Network


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

Hotspot


Dive into the research topics where John Stellwagen is active.

Publication


Featured researches published by John Stellwagen.


Journal of Medicinal Chemistry | 2012

The Discovery and Optimization of a Novel Class of Potent, Selective, and Orally Bioavailable Anaplastic Lymphoma Kinase (ALK) Inhibitors with Potential Utility for the Treatment of Cancer.

Richard T. Lewis; Christiane Bode; Deborah Choquette; Michele Potashman; Karina Romero; John Stellwagen; Yohannes Teffera; Earl Moore; Douglas A. Whittington; Hao Chen; Linda F. Epstein; Renee Emkey; Paul S. Andrews; Violeta Yu; Douglas Saffran; Man Xu; Allison Drew; Patricia Merkel; Steven Szilvassy; Rachael L. Brake

A class of 2-acyliminobenzimidazoles has been developed as potent and selective inhibitors of anaplastic lymphoma kinase (ALK). Structure based design facilitated the rapid development of structure-activity relationships (SAR) and the optimization of kinase selectivity. Introduction of an optimally placed polar substituent was key to solving issues of metabolic stability and led to the development of potent, selective, orally bioavailable ALK inhibitors. Compound 49 achieved substantial tumor regression in an NPM-ALK driven murine tumor xenograft model when dosed qd. Compounds 36 and 49 show favorable potency and PK characteristics in preclinical species indicative of suitability for further development.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery and optimization of potent and selective imidazopyridine and imidazopyridazine mTOR inhibitors

Emily A. Peterson; Alessandro Boezio; Paul S. Andrews; Christiane Boezio; Tammy L. Bush; Alan C. Cheng; Deborah Choquette; James R. Coats; Adria E. Colletti; Katrina W. Copeland; Michelle DuPont; Russell Graceffa; Barbara Grubinska; Joseph L. Kim; Richard T. Lewis; Jingzhou Liu; Erin L. Mullady; Michele Potashman; Karina Romero; Paul L. Shaffer; Mary K. Stanton; John Stellwagen; Yohannes Teffera; Shuyan Yi; Ti Cai; Daniel S. La

mTOR is a critical regulator of cellular signaling downstream of multiple growth factors. The mTOR/PI3K/AKT pathway is frequently mutated in human cancers and is thus an important oncology target. Herein we report the evolution of our program to discover ATP-competitive mTOR inhibitors that demonstrate improved pharmacokinetic properties and selectivity compared to our previous leads. Through targeted SAR and structure-guided design, new imidazopyridine and imidazopyridazine scaffolds were identified that demonstrated superior inhibition of mTOR in cellular assays, selectivity over the closely related PIKK family and improved in vivo clearance over our previously reported benzimidazole series.


Journal of Medicinal Chemistry | 2017

Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency, Pharmacokinetics, and Metabolic Properties to Obtain Atropisomeric Quinolinone (AM-0466) that Affords Robust in Vivo Activity

Russell Graceffa; Alessandro Boezio; Jessica Able; Steven Altmann; Loren Berry; Christiane Boezio; John R. Butler; Margaret Y. Chu-Moyer; Melanie Cooke; Erin F. DiMauro; Thomas Dineen; Elma Feric Bojic; Robert S. Foti; Robert T. Fremeau; Angel Guzman-Perez; Hua Gao; Hakan Gunaydin; Hongbing Huang; Liyue Huang; Christopher P. Ilch; Michael Jarosh; Thomas Kornecook; Charles Kreiman; Daniel S. La; Joseph Ligutti; Benjamin C. Milgram; Min-Hwa Jasmine Lin; Isaac E. Marx; Hanh Nho Nguyen; Emily A. Peterson

Because of its strong genetic validation, NaV1.7 has attracted significant interest as a target for the treatment of pain. We have previously reported on a number of structurally distinct bicyclic heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. Herein, we report the discovery and optimization of a series of atropisomeric quinolinone sulfonamide inhibitors [ Bicyclic sulfonamide compounds as sodium channel inhibitors and their preparation . WO 2014201206, 2014 ] of NaV1.7, which demonstrate nanomolar inhibition of NaV1.7 and exhibit high levels of selectivity over other sodium channel isoforms. After optimization of metabolic and pharmacokinetic properties, including PXR activation, CYP2C9 inhibition, and CYP3A4 TDI, several compounds were advanced into in vivo target engagement and efficacy models. When tested in mice, compound 39 (AM-0466) demonstrated robust pharmacodynamic activity in a NaV1.7-dependent model of histamine-induced pruritus (itch) and additionally in a capsaicin-induced nociception model of pain without any confounding effect in open-field activity.


Cancer Research | 2012

Abstract 1795: Characterization of a novel series of potent, selective inhibitors of wild type and mutant/fusion anaplastic lymphoma kinase

Keith Wilcoxen; Rachael L. Brake; Doug Saffran; Yohannes Teffera; Deborah Choquette; Doug Whittington; Violeta Yu; Karina Romero; Christiane Bode; John Stellwagen; Michelle Potashman; Renee Emkey; Paul S. Andrews; Allison Drew; Man Xu; Stephen J. Szilvassy; Samer Al-Assad; Richard T. Lewis

Anaplastic lymphoma kinase (ALK) is a tyrosine kinase that has been implicated as a driving oncogene in a number of cancers, including non-small cell lung cancer (NSCLC), anaplastic large cell lymphoma (ALCL), neuroblastoma and inflammatory myofibroblastic tumors (IMT). Numerous genetic aberrations at the ALK locus are observed in cancer including point mutations, amplifications, translocations and inversions. Inversions are exemplified by inv(2)(p21;p23), which leads to the constitutively active oncogenic fusion protein EML4-ALK present in ∼5% of NSCLC. Crizotinib, a dual cMet/ALK kinase inhibitor, was recently approved by the FDA for locally advanced or metastatic NSCLC that is ALK-positive, thereby validating ALK as therapeutic target. Here we describe the pharmacological characterization of a novel series of potent, selective and orally bioavailable ALK kinase inhibitors. Members of this series inhibit wild type ALK, NPM-ALK fusion and crizotinib resistant ALK[L1196M] kinase activity at sub-nanomolar concentrations, displaying up to ∼200 fold increased inhibitory activity over crizotinib. Kinase profiling indicate that members of this series display increased selectivity scores relative crizotinib. In Karpas-299 cells, selected compounds inhibited both pY1604 ALK activation (IC50 = 2 nM) and cell proliferation (IC50 = 1 nM). Members of this series were also evaluated in the EML4-ALK expressing NSCLC cell line H3122, and displayed equipotent inhibition of pY1604 ALK activation and inhibition of cell proliferation (both IC50 = 1 nM). Members of this class did not inhibit growth of an ALK negative lymphoma cell line (HT). The in vivo activity of this series was examined in the Karpas-299 ALCL xenograft model. Compound was dosed daily (PO) at 10, 30 and 60 mg/kg. Tumor growth inhibition was observed at all dose levels, and the highest dose level resulted in significant tumor regression (96%, p 80% pALK inhibition was observed at the lowest doses tested, and complete inhibition was seen at doses of 30 mg/kg and above. In a direct comparison, members of this series achieved ALK inhibition in these tumors at a ∼15 fold lower plasma concentration than crizotinib. A PK/PD time course study was performed in the Karpas-299 model. A single 60 mg/kg dose of an inhibitor was able to maintain >90% ALK inhibition in tumors up to 24 hours post-dose, indicating significant tumor penetration and sustained ALK kinase inhibition. In conclusion, the described compounds are potent and selective inhibitors of ALK kinase, possess an impressive efficacy profile and drug-like pharmacokinetic properties. These features together indicate the potential for significant advantages over crizotinib. There is a compelling case for their clinical evaluation in patients with ALK-driven cancers. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1795. doi:1538-7445.AM2012-1795


Archive | 2011

Benzimidazole and azabenzimidazole compounds that inhibit anaplastic lymphoma kinase

Christiane Boezio; Alan C. Cheng; Deborah Choquette; Richard T. Lewis; Michele Potashman; Karina Romero; John Stellwagen; Douglas A. Whittington


Archive | 2013

Amino - dihydrothiazine and amino - dioxido dihydrothiazine compounds as beta-secretase antagonists and methods of use

Richard T. Lewis; Deborah Choquette; Oleg Epstein; Angel Guzman-Perez; Zihao Hua; Jason Brooks Human; Ted Judd; Qingyian Liu; Patricia Lopez; Ana Elena Minatti; Philip R. Olivieri; Karina Romero; Laurie B. Schenkel; John Stellwagen; Ryan White; Xiao Mei Zheng; Jennifer R. Allen; Yuan Cheng; Paul Harrington; Randall W. Hungate; Shannon Rumfelt; Robert M. Rzasa; Qiufen Xue; Wenge Zhong


Archive | 2010

Heteroaryl compounds as pikk inhibitors

Christiane Bode; Alessandro Boezio; Alan C. Cheng; Deborah Choquette; James R. Coats; Katrina W. Copeland; Hongbing Huang; Daniel La; Richard T. Lewis; Hongyu Liao; Michele Potashman; John Stellwagen; Shuyan Yi; Mark H. Norman; Markian Stec; Emily A. Peterson; Russell Graceffa


Archive | 2015

Bridged bicyclic amino thiazine dioxide compounds as inhibitors of beta- secretase

Richard T. Lewis; Jennifer R. Allen; James Brown; Angel Guzman-Perez; Zihao Hua; Ted Judd; Qingyian Liu; Philip R. Olivieri; Laurie B. Schenkel; John Stellwagen; Karina R. Vaida; Ryan White


Archive | 2014

Fused multi-cyclic sulfone compounds as inhibitors of beta-secretase and methods of use thereof

Ryan White; Jennifer R. Allen; Oleg Epstein; Fang-Tsao Hong; Zihao Hua; Jason Brooks Human; Patricia Lopez; Philip R. Olivieri; Karina Romero; Laurie B. Schenkel; John Stellwagen; Nuria A. Tamayo; Xiao Mei Zheng


Archive | 2014

FUSED MULTICYCLIC 3-AMINO-5,6-DIHYDRO-2H-1,4-THIAZINE DERIVATIVES AND THEIR USE AS BETA-SECRETASE INHIBITORS

Ryan White; Jennifer R. Allen; Oleg Epstein; Fang-Tsao Hong; Zihao Hua; Jason Brooks Human; Patricia Lopez; Philip R. Olivieri; Karina Romero; Laurie B. Schenkel; John Stellwagen; Nuria A. Tamayo; Xiao Mei Zheng

Collaboration


Dive into the John Stellwagen's collaboration.

Researchain Logo
Decentralizing Knowledge