimization of a novel series of dihydrobenzofuran amides as γ-secretase modulators (GSMs). Strategies for aligning in vitro potency with drug-like physicochemical properties and good microsomal stability while avoiding P-gp mediated efflux are discussed. Lead compounds such as 35 and 43 have moderate to good in vitro potency and excellent selectivity against Notch. Good oral bioavailability was achieved as well as robust brain Aβ42 lowering activity at 100 mg/kg po dose.


Bioorganic & Medicinal Chemistry Letters | 1994

A benzisothiazolone class of potent, selective mechanism-based inhibitors of human leukocyte elastase

Dennis J. Hlasta; Malcolm R. Bell; Neil Warren Boaz; John J. Court; Ranjit C. Desai; Catherine A. Franke; Albert J. Mura; Chakrapani Subramanyam; Richard P. Dunlap

Abstract A new type of mechanism-based inhibitor of human leukocyte elastase (HLE) is described that are designed to inhibit HLE by a suicide route to form an inhibitor-enzyme complex by cross linking the enzyme active site. A mechanism of HLE inhibition is proposed and was used to design analogues with improved potency. The structure-activity relationships described in this paper are consistent with the proposed mechanism of HLE inhibition, and led to WIN 62785 ( 12 ), the most potent compound in this series with a K i * = 0.3 nM.


Bioorganic & Medicinal Chemistry Letters | 1995

The design of potent and stable benzisothiazolone inhibitors of human leukocyte elastase

Dennis J. Hlasta; Malcolm R. Bell; John J. Court; Kenneth C. Cundy; Ranjit C. Desai; Edward Ferguson; Robert J. Gordon; Virendra Kumar; Alan L. Maycock; Chakrapani Subramanyam; Richard P. Dunlap; Shari L. Eiff; Catherine A. Franke; Albert J. Mura; Anne G. Rowlands

Abstract The lead compound for this SAR study, benzisothiazolone 1a, was a 15 nM inhibitor of HLE, but was unstable in human blood ( t 1 2 min). The introduction of lipophilic substituents at the R4-position such as ethyl or isopropyl and modulation of the electrophilicity of the benzisothiazolone carbonyl led to the identification of a potent ( K i ∗ =0.27 nM) and blood stable ( t 1 2 =260 min) inhibitor 2e, WIN 63395.

Collaboration


Dive into the Chakrapani Subramanyam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John J. Court

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar

Malcolm R. Bell

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar

Virendra Kumar

Banaras Hindu University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge