Network


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

Hotspot


Dive into the research topics where Sajiv K. Nair is active.

Publication


Featured researches published by Sajiv K. Nair.


Molecular Cancer Therapeutics | 2012

Effective Targeting of Hedgehog Signaling in a Medulloblastoma Model with PF-5274857, a Potent and Selective Smoothened Antagonist That Penetrates the Blood–Brain Barrier

Allison Rohner; Mary E. Spilker; Justine L. Lam; Bernadette Pascual; Darian Bartkowski; Qing John Li; Amy H. Yang; Greg Stevens; Meirong Xu; Peter A. Wells; Simon Paul Planken; Sajiv K. Nair; Shaoxian Sun

Inhibition of the Smoothened (Smo) represents a promising therapeutic strategy for treating malignant tumors that are dependent on the Hedgehog (Hh) signaling pathway. PF-5274857 is a novel Smo antagonist that specifically binds to Smo with a Ki of 4.6 ± 1.1 nmol/L and completely blocks the transcriptional activity of the downstream gene Gli1 with an IC50 of 2.7 ± 1.4 nmol/L in cells. This Smo antagonist showed robust antitumor activity in a mouse model of medulloblastoma with an in vivo IC50 of 8.9 ± 2.6 nmol/L. The downregulation of Gli1 is closely linked to the tumor growth inhibition in patched+/− medulloblastoma mice. Mathematical analysis of the relationship between the drugs pharmacokinetics and Gli1 pharmacodynamics in patched+/− medulloblastoma tumor models yielded similar tumor and skin Gli1 IC50 values, suggesting that skin can be used as a surrogate tissue for the measurement of tumor Gli1 levels. In addition, PF-5274857 was found to effectively penetrate the blood–brain barrier and inhibit Smo activity in the brain of primary medulloblastoma mice, resulting in improved animal survival rates. The brain permeability of PF-5274857 was also confirmed and quantified in nontumor-bearing preclinical species with an intact blood–brain barrier. PF-5274857 was orally available and metabolically stable in vivo. These findings suggest that PF-5274857 is a potentially attractive clinical candidate for the treatment of tumor types including brain tumors and brain metastasis driven by an activated Hh pathway. Mol Cancer Ther; 11(1); 57–65. ©2011 AACR.


Bioorganic & Medicinal Chemistry Letters | 2009

N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Discovery of PF-915275

Michael Siu; Theodore Otto Johnson; Yong Wang; Sajiv K. Nair; Wendy D. Taylor; Stephan James Cripps; Jean Matthews; Martin Paul Edwards; Thomas A. Pauly; Jacques Ermolieff; Arturo Castro; Natilie Hosea; Amy LaPaglia; Andrea Fanjul; Jennifer E. Vogel

N-(Pyridin-2-yl) arylsulfonamides are identified as inhibitors of 11beta-hydroxysteroid dehydrogenase type 1 (11betaHSD1), an enzyme that catalyzes the reduction of the glucocorticoid cortisone to cortisol. Dysregulation of glucocorticoids has been implicated in the pathogenesis of diabetes and the metabolic syndrome. In this Letter, we present the development of an initial lead to an efficient ligand with improved physiochemical properties using a deletion strategy. This strategy allowed for further optimization of potency leading to the discovery of the clinical candidate PF-915275.


Bioorganic & Medicinal Chemistry Letters | 2010

The development and SAR of pyrrolidine carboxamide 11β-HSD1 inhibitors

Hengmiao Cheng; Jacqui Elizabeth Hoffman; Phuong T. Le; Sajiv K. Nair; Stephan James Cripps; Jean Matthews; Christopher Ronald Smith; Michele Yang; Stan Kupchinsky; Klaus Ruprecht Dress; Martin Paul Edwards; Bridget Mccarthy Cole; Evan Walters; Christine Loh; Jacques Ermolieff; Andrea Fanjul; Ganesh B. Bhat; Jocelyn Herrera; Tom Pauly; Natilie Hosea; Genevieve Paderes; Paul A. Rejto

The design and development of a series of highly selective pyrrolidine carboxamide 11beta-HSD1 inhibitors are described. These compounds including PF-877423 demonstrated potent in vitro activity against both human and mouse 11beta-HSD1 enzymes. In an in vivo assay, PF-877423 inhibited the conversion of cortisone to cortisol. Structure guided optimization effort yielded potent and stable 11beta-HSD1 selective inhibitor 42.


Bioorganic & Medicinal Chemistry Letters | 2010

Modifications of C-2 on the pyrroloquinoline template aimed at the development of potent herpesvirus antivirals with improved aqueous solubility

James A. Nieman; Sajiv K. Nair; Steven E. Heasley; Brenda L. Schultz; Herbert M. Zerth; Richard A. Nugent; Ke Chen; Kevin J. Stephanski; Todd A. Hopkins; Mary L. Knechtel; Nancee L. Oien; Janet L. Wieber; Michael W. Wathen

A series of C-2 pyrroloquinoline analogs designed to improve aqueous solubility were examined for herpesvirus polymerase and antiviral activity. Several analogs were identified that maintained the antiviral activity of the previous development candidate against HCMV, HSV-1 and VZV, but with significantly improved aqueous solubility.


Experimental Eye Research | 2009

Ocular pharmacokinetics and hypotensive activity of PF-04475270, an EP4 prostaglandin agonist in preclinical models.

Ganesh Prasanna; Jay Fortner; Cathie Xiang; Eric Zhang; Samantha Carreiro; Scott Anderson; Soisurin Sartnurak; Grace Wu; Hovhannes J. Gukasyan; M.R. Niesman; Sajiv K. Nair; Eugene Rui; Jennifer Lafontaine; Chau Almaden; Peter A. Wells; A. Krauss

Prostaglandins are widely used to lower intraocular pressure (IOP) as part of the treatment regimen for glaucoma. While FP and EP2 agonists are known to lower IOP, we investigated the ocular hypotensive activity and ocular drug distribution of PF-04475270, a novel EP4 agonist following topical administration in normotensive Beagle dogs. PF-04475270 is a prodrug of CP-734432, which stimulated cAMP formation in HEK293 cells expressing EP4 receptor and beta-lactamase activity in human EP4 expressing CHO cells transfected with a cAMP response element (CRE) with an EC(50) of 1 nM. Prodrug conversion and transcorneal permeability were assessed in rabbit corneal homogenates and a human corneal epithelial cell (cHCE) model. The compound underwent rapid hydrolysis to CP-734432 in corneal homogenates, and exhibited good permeability in the cHCE model. The descending order of ocular exposure to CP-734432 after topical dosing of PF-04475270 in dogs was as follows: cornea > aqueous humor >or= iris/ciliary body. When administered q.d., PF-04475270 lowered IOP effectively in the dog IOP model both after single and multiple days of dosing. A maximum decrease in IOP with PF-04475270 was between 30 and 45% at 24h post-dose relative to that observed with vehicle. In conclusion, PF-04475270 is a novel ocular hypotensive compound which is bioavailable following topical dosing, effectively lowering IOP in dogs. EP4 agonists could be considered as potential targets for lowering IOP for the treatment of glaucoma and ocular hypertension.


Bioorganic & Medicinal Chemistry Letters | 2016

Recent progress on third generation covalent EGFR inhibitors

Hengmiao Cheng; Sajiv K. Nair; Brion W. Murray

First generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (gefitinib and erlotinib) demonstrate excellent clinical efficacy for NSCLC patients carrying EGFR oncogenic mutations (L858R, del exon 19 deletions between amino acids 746 and 750). Invariable, drug resistance occurs with around 60% of it driven by the EGFR-T790M gatekeeper mutation. To counter the T790M-dependent resistance, third generation covalent EGFR inhibitors have been developed with high potency toward T790M containing mutants and selectivity over WT EGFR. This review provides an overview of the third generation drugs currently in clinical trials and also encompasses novel methodologies developed to discover third generation covalent EGFR drugs.


Bioorganic & Medicinal Chemistry Letters | 2013

N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Strategies to eliminate reactive metabolites

Sajiv K. Nair; Jean Matthews; Stephan James Cripps; Hengmiao Cheng; Jacqui Elizabeth Hoffman; Christopher Ronald Smith; Stanley William Kupchinsky; Michael Siu; Wendy D. Taylor; Yong Wang; Theodore Otto Johnson; Klaus Ruprecht Dress; Martin Paul Edwards; Sue Zhou; Natilie Hosea; Amy LaPaglia; Ping Kang; Arturo Castro; Jacques Ermolieff; Andrea Fanjul; Jennifer E. Vogel; Paul A. Rejto; Deepak Dalvie

N-(Pyridin-2-yl) arylsulfonamides 1 and 2 (PF-915275) were identified as potent inhibitors of 11β-hydroxysteroid dehydrogenase type 1. A screen for bioactivation revealed that these compounds formed glutathione conjugates. This communication presents the results of a risk benefit analysis carried out to progress 2 (PF-915275) to a clinical study and the strategies used to eliminate reactive metabolites in this series of inhibitors. Based on the proposed mechanism of bioactivation and structure-activity relationships, design efforts led to N-(pyridin-2-yl) arylsulfonamides such as 18 and 20 that maintained potent 11β-hydroxysteroid dehydrogenase type 1 activity, showed exquisite pharmacokinetic profiles, and were negative in the reactive metabolite assay.


Cancer Research | 2015

Abstract 2594: Characterization of a novel irreversible third generation EGFR TKI that targets T790M-mediated resistant EGFR-mutant NSCLC while sparing wild type EGFR

Mike Zientek; Sangita M. Baxi; Henry Cheng; Valeria R. Fantin; Jun Li Feng; Allison M. Given; Zelanna Goldberg; Jie Guo; Michelle Hemkens; John Charles Kath; Jennifer Lafontaine; Gary Li; Pramod P. Mehta; Brion W. Murray; Sajiv K. Nair; Simon Paul Planken; Chad Ray; Yuli Wang; Manli Shi; Anand Sistla; Tod Smeal; Greg Stevens; Wei Tan; Paolo Vicini; Marlena Walls; Liu Yang; Min-Jean Yin; Scott Weinrich

Activating mutations in EGFR confer constitutive activity providing the oncogenic drive in EGFR-mutant NSCLC. First and 2nd generation EGFR tyrosine kinase inhibitors (TKIs) are effective drugs in this setting, but are constrained by dose-limiting toxicities attributed to inhibition of wild type (WT) EGFR and by drug resistance caused, in the majority of cases, via a T790M secondary mutation in EGFR. We report the pharmacology of a novel irreversible 3rd generation EGFR TKI active against EGFR with activating and T790M mutations, but sparing WT EGFR. Our novel 3rd generation EGFR TKI was studied in a variety of in vitro and in vivo models to determine its inhibitory potencies on different EGFR variants, pharmacokinetics (PK), antitumor efficacy, exposure-response relationships, mechanism of action, and predicted human efficacious dose. In enzyme and cell assays, our compound is a highly potent inhibitor of EGFR double mutants (L858R/T790M and Del/T790M) and EGFR activating mutants (L858R and Del), but a weak inhibitor of WT EGFR (26-fold margin over mutant target potencies). Effects on downstream signaling and function indicate the underlying mechanism of the compound is direct inhibition of EGFR, with subsequent inhibition of downstream signaling that results in apoptosis and viable cell decline. In xenograft mouse models, the compound demonstrates tumor growth inhibition and regression at well-tolerated doses in models driven by EGFR double mutants and EGFR activating mutants. The antitumor efficacy is dose-dependent and strongly correlates with inhibition of EGFR phosphorylation and EGFR-mediated downstream signaling, and induction of apoptosis. Plasma concentrations assumed to be sufficient for efficacy (Ceff) were defined using a mathematical model incorporating the plasma levels of the compound, the associated inhibitory effects on EGFR phosphorylation, and the antitumor efficacy in the double and activating mutant xenograft models. Ceff was in agreement across several models and was used with in vitro human PK properties to calculate required human dose. While our compound possesses a similar profile as other recently disclosed 3rd generation EGFR TKIs, this molecule is distinguished by better potency on the activating mutants and by the widest potency margin on WT EGFR. Given that the target potencies and WT margins of 3rd generation EGFR TKIs have been sufficient for tolerated clinical efficacy in preliminary results, it can be inferred that our compound will have similar promise in the clinic. These results support our compound as a novel EGFR TKI with an inhibitory profile and favorable drug-like properties that suggest utility for treating patients with NSCLC with EGFR activating and resistance mutations. Citation Format: Mike Zientek, Sangita Baxi, Henry Cheng, Valeria Fantin, Jun Li Feng, Allison Given, Zelanna Goldberg, Jie Guo, Michelle Hemkens, John Kath, Jennifer Lafontaine, Gary Li, Pramod Mehta, Brion Murray, Sajiv Nair, Simon Planken, Chad Ray, Yuli Wang, Manli Shi, Anand Sistla, Tod Smeal, Greg Stevens, Wei Tan, Paolo Vicini, Marlena Walls, Liu Yang, Min-Jean Yin, Scott L. Weinrich. Characterization of a novel irreversible third generation EGFR TKI that targets T790M-mediated resistant EGFR-mutant NSCLC while sparing wild type EGFR. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2594. doi:10.1158/1538-7445.AM2015-2594


Archive | 2004

Benzenesulfonylamino-pyridin-2-yl derivatives and related compounds as inhibitors of 11-beta-hydroxysteroid dehydrogenase type 1 (11-beta-hsd-1) for the treatment of diabetes and obesity

Martin Paul Edwards; Theodore Otto Johnson; Sajiv K. Nair; Michael Siu; Wendy Dianne Taylor; Stephan James Cripps; Yong Wang; Hengmiao Cheng; Christopher Ronald Smith


Archive | 2005

Novel compounds of proline and morpholine derivatives

Hengmiao Cheng; Stephan James Cripps; Klaus Ruprecht Dress; Jacqui Elizabeth Hoffman; Buwen Huang; Stanley William Kupchinsky; Phuong T. Le; Sajiv K. Nair; Timothy James Parrott; Christopher Ronald Smith; Yong Wang; Yi Yang

Collaboration


Dive into the Sajiv K. Nair's collaboration.

Researchain Logo
Decentralizing Knowledge